ArticlePDF AvailableLiterature Review

Abstract

The melanocortin-2-receptor (MC2R), also known as the ACTH receptor, is a critical component of the hypothalamic-pituitary-adrenal axis. The importance of MC2R in adrenal physiology is exemplified by the condition familial glucocorticoid deficiency (FGD), a potentially fatal disease characterised by isolated cortisol deficiency. MC2R mutations cause ~25% of cases. The discovery of a MC2R accessory protein MRAP, mutations of which account for ~20% of FGD, has provided insight into MC2R trafficking and signalling. MRAP is a single transmembrane domain accessory protein highly expressed in the adrenal gland and essential for MC2R expression and function. Mouse models helped elucidate the action of ACTH. The Mc2r knockout (Mc2r−/−) mice was the first mouse model developed to have adrenal insufficiency with deficiencies in glucocorticoid, mineralocorticoid and catecholamines. We recently reported the generation of the Mrap-/- mice which better mimics the human FGD phenotype with isolated glucocorticoid deficiency alone. The adrenal glands of adult Mrap−/− mice were grossly dysmorphic with a thickened capsule, deranged zonation and deranged WNT4/beta-catenin and sonic hedgehog (SHH) pathway signalling. Collectively, these mouse models of FGD highlight the importance of ACTH and MRAP in adrenal progenitor cell regulation, cortex maintenance and zonation.
1
ACTH signalling and adrenal development: lessons from mouse models
Tatiana V Novoselova, Peter J King, Leonardo Guasti, Louise A Metherell, Adrian JL
Clark, Li F Chan
Affiliation
Centre for Endocrinology, William Harvey Research Institute, Barts and the London
School of Medicine, Queen Mary University of London, Charterhouse Square,
London EC1M 6BQ, UK.
Corresponding Author
Dr Li Chan
Centre for Endocrinology,
William Harvey Research Institute,
Barts and the London School of Medicine,
Queen Mary University of London,
Charterhouse Square,
London EC1M 6BQ, UK.
Tel: +44 207 882 8284;
Fax: +44 207 882 6197;
Email: l.chan@qmul.ac.uk
Page 1 of 19 Accepted Manuscript published as EC-19-0190.R1. Accepted for publication: 10-Jun-2019
Copyright © 2019 the authors Downloaded from Bioscientifica.com at 06/12/2019 01:24:10AM
via free access
2
Abstract
The melanocortin-2-receptor (MC2R), also known as the ACTH receptor, is a critical
component of the hypothalamic-pituitary-adrenal axis. The importance of MC2R in
adrenal physiology is exemplified by the condition familial glucocorticoid deficiency
(FGD), a potentially fatal disease characterised by isolated cortisol deficiency. MC2R
mutations cause ~25% of cases. The discovery of a MC2R accessory protein MRAP,
mutations of which account for ~20% of FGD, has provided insight into MC2R
trafficking and signalling. MRAP is a single transmembrane domain accessory
protein highly expressed in the adrenal gland and essential for MC2R expression
and function. Mouse models helped elucidate the action of ACTH. The Mc2r
knockout (Mc2r−/−) mice was the first mouse model developed to have adrenal
insufficiency with deficiencies in glucocorticoid, mineralocorticoid and
catecholamines. We recently reported the generation of the Mrap-/- mice which better
mimics the human FGD phenotype with isolated glucocorticoid deficiency alone. The
adrenal glands of adult Mrap−/− mice were grossly dysmorphic with a thickened
capsule, deranged zonation and deranged WNT4/beta-catenin and sonic hedgehog
(SHH) pathway signalling. Collectively, these mouse models of FGD highlight the
importance of ACTH and MRAP in adrenal progenitor cell regulation, cortex
maintenance and zonation.
Page 2 of 19Accepted Manuscript published as EC-19-0190.R1. Accepted for publication: 10-Jun-2019
Copyright © 2019 the authors Downloaded from Bioscientifica.com at 06/12/2019 01:24:10AM
via free access
3
Hypothalamo-pituitary-adrenal axis
The hypothalamo-pituitary adrenal (HPA) axis dictates the production of
glucocorticoids secreted from the adrenal gland. Parvocellular neurosecretory
neurons within the hypothalamic paraventricular nucleus (PVN) secrete corticotropin-
releasing hormone (CRH) and arginine vasopressin (AVP) (Sawchenko and
Swanson, 1985) into the hypophyseal portal circulation, which then acts on anterior
pituitary corticotroph cells to trigger secretion of adrenocorticotropic hormone
(ACTH) (1, 2). ACTH, a 39 amino acid peptide, is produced by cleavage of its
precursor protein, pro-opiomelanocortin (POMC). Other cleavage products of POMC
include α−, β−, γ-melanocyte stimulating hormones (MSH), β−endorphin, N-Terminal
peptide of proopiomelanocortin, lipotrophins and Met-enkephalin, Corticotropin-like
Intermediate Peptide (CLIP) (reviewed in (3, 4)). ACTH is released into the circulation
to act on peripheral sites, mainly the adrenal glands to stimulate glucocorticoid
hormone production. Glucocorticoids negatively feedback on the release of CRH and
AVP at the hypothalamus and ACTH at the pituitary, thus providing tight regulation of
cortisol production.
ACTH receptor/Melanocortin-2-receptor
The ACTH receptor also known as the melanocortin-2-receptor (MC2R), cloned in
1992 (5), is a critical component of the HPA axis and a member of the melanocortin
receptor family. Other members are MC1R, MC3R, MC4R and MC5R, the functions
of which were recently reviewed elsewhere (3). Unlike the other MCRs that also bind
melanocortins α−,β−, γ-MSH, MC2R is unique in that it only binds ACTH. In the
adrenal gland, MC2R is expressed in all zones of the adrenal cortex. Its principal site
of action is the zona fasciculata (ZF) in the generation of glucocorticoids in response
to ACTH, although action on the zona glomerulosa (ZG) and zona reticularis (ZR)
have been implicated in a number of physiological and disease states (6). In the
case of the ZG, ACTH can acutely stimulate aldosterone production and there is now
growing interest in the role of ACTH/MC2R in primary hyperaldosteronism (7, 8).
ACTH resistance syndromes and Familial Glucocorticoid Deficiency (FGD)
Much of what we know about MC2R has been through the study of the ACTH
resistance syndrome, Familial Glucocorticoid Deficiency (FGD). FGD is a rare
autosomal recessive condition clinically characterised by isolated glucocorticoid
Page 3 of 19 Accepted Manuscript published as EC-19-0190.R1. Accepted for publication: 10-Jun-2019
Copyright © 2019 the authors Downloaded from Bioscientifica.com at 06/12/2019 01:24:10AM
via free access
4
deficiency in the presence of normal mineralocorticoid function. FGD was first
reported in two siblings diagnosed with Addison’s disease without
hypoaldosteronism (9). Patients with FGD usually present in the neonatal period or
early childhood with symptoms of hypocortisolaemia such as hypoglycaemia, failure
to thrive, recurrent infections, collapse and seizures along with severe
hyperpigmentation due to the extra-adrenal action of excessive plasma ACTH on
MC1R in the skin melanocytes (10). Biochemically, FGD patients present with very
high plasma ACTH levels often greater than 1000 pg/ml paired with very low or
absent serum cortisol concentrations, hence the term ACTH resistance. Aldosterone
levels are usually unaffected although derangements have been reported in a subset
of patients (10-12). With the identification of additional FGD causative genes such as
steroidogenic acute regulatory protein (STAR), minichromosome maintenance 4
(MCM4), nicotinamide nucleotide transhydrogenase (NNT), thioredoxin reductase 2
(TXNRD2), cytochrome p450scc (CYP11A1), glutathione peroxidase 1 (GPX1),
peroxiredoxin 3 (PRDX3) and sphingosine 1-phosphate lyase (SGPL1), additional
phenotypes including permanent or evolving mineralocorticoid deficiency have been
reported, which has been recently reviewed elsewhere (13).
Loss of function mutations in MC2R and FGD type 1
The first loss of function missense mutation in MC2R, S74I, was identified in 1993
after candidate-gene sequencing of two siblings with FGD (14). Following this,
numerous loss-of-function mutations have been identified scattered along the whole
receptor (15), cementing the importance of MC2R in HPA biology. MC2R gene
mutations account for approximately 25% of all FGD cases (16, 17) and are referred
to as FGD type I (OMIM#202200). The majority of missense mutations result in a
misfolded protein and its retention in the endoplasmic reticulum while some genetic
defects affect ligand binding, signal transduction or lead to a truncated protein
product (3, 18). Patients harbouring MC2R mutations have been shown to have tall
stature (15, 19-21) along with advanced or dissociated bone age (22). The
mechanism for this phenomenon remains unclear but is not due to the insulin-like
growth factor I-growth hormone axis as this is normal (19). One potential explanation
is that the phenotype is due to high plasma ACTH levels acting on MCR in bone (23,
24). In support of this, ACTH has been shown to directly increase chondroprogenitor
cell proliferation in-vitro (25) and promote chondrocyte differentiation although the
Page 4 of 19Accepted Manuscript published as EC-19-0190.R1. Accepted for publication: 10-Jun-2019
Copyright © 2019 the authors Downloaded from Bioscientifica.com at 06/12/2019 01:24:10AM
via free access
5
target MCR responsible for this action is uncertain (23). Interestingly, growth velocity
falls after the introduction of glucocorticoid replacement, during which plasma ACTH
levels often remain high. Another feature of absent adrenarche has also been
described (26) and, in conjunction with findings that have linked polymorphisms in
MC2R with age of adrenarche, highlights the importance of ACTH in the regulation of
adrenarche in children (27). The adrenal glands have been reported to be small in
FGD, with histopathology showing the absence of fasciculata or reticularis cells
together with disorganization of zona granulosa (ZG) cells (15). Recent data in
mouse models show that PKA signaling overactivation drives differentiation of a
reticularis-like zone in mice, supportive of the importance of ACTH in adrenal
androgen regulation (28).
Melanocortin-2-receptor accessory protein (MRAP) mutations and FGD type 2
Genetic studies of FGD patients with a normal MC2R, identified variants in the
C21orf61 gene. These variants would result in a truncation or complete absence of
the protein product, which was found to be highly expressed in the adrenal gland and
adipose tissue (16). In vitro studies revealed that this protein interacted with MC2R
and was the adrenal specific factor necessary for the transport of the receptor to the
plasma membrane and for the receptor signalling (16, 29-31) and hence was
renamed Melanocortin 2 Receptor Accessory Protein (MRAP). It is now known that
mutations in MRAP cause approximately 20% of all FGD cases and these are
termed FGD type 2 (32, 33). In comparison with FGD type 1, patients with MRAP
mutations present earlier with more severe disease. Tall stature is not seen, which is
probably a reflection of commencement of glucocorticoid replacement at an earlier
age (20). To date two FGD type 2 patients have been reported as requiring
fludrocortisone treatment, although it is unclear if this is a transient phenomenon
(34).
MRAP is a single transmembrane domain protein which is highly evolutionarily
conserved in the N-terminal and transmembrane regions (16). Human MRAP has
two isoforms produced by alternative splicing, MRAP-α (19 kDa) and MRAP-β (11.5
kDa), which differ in the C-terminus. The functional difference between the two
isoforms is unclear although differences have been reported in ACTH binding
capacity and cAMP generation have been reported (35). Interestingly, MRAP can
Page 5 of 19 Accepted Manuscript published as EC-19-0190.R1. Accepted for publication: 10-Jun-2019
Copyright © 2019 the authors Downloaded from Bioscientifica.com at 06/12/2019 01:24:10AM
via free access
6
form unique antiparallel transmembrane homodimers, which together with its
paralogue MRAP2 are as yet the only group of proteins known to do so in eukaryotic
cells. These MRAP homodimers form multimers with MC2R (36-38). The orientation
of the dimers is critical to function (36-38), moreover these antiparallel homodimers
are made early on in the biogenesis of the protein, are maintained and stable and in
a heterologous cell based system with a half-life of 2 hours (39). Although MRAP in
vitro binds and modulates the function of other melanocortin receptors (29), it is
unclear what physiological relevance this has at present.
Mouse models of FGD
The first mouse model of FGD was generated by Chida et al. in 2007 (40). The Mc2r
KO mouse model was the last member of the melanocortin receptor family to be
knocked out in mice. We recently generated a novel Mrap KO mouse model which,
like FGD, has isolated glucocorticoid deficiency. Both models are discussed in more
detail below.
Mc2r knockout mouse model
A mouse model of MC2R deficiency was generated by replacing the whole coding
region of Mc2r with a neomycin-resistance gene cassette (40), leading to complete
absence of Mc2r transcript in Mc2r-/- mice. On a C57BL/6J background the majority
of Mc2r-/- mice did not survive 48 hours after birth. Genotyping of 129 living mice at 4
weeks of age identified that only 9 (7%) were Mc2r-/-. It is worth noting that on a
B6/Balbc mix background approximately half Mc2r-/- mice survived to adulthood,
suggesting genetic modifiers at play (41). Detailed analysis demonstrated that Mc2r-
/- mice failed to produce glucocorticoids and developed profound neonatal
hypoglycemia resulting in high mortality levels during the first days of life. Moreover,
pups born to homozygous Mc2r null parents died before postnatal day 0.5 due to
lung failure, highlighting the importance of glucocorticoids in foetal lung maturation
(40). In addition to glucocorticoid deficiency, Mc2r-/- mice also had significantly lower
serum levels of aldosterone and catecholamines (40). Epinephrine levels were
significantly reduced in Mc2r-/- animals, whilst dopamine and norepinephrine
concentrations were unchanged. Examination of the adrenals of surviving adult mice
(never replaced with glucocorticoids) revealed dysmorphic glands with gross
hypoplasia of the ZF. Moreover, lipid droplets within the ZF cells were markedly
Page 6 of 19Accepted Manuscript published as EC-19-0190.R1. Accepted for publication: 10-Jun-2019
Copyright © 2019 the authors Downloaded from Bioscientifica.com at 06/12/2019 01:24:10AM
via free access
7
reduced or absent, but cell nuclei were normal. However, the precise cell identity of
such ‘ZF’ cells is not known. Interestingly, the adrenal capsule in the Mc2r null mice
was thickened compared to Mc2r+/+ littermates. The majority of naturally occurring
MC2R loss-of-function mutations are missense mutations, many with residual
function (3, 18). Location and type of reported MC2R are summarised in a recent
review (3). As a demonstration of this, patients with missense MC2R mutations have
been known to present later on in childhood (20). Several more severe cases
including homozygous nonsense or frame shift mutations in MC2R have been
described to have hyponatraemia and/or disruption of renin-angiotensin-aldosterone
system (11, 12, 34). The majority of these are transient though permanent
fludrocortisone replacement have been described in several children (34). Hence,
although the complete KO of Mc2r in mice is not fully representative of human FGD
type 1, the model has nevertheless highlighted crucial actions of ACTH and/or
glucocorticoids during development.
Mrap knockout mouse model
We recently reported a Mrap KO (Mrap-/-) mouse model created by targeting the first
coding exon of Mrap, which led to complete absence of transcript and protein in
homozygote mice (42). Intercrossing heterozygote (Mrap+/-), also on a C57BL/6J
background, demonstrated high neonatal mortality. Out of 325 mice generated from
breeding Mrap+/- mice, only three Mrap-/- mice (<1%) survived until weaning (42). The
new-born pups born from heterozygous parents with normal adrenal function died
before postnatal day 1 and morphologically showed immature lungs and lack of
hepatic glycogen stores (42). Glucocorticoids treatment of pregnant Mrap+/- dams
rescued the phenotype, resulting in Mrap-/- mice born at the expected ratio of 25%.
This indicated that the observed high mortality in homozygous Mrap-/- newborns was
likely to be due to glucocorticoid deficiency rather than a direct effect of Mrap
deletion. Moreover, this phenotype highlights the importance of foetal rather than
maternal glucocorticoids in pre-partum neonatal adaptation in the Mrap-/- mouse.
The severity of this phenotype is consistent with the human data whereby FGD type
2 patients present soon after birth (20), thought to be due to the severity of the
mutations in MRAP that lead to complete disruption or absence of protein (20).
Page 7 of 19 Accepted Manuscript published as EC-19-0190.R1. Accepted for publication: 10-Jun-2019
Copyright © 2019 the authors Downloaded from Bioscientifica.com at 06/12/2019 01:24:10AM
via free access
8
Naturally occurring mutations in MRAP (location and type) have recently been
reviewed (3).
Adult Mrap-/- mice of both genders exhibited a grossly dysmorphic adrenal cortex
with the glucocorticoid synthesis pathway severely downregulated and unable to
produce glucocorticoids in the presence of high plasma ACTH levels. Surprisingly,
unlike the Mc2r-/- mice, circulating aldosterone and catecholamine levels were
unaffected (40, 42). The enzyme, phenylethanolamine N-methyltransferase (PNMT)
which is responsible for conversion of norepinephrine to epinephrine appeared to be
unaffected in Mrap-/- mice even though PNMT expression is known to be dependent
on glucocorticoid action. In contrast, PNMT is markedly reduced in Mc2r-/- (40). As
both knockout models are on a C57BL/6J background, one obvious difference is that
the Mrap-/- mice received relatively high doses of glucocorticoids between E17.5 until
weaning. This could contribute to the discrepancy in PNMT between Mc2r-/- and
Mrap-/- mice, especially in light of the fact that prenatal exposure to glucocorticoids
have been shown to lead to increased PNMT mRNA by RT-PCR and elevated
plasma epinephrine in adult rats (43). The discrepancy between aldosterone levels in
Mc2r-/- and Mrap-/- mice could be due to a number of possibilities, such as Mrap
independent aldosterone production (although in-vitro the MC2R is non-functional in
the absence of MRAP and MRAP protein expression closely mirrors that of MC2R in
the ZG), differences in salt intake between the 2 models (both rodent lines are fed
ad-libitum on a standard chow diet although, the salt content may differ) and finally
alterations of the renin-angiotensin-aldosterone system have also been reported in
animals exposed to glucocorticoids prenatally (44). Determining the mechanism of
such differences would further dissect adrenal ACTH action. Importantly however,
the absence of mineralocorticoid and catecholamine deficiency in Mrap-/- mice
makes it a unique model for studying FGD and isolated glucocorticoid deficiency.
Similar to the Mc2r-/- mouse model, the Mrap-/- adrenal capsule is thickened, which in
the Mrap-/- mice was shown to be due to an increased cell number, rather than
hyperplasia. This is of particular interest as the adrenal capsule and the subcapsular
region is known to contain adrenocortical stem/progenitor cells capable of dividing,
migrating centripetally and differentiating into mature steroid producing cell types
(45-47). In keeping with this, the absence of MRAP and therefore ACTH signalling,
Page 8 of 19Accepted Manuscript published as EC-19-0190.R1. Accepted for publication: 10-Jun-2019
Copyright © 2019 the authors Downloaded from Bioscientifica.com at 06/12/2019 01:24:10AM
via free access
9
resulted in small adrenals with grossly deranged cortex zonation (discussed in
subsequent sections).
Other MRAP mouse models: Transgenic MRAP adipose tissue overexpression
mouse model
Prior to its renaming in 2005, MRAP was first identified as a putative novel
membrane protein selectively expressed during adipogenic conversion of 3T3-L1
cells and called FALP (fat tissue-specific low molecular weight protein) (48). The
protein expressed in both brown and white fat tissue and highly expressed during
adipogenesis (48, 49). More recently, the importance of MRAP in energy balance
was demonstrated using transgenic mice overexpressing MRAP in a fat specific
manner, under the control of the aP2 (adipocyte fatty acid binding protein) promoter
(50). These mice were shown to be protected against diet induced obesity and
diabetes, through enhancement of ACTH induced lipolysis. Therefore, this
demonstrated for the first time the emerging importance of MRAP in metabolism.
ACTH and adrenocortical renewal and zonation – contribution of Mrap and
Mc2r KO mouse models
Adrenocortical renewal and regeneration
The adrenal gland has a great capacity to respond to changes, renew and
regenerate. For example, activation of the HPA axis leads to expansion of the ZF
and increased expression of CYP11B1 and glucocorticoid production, whilst
suppression by dexamethasone leads to the contraction/atrophy of the ZF, reduced
CYP11B1 and glucocorticoid production (51). This is not specific to the ZF and
activation or inhibition of the renin-angiotensin-aldosterone system (ie triggered by a
diet low in sodium or treatment with ACE inhibitors, respectively) results in similar
changes in ZG morphology, CYP11B2 expression and aldosterone production (52).
This together with the appearance of compensatory growth of the contralateral
adrenal gland following unilateral adrenalectomy (53) demonstrates the dynamic
nature of the adrenal gland. Apart from remodelling, the adrenal gland can also
regenerate from residual adrenal capsular and adherent ZG cells in enucleation
studies (54, 55). Regeneration arises from cell proliferation and differentiation and is
associated with a thickened adrenal cortex (56).
Page 9 of 19 Accepted Manuscript published as EC-19-0190.R1. Accepted for publication: 10-Jun-2019
Copyright © 2019 the authors Downloaded from Bioscientifica.com at 06/12/2019 01:24:10AM
via free access
10
Role of ACTH in adrenal progenitor cell differentiation and maintenance
ACTH administration can induce ZF hyperplasia, with no effect on the ZG (57). More
recently, the localisation of Mc2r and Mrap to the undifferentiated zone (layer of
cyp11b1/b2 negative cells located between the ZG and ZF) in the rat adrenal (58,
59), suggested a role for ACTH in the differentiation of progenitor cells towards the
ZF phenotype in vivo. However, the data from remodelling experiments provides
somewhat conflicting evidence for the exact role of ACTH in adrenal progenitor cell
differentiation and maintenance. For example, some studies show that treatment
with dexamethasone blocks ZF proliferation and compensatory growth of the
contralateral gland following unilateral adrenalectomy (53, 60), whilst other studies
do not show this (61, 62). Hypophysectomy, with the complete removal of the
pituitary gland, blocks adrenal gland regeneration following enucleation (63) but does
not completely block compensatory growth (51, 64), and ACTH has even been
shown to inhibit this process (64). However, overall there is agreement that
hypophysectomy reduces the extent of compensatory growth suggesting the
presence of a pituitary factor. One possible factor is pituitary derived N-POMC,
derived from the POMC cleavage product pro--MSH. Neutralising antibodies against
pro--MSH inhibit both adrenal regeneration and compensatory growth. However,
pro--MSH has no direct mitogenic activity (65), hence it has been suggested that
further processing of pro--MSH to peptides such as N-POMC with mitogenic activity
is required (66). Evidence of the need for pituitary factors, including ACTH, comes
from work on POMC-/- animals that have complete absence of the POMC peptide
(67, 68). POMC-/- mice have adrenal glands that fail to proliferate postnatally leading
to atrophic adrenals which become undetectable with age (69). These adrenal
glands from POMC-/- animals can be rescued in size and function, following
transplantation into a wild-type recipient animal with physiological levels of all POMC
peptides. Interestingly, N-POMC cannot restore adrenal growth in POMC null mice
(70). However, high dose replacement of ACTH over several days restores POMC-/-
adrenal weight, morphology and corticosterone secretion (36), but it has been
suggested this is due to hypertrophy of the ZF rather than full regeneration of the
adrenal gland (71).
Page 10 of 19Accepted Manuscript published as EC-19-0190.R1. Accepted for publication: 10-Jun-2019
Copyright © 2019 the authors Downloaded from Bioscientifica.com at 06/12/2019 01:24:10AM
via free access
11
Contribution of Mc2r and Mrap KO mouse models to understanding of adrenal
stem/progenitor cell differentiation and maintenance
The MC2R and MRAP KO mouse models add to our existing knowledge of the role
of ACTH in adrenal gland stem/progenitor cell differentiation and gland maintenance.
Assessment of the adrenal glands of Mrap-/- mice at embryonic day 17.5 shows that
the adrenal sizes are comparable to Mrap+/+ mice, derived from intercrosses of
heterozygous mice with normal HPA activity. However, when assessed at 8 weeks of
age the gland is significantly reduced in size with a thickened adrenal capsule (42).
One major issue is the separation of the effects of absent ACTH action and
glucocorticoid deficiency that are both present in adult Mrap-/- mice. After lifetime
glucocorticoid replacement the capsule reduced in thickness but still remained
significantly increased compared to wild-type littermate animals suggesting that both
glucocorticoid deficiency and absent ACTH action contribute to the expansion of the
stem cell niche in the adrenal gland, in keeping with some of the data described
above. Treatment of wild-type control mice with glucocorticoid resulted in
biochemically undetectable plasma ACTH coupled with corticosterone within the
normal range. These mice have a thickened capsule and thus taken together this is
highly suggestive that ACTH deficiency is a key contributor to capsule thickening.
The Mrap-/- mouse model also introduces some novel concepts in the growing
landscape of molecular pathways and factors involved in adrenal stem/progenitor
cell determination and differentiation. In fact this field is rapidly gaining pace with the
identification of new factors required for the adrenal homeostasis and zonation such
as RSPO3, EZH2 and ZNRF3 (72-74) and [reviewed in (46, 52)]. The WNT4/-
catenin and SHH pathways have been described to be key drivers of ZG identity
(75).
Cells that express WNT/-catenin, which co-express SHH and CYP11B2, normally
reside in the subcapsular region (40-42). It has been shown that cAMP/PKA,
activated by ACTH signalling in the adrenal gland, represses the WNT/beta-catenin
pathway to allow lineage conversion and correct adrenal cortex zonation (45).
Consistent with this, is the absence of ZF in the adrenals of Mrap-/- mice, whilst ZG is
intact and functional. However, the Mrap-/- mice identifies a concentric zone of cells
Page 11 of 19 Accepted Manuscript published as EC-19-0190.R1. Accepted for publication: 10-Jun-2019
Copyright © 2019 the authors Downloaded from Bioscientifica.com at 06/12/2019 01:24:10AM
via free access
12
between the ZG and adrenal medulla that are negative for Cyp11b2, Cyp11b1 and
20-HSD and hence not terminally differentiated ZG, ZF or ZR/fetal X-zone cells
respectively. These cells are WNT4/-catenin positive but negative for downstream
canonical targets of WNT signalling, LEF1 and DAB2 suggesting that canonical WNT
signalling is not active in these cells. Another option is that these cells were once
LEF1 and DAB2 positive and have subsequently lost some of ZG features. However,
the precise origin, fate and signalling potential within these cells are currently being
investigated.
SHH signalling is another key pathway regulating the differentiation of progenitor
cells into steroidogenic cells (76, 77). SHH positive cells in the stem cell niche, which
are located in the subcapsular region in mice and in the undifferentiated zone in rats,
have been shown through lineage tracing to differentiate into all cortical cell
populations in mice. The majority of Shh-descendants become ZG cells first and
then transition to ZF cells during centripetal migration (47, 76). In SHH KO mice, the
adrenal capsule is reduced to a single cell layer, suggesting that SHH could act as a
capsule cell mitogen/chemoattractant for noncapsule mesenchyme or to maintain
capsule progenitors (76). In the absence of MRAP, we see thickened capsule,
upregulated SHH expression and ectopic SHH expression throughout the cortex, no
longer restricted to the subscapular region. Together with the co-expression of
WNT4 and CYP11B2 this is suggestive that these cells have acquired ZG features.
Glucocorticoid treatment only partially attenuated this phenotype. Interestingly, the
levels of Gli1, which is a canonical target of SHH, were unchanged despite high SHH
expression. Determining the reason for this dissociation between high SHH and Gli1
which is unchanged as well as the co-ordination with other factors involved with
capsule thickness such as FGF signalling will be investigated in the future (Fig 1).
In conclusion, the mouse models of FGD have helped elucidate the action of ACTH
and glucocorticoids in adrenal development, progenitor cell renewal and zonation.
Further studies will focus on the role of ACTH signalling in physiology as well as how
this can be altered in disease states.
Declaration of interest
Page 12 of 19Accepted Manuscript published as EC-19-0190.R1. Accepted for publication: 10-Jun-2019
Copyright © 2019 the authors Downloaded from Bioscientifica.com at 06/12/2019 01:24:10AM
via free access
13
We have no conflict of interest that could be perceived as prejudicing the impartiality
of the research reported.
Funding
The work reviewed was funded by The Medical Research Council UK
(MRC/Academy of Medical Sciences Clinician Scientist Fellowship Grant G0802796
to LF Chan).
Page 13 of 19 Accepted Manuscript published as EC-19-0190.R1. Accepted for publication: 10-Jun-2019
Copyright © 2019 the authors Downloaded from Bioscientifica.com at 06/12/2019 01:24:10AM
via free access
14
1. Aguilera G, Harwood JP, Wilson JX, Morell J, Brown JH, Catt KJ. Mechanisms of action of
corticotropin-releasing factor and other regulators of corticotropin release in rat pituitary cells. J Biol
Chem 1983 258 8039-8045.
2. Vale W, Spiess J, Rivier C, Rivier J. Characterization of a 41-residue ovine hypothalamic
peptide that stimulates secretion of corticotropin and beta-endorphin. Science 1981 213 1394-1397.
3. Novoselova TV, Chan LF, Clark AJL. Pathophysiology of melanocortin receptors and their
accessory proteins. Best Practice & Research Clinical Endocrinology & Metabolism 2018.
4. Harno E, Gali Ramamoorthy T, Coll AP, White A. POMC: The Physiological Power of Hormone
Processing. Physiol Rev 2018 98 2381-2430.
5. Mountjoy KG, Robbins LS, Mortrud MT, Cone RD. The cloning of a family of genes that
encode the melanocortin receptors. Science 1992 257 1248-1251.
6. Clark AJ, Forfar R, Hussain M, Jerman J, McIver E, Taylor D, Chan L. ACTH Antagonists. Front
Endocrinol (Lausanne) 2016 7 101.
7. El Ghorayeb N, Bourdeau I, Lacroix A. Role of ACTH and Other Hormones in the Regulation of
Aldosterone Production in Primary Aldosteronism. Front Endocrinol (Lausanne) 2016 7 72.
8. Funder JW. The Potential of ACTH in the Genesis of Primary Aldosteronism. Front Endocrinol
(Lausanne) 2016 7 40.
9. Shepard TH, Landing BH, Mason DG. Familial Addison's disease; case reports of two sisters
with corticoid deficiency unassociated with hypoaldosteronism. AMA J Dis Child 1959 97 154-162.
10. Turan S, Hughes C, Atay Z, Guran T, Haliloglu B, Clark AJ, Bereket A, Metherell LA. An atypical
case of familial glucocorticoid deficiency without pigmentation caused by coexistent homozygous
mutations in MC2R (T152K) and MC1R (R160W). J Clin Endocrinol Metab 2012 97 E771-774.
11. Lin L, Hindmarsh PC, Metherell LA, Alzyoud M, Al-Ali M, Brain CE, Clark AJ, Dattani MT,
Achermann JC. Severe loss-of-function mutations in the adrenocorticotropin receptor (ACTHR,
MC2R) can be found in patients diagnosed with salt-losing adrenal hypoplasia. Clin Endocrinol (Oxf)
2007 66 205-210.
12. Chan LF, Metherell LA, Krude H, Ball C, O'Riordan SM, Costigan C, Lynch SA, Savage MO,
Cavarzere P, Clark AJ. Homozygous nonsense and frameshift mutations of the ACTH receptor in
children with familial glucocorticoid deficiency (FGD) are not associated with long-term
mineralocorticoid deficiency. Clin Endocrinol (Oxf) 2009 71 171-175.
13. Maharaj A MA, Chan L, Novoselova T, Prasad R, Metherell L, Guasti L. Isolated Glucocorticoid
Deficiency: genetic causes and animal models. 2019.
14. Clark AJ, McLoughlin L, Grossman A. Familial glucocorticoid deficiency associated with point
mutation in the adrenocorticotropin receptor. Lancet 1993 341 461-462.
15. Clark AJ, Weber A. Adrenocorticotropin insensitivity syndromes. Endocr Rev 1998 19 828-
843.
16. Metherell LA, Chapple JP, Cooray S, David A, Becker C, Ruschendorf F, Naville D, Begeot M,
Khoo B, Nurnberg P et al. Mutations in MRAP, encoding a new interacting partner of the ACTH
receptor, cause familial glucocorticoid deficiency type 2. Nat Genet 2005 37 166-170.
17. Weber A, Clark AJ. Mutations of the ACTH receptor gene are only one cause of familial
glucocorticoid deficiency. Hum Mol Genet 1994 3 585-588.
18. Chung TT, Webb TR, Chan LF, Cooray SN, Metherell LA, King PJ, Chapple JP, Clark AJ. The
majority of adrenocorticotropin receptor (melanocortin 2 receptor) mutations found in familial
glucocorticoid deficiency type 1 lead to defective trafficking of the receptor to the cell surface. J Clin
Endocrinol Metab 2008 93 4948-4954.
19. Elias LL, Huebner A, Metherell LA, Canas A, Warne GL, Bitti ML, Cianfarani S, Clayton PE,
Savage MO, Clark AJ. Tall stature in familial glucocorticoid deficiency. Clin Endocrinol (Oxf) 2000 53
423-430.
20. Chung TT, Chan LF, Metherell LA, Clark AJ. Phenotypic characteristics of familial
glucocorticoid deficiency (FGD) type 1 and 2. Clin Endocrinol (Oxf) 2010 72 589-594.
Page 14 of 19Accepted Manuscript published as EC-19-0190.R1. Accepted for publication: 10-Jun-2019
Copyright © 2019 the authors Downloaded from Bioscientifica.com at 06/12/2019 01:24:10AM
via free access
15
21. Imamine H, Mizuno H, Sugiyama Y, Ohro Y, Sugiura T, Togari H. Possible relationship
between elevated plasma ACTH and tall stature in familial glucocorticoid deficiency. Tohoku J Exp
Med 2005 205 123-131.
22. Clark AJ, Weber A. Molecular insights into inherited ACTH resistance syndromes. Trends
Endocrinol Metab 1994 5 209-214.
23. Bohm M, Grassel S. Role of proopiomelanocortin-derived peptides and their receptors in the
osteoarticular system: from basic to translational research. Endocr Rev 2012 33 623-651.
24. Zhong Q, Sridhar S, Ruan L, Ding KH, Xie D, Insogna K, Kang B, Xu J, Bollag RJ, Isales CM.
Multiple melanocortin receptors are expressed in bone cells. Bone 2005 36 820-831.
25. Yeh JK, Evans JF, Niu QT, Aloia JF. A possible role for melanocortin peptides in longitudinal
growth. J Endocrinol 2006 191 677-686.
26. Weber A, Clark AJ, Perry LA, Honour JW, Savage MO. Diminished adrenal androgen secretion
in familial glucocorticoid deficiency implicates a significant role for ACTH in the induction of
adrenarche. Clin Endocrinol (Oxf) 1997 46 431-437.
27. Lappalainen S, Utriainen P, Kuulasmaa T, Voutilainen R, Jaaskelainen J. ACTH receptor
promoter polymorphism associates with severity of premature adrenarche and modulates
hypothalamo-pituitary-adrenal axis in children. Pediatr Res 2008 63 410-414.
28. Dumontet T, Sahut-Barnola I, Septier A, Montanier N, Plotton I, Roucher-Boulez F, Ducros V,
Lefrancois-Martinez AM, Pointud JC, Zubair M et al. PKA signaling drives reticularis differentiation
and sexually dimorphic adrenal cortex renewal. JCI Insight 2018 3.
29. Chan LF, Webb TR, Chung TT, Meimaridou E, Cooray SN, Guasti L, Chapple JP, Egertova M,
Elphick MR, Cheetham ME et al. MRAP and MRAP2 are bidirectional regulators of the melanocortin
receptor family. Proc Natl Acad Sci U S A 2009 106 6146-6151.
30. Webb TR, Chan L, Cooray SN, Cheetham ME, Chapple JP, Clark AJ. Distinct melanocortin 2
receptor accessory protein domains are required for melanocortin 2 receptor interaction and
promotion of receptor trafficking. Endocrinology 2009 150 720-726.
31. Cooray SN, Almiro Do Vale I, Leung KY, Webb TR, Chapple JP, Egertova M, Cheetham ME,
Elphick MR, Clark AJ. The melanocortin 2 receptor accessory protein exists as a homodimer and is
essential for the function of the melanocortin 2 receptor in the mouse y1 cell line. Endocrinology
2008 149 1935-1941.
32. Metherell LA, Chan LF, Clark AJ. The genetics of ACTH resistance syndromes. Best Pract Res
Clin Endocrinol Metab 2006 20 547-560.
33. Chan LF, Clark AJ, Metherell LA. Familial glucocorticoid deficiency: advances in the molecular
understanding of ACTH action. Horm Res 2008 69 75-82.
34. Guran T, Buonocore F, Saka N, Ozbek MN, Aycan Z, Bereket A, Bas F, Darcan S, Bideci A,
Guven A et al. Rare Causes of Primary Adrenal Insufficiency: Genetic and Clinical Characterization of
a Large Nationwide Cohort. J Clin Endocrinol Metab 2016 101 284-292.
35. Roy S, Rached M, Gallo-Payet N. Differential regulation of the human adrenocorticotropin
receptor [melanocortin-2 receptor (MC2R)] by human MC2R accessory protein isoforms alpha and
beta in isogenic human embryonic kidney 293 cells. Mol Endocrinol 2007 21 1656-1669.
36. Sebag JA, Hinkle PM. Melanocortin-2 receptor accessory protein MRAP forms antiparallel
homodimers. Proc Natl Acad Sci U S A 2007 104 20244-20249.
37. Cooray SN, Chung TT, Mazhar K, Szidonya L, Clark AJ. Bioluminescence resonance energy
transfer reveals the adrenocorticotropin (ACTH)-induced conformational change of the activated
ACTH receptor complex in living cells. Endocrinology 2011 152 495-502.
38. Malik S, Dolan TM, Maben ZJ, Hinkle PM. Adrenocorticotropic Hormone (ACTH) Responses
Require Actions of the Melanocortin-2 Receptor Accessory Protein on the Extracellular Surface of the
Plasma Membrane. J Biol Chem 2015 290 27972-27985.
39. Maben ZJ, Malik S, Jiang LH, Hinkle PM. Dual Topology of the Melanocortin-2 Receptor
Accessory Protein Is Stable. Front Endocrinol (Lausanne) 2016 7 96.
Page 15 of 19 Accepted Manuscript published as EC-19-0190.R1. Accepted for publication: 10-Jun-2019
Copyright © 2019 the authors Downloaded from Bioscientifica.com at 06/12/2019 01:24:10AM
via free access
16
40. Chida D, Nakagawa S, Nagai S, Sagara H, Katsumata H, Imaki T, Suzuki H, Mitani F, Ogishima
T, Shimizu C et al. Melanocortin 2 receptor is required for adrenal gland development,
steroidogenesis, and neonatal gluconeogenesis. Proc Natl Acad Sci U S A 2007 104 18205-18210.
41. Chida D, Sato T, Sato Y, Kubo M, Yoda T, Suzuki H, Iwakura Y. Characterization of mice
deficient in melanocortin 2 receptor on a B6/Balbc mix background. Mol Cell Endocrinol 2009 300
32-36.
42. Novoselova TV, Hussain M, King PJ, Guasti L, Metherell LA, Charalambous M, Clark AJL, Chan
LF. MRAP deficiency impairs adrenal progenitor cell differentiation and gland zonation. FASEB J 2018
fj201701274RR.
43. Nguyen P, Khurana S, Peltsch H, Grandbois J, Eibl J, Crispo J, Ansell D, Tai TC. Prenatal
glucocorticoid exposure programs adrenal PNMT expression and adult hypertension. J Endocrinol
2015 227 117-127.
44. Waddell BJ, Bollen M, Wyrwoll CS, Mori TA, Mark PJ. Developmental programming of adult
adrenal structure and steroidogenesis: effects of fetal glucocorticoid excess and postnatal dietary
omega-3 fatty acids. J Endocrinol 2010 205 171-178.
45. Drelon C, Berthon A, Sahut-Barnola I, Mathieu M, Dumontet T, Rodriguez S, Batisse-Lignier
M, Tabbal H, Tauveron I, Lefrancois-Martinez AM et al. PKA inhibits WNT signalling in adrenal cortex
zonation and prevents malignant tumour development. Nat Commun 2016 7 12751.
46. Lerario AM, Finco I, LaPensee C, Hammer GD. Molecular Mechanisms of Stem/Progenitor
Cell Maintenance in the Adrenal Cortex. Front Endocrinol (Lausanne) 2017 8 52.
47. Freedman BD, Kempna PB, Carlone DL, Shah M, Guagliardo NA, Barrett PQ, Gomez-Sanchez
CE, Majzoub JA, Breault DT. Adrenocortical zonation results from lineage conversion of
differentiated zona glomerulosa cells. Dev Cell 2013 26 666-673.
48. Xu A, Choi KL, Wang Y, Permana PA, Xu LY, Bogardus C, Cooper GJ. Identification of novel
putative membrane proteins selectively expressed during adipose conversion of 3T3-L1 cells.
Biochem Biophys Res Commun 2002 293 1161-1167.
49. Menssen A, Haupl T, Sittinger M, Delorme B, Charbord P, Ringe J. Differential gene
expression profiling of human bone marrow-derived mesenchymal stem cells during adipogenic
development. BMC Genomics 2011 12 461.
50. Zhang X, Saarinen AM, Campbell LE, De Filippis EA, Liu J. Regulation of Lipolytic Response
and Energy Balance by Melanocortin 2 Receptor Accessory Protein (MRAP) in Adipocytes. Diabetes
2018 67 222-234.
51. Dallman MF. Control of adrenocortical growth in vivo. Endocr Res 1984 10 213-242.
52. Yates R, Katugampola H, Cavlan D, Cogger K, Meimaridou E, Hughes C, Metherell L, Guasti L,
King P. Adrenocortical development, maintenance, and disease. Curr Top Dev Biol 2013 106 239-312.
53. Engeland WC, Ennen WB, Elayaperumal A, Durand DA, Levay-Young BK. Zone-specific cell
proliferation during compensatory adrenal growth in rats. Am J Physiol Endocrinol Metab 2005 288
E298-306.
54. Bland ML, Desclozeaux M, Ingraham HA. Tissue growth and remodeling of the embryonic
and adult adrenal gland. Ann N Y Acad Sci 2003 995 59-72.
55. Feuerstein B, Streeten DH. Recovery of adrenal function after failure resulting from
traumatic bilateral adrenal hemorrhages. Ann Intern Med 1991 115 785-786.
56. Skelton FR. Adrenal regeneration and adrenal-regeneration hypertension. Physiol Rev 1959
39 162-182.
57. McEwan PE, Lindop GB, Kenyon CJ. Control of cell proliferation in the rat adrenal gland in
vivo by the renin-angiotensin system. Am J Physiol 1996 271 E192-198.
58. Gorrigan RJ, Guasti L, King P, Clark AJ, Chan LF. Localisation of the melanocortin-2-receptor
and its accessory proteins in the developing and adult adrenal gland. J Mol Endocrinol 2011 46 227-
232.
Page 16 of 19Accepted Manuscript published as EC-19-0190.R1. Accepted for publication: 10-Jun-2019
Copyright © 2019 the authors Downloaded from Bioscientifica.com at 06/12/2019 01:24:10AM
via free access
17
59. Guasti L, Cavlan D, Cogger K, Banu Z, Shakur A, Latif S, King PJ. Dlk1 up-regulates Gli1
expression in male rat adrenal capsule cells through the activation of beta1 integrin and ERK1/2.
Endocrinology 2013 154 4675-4684.
60. Phillips R, Crock C, Funder J. Effects of mineralocorticoids and glucocorticoids on
compensatory adrenal growth in rats. Am J Physiol 1985 248 E450-456.
61. Engeland WC, Dallman MF. Compensatory adrenal growth is neurally mediated.
Neuroendocrinology 1975 19 352-362.
62. Grizzle WE, Dunlap NE. Aldosterone blocks adrenal compensatory hypertrophy in the rat.
Am J Physiol 1984 246 E306-310.
63. Estivariz FE, Carino M, Lowry PJ, Jackson S. Further evidence that N-terminal pro-
opiomelanocortin peptides are involved in adrenal mitogenesis. J Endocrinol 1988 116 201-206.
64. Dallman MF, Engeland WC, Holzwarth MA, Scholz PM. Adrenocorticotropin inhibits
compensatory adrenal growth after unilateral adrenalectomy. Endocrinology 1980 107 1397-1404.
65. Lowry PJ, Silas L, McLean C, Linton EA, Estivariz FE. Pro-gamma-melanocyte-stimulating
hormone cleavage in adrenal gland undergoing compensatory growth. Nature 1983 306 70-73.
66. Estivariz FE, Iturriza F, McLean C, Hope J, Lowry PJ. Stimulation of adrenal mitogenesis by N-
terminal proopiocortin peptides. Nature 1982 297 419-422.
67. Coll AP, Challis BG, Yeo GS, Snell K, Piper SJ, Halsall D, Thresher RR, O'Rahilly S. The effects of
proopiomelanocortin deficiency on murine adrenal development and responsiveness to
adrenocorticotropin. Endocrinology 2004 145 4721-4727.
68. Karpac J, Ostwald D, Bui S, Hunnewell P, Shankar M, Hochgeschwender U. Development,
maintenance, and function of the adrenal gland in early postnatal proopiomelanocortin-null mutant
mice. Endocrinology 2005 146 2555-2562.
69. Karpac J, Kern A, Hochgeschwender U. Pro-opiomelanocortin peptides and the adrenal
gland. Mol Cell Endocrinol 2007 265-266 29-33.
70. Coll AP, Fassnacht M, Klammer S, Hahner S, Schulte DM, Piper S, Tung YC, Challis BG,
Weinstein Y, Allolio B et al. Peripheral administration of the N-terminal pro-opiomelanocortin
fragment 1-28 to Pomc-/- mice reduces food intake and weight but does not affect adrenal growth
or corticosterone production. J Endocrinol 2006 190 515-525.
71. Karpac J, Czyzewska K, Kern A, Brush RS, Anderson RE, Hochgeschwender U. Failure of
adrenal corticosterone production in POMC-deficient mice results from lack of integrated effects of
POMC peptides on multiple factors. Am J Physiol Endocrinol Metab 2008 295 E446-455.
72. Vidal V, Sacco S, Rocha AS, da Silva F, Panzolini C, Dumontet T, Doan TM, Shan J, Rak-
Raszewska A, Bird T et al. The adrenal capsule is a signaling center controlling cell renewal and
zonation through Rspo3. Genes Dev 2016 30 1389-1394.
73. Mathieu M, Drelon C, Rodriguez S, Tabbal H, Septier A, Damon-Soubeyrand C, Dumontet T,
Berthon A, Sahut-Barnola I, Djari C et al. Steroidogenic differentiation and PKA signaling are
programmed by histone methyltransferase EZH2 in the adrenal cortex. Proc Natl Acad Sci U S A 2018
115 E12265-E12274.
74. Basham KJ, Rodriguez S, Turcu AF, Lerario AM, Logan CY, Rysztak MR, Gomez-Sanchez CE,
Breault DT, Koo BK, Clevers H et al. A ZNRF3-dependent Wnt/beta-catenin signaling gradient is
required for adrenal homeostasis. Genes Dev 2019 33 209-220.
75. Finco I, Lerario AM, Hammer GD. Sonic Hedgehog and WNT Signaling Promote Adrenal
Gland Regeneration in Male Mice. Endocrinology 2018 159 579-596.
76. King P, Paul A, Laufer E. Shh signaling regulates adrenocortical development and identifies
progenitors of steroidogenic lineages. Proc Natl Acad Sci U S A 2009 106 21185-21190.
77. Guasti L, Paul A, Laufer E, King P. Localization of Sonic hedgehog secreting and receiving cells
in the developing and adult rat adrenal cortex. Mol Cell Endocrinol 2011 336 117-122.
Page 17 of 19 Accepted Manuscript published as EC-19-0190.R1. Accepted for publication: 10-Jun-2019
Copyright © 2019 the authors Downloaded from Bioscientifica.com at 06/12/2019 01:24:10AM
via free access
18
Figure Legends
Figure 1. Model illustrating the morphological alterations and pathway
deregulation in the adrenal gland of FGD type 2.
In the normal adrenal cortex, activation of SHH and WNT4 signalling results in
driving the expression of CYP11B2, DAB2 and LEF1 leading to a zona glomerulosa
(ZG) lineage. Activation of PKA due to the action of ACTH on MC2R/MRAP complex
suppresses WNT4 resulting in inhibition of CYP11B2, DAB2 and LEF1 and
differentiation into zona fasciculata (ZF) cells. In FGD type 2 (Mrap-/- mice) adrenal
glands, the lack of PKA activation results in complete absence of ZF and WNT4/SHH
accumulation outside the ZG, where WNT4 expression in such cells do not lead to a
functional ZG identity. Morphologically the ZG is expanded and is able to secrete
aldosterone. The adrenal medulla in FGD type 2 is morphologically intact and
secretes both epinephrine and norepinephrine.
Page 18 of 19Accepted Manuscript published as EC-19-0190.R1. Accepted for publication: 10-Jun-2019
Copyright © 2019 the authors Downloaded from Bioscientifica.com at 06/12/2019 01:24:10AM
via free access
MC2R
MRAP
ACTH
PKA
MC2R
ACTH
PKA
ACTH
ACTH ACTH
ACTH
CYP11B1 (-)
CYP11B2 (-)
DAB2 (-)
LEF1 (-)
20-α-HSD (-)
CYP11B2 (+)
DAB2 (+)
LEF1 (+)
CYP11B2 (+)
DAB2 (+)
LEF1 (+)
capsular cell
zona glomerulosa cell
zona fasciculata cell
abnormal cells in FGD
Normal adrenal gland Adrenal gland in FGD type 2
CYP11B1 (+)
CYP11B2 (-)
DAB2 (-)
LEF1 (-)
WNT4/beta-catenin
SHH
*
*Activated Protein kinase A (PKA)
X zone
Inactive Protein kinase A (PKA)
PKA
PKA
*
PKA
ACTH
ACTH plasma ACTH
ALDO
ALDO
GC
adrenal medulla
CA
ALDO
GC
CA
aldosterone
glucocorticoids
catecholamines
CA
PNMT (+) PNMT (+)
X
Page 19 of 19 Accepted Manuscript published as EC-19-0190.R1. Accepted for publication: 10-Jun-2019
Copyright © 2019 the authors Downloaded from Bioscientifica.com at 06/12/2019 01:24:10AM
via free access
... MC2R is the most common genetic mutation in FGD-over 40 different MC2R mutations have been reported (accounting for 25% of FGD cases, termed FGD Type 1) [15]. Most are non-conservative missense mutations, which alter the MC2R protein structure so that it remains sequestered within the endoplasmic reticulum with a minority affecting ligand binding [8,12,15]. ...
... MRAP variants present earlier than MC2R mutations (often within one month of birth). People with MRAP mutations (termed FGD Type 2) account for 20% of solved FGD cases and typically have a more severe phenotype [12,14,15]. These splice site or non-sense mutations produce truncated MRAP proteins, which lack key transmembrane domains and are non-functional [14,15]. ...
... FGD is characterised by isolated GC deficiency due to ACTH resistance [8,11,15,41]. Serum cortisol levels are often undetectable with very high ACTH, but normal mineralocorticoid concentrations and electrolytes [8,11,12,41]. ...
Article
Full-text available
Cortisol is central to several homeostatic mechanisms including the stress and immune response. Adrenal insufficiency and impaired cortisol production leads to severe, potentially fatal disorders. Several fundamental stages of steroidogenesis occur within the mitochondria. These dynamic organelles not only contribute ATP for steroidogenesis, but also detoxify harmful by-products generated during cortisol synthesis (reactive oxygen species). Mutations in nuclear or mitochondrial DNA that impair mitochondrial function lead to debilitating multi-system diseases. Recently, genetic variants that impair mitochondrial function have been identified in people with isolated cortisol insufficiency. This review aimed to clarify the association between mitochondrial diseases and adrenal insufficiency to produce cortisol. Mitochondrial diseases are rare and mitochondrial diseases that feature adrenal insufficiency are even rarer. We identified only 14 cases of adrenal insufficiency in people with confirmed mitochondrial diseases globally. In line with previous reviews, adrenal dysfunction was most prevalent in mitochondrial deletion syndromes (particularly Pearson syndrome and Kearns–Sayre syndrome) and with point mutations that compromised oxidative phosphorylation. Although adrenal insufficiency has been reported with mitochondrial diseases, the incidence reflects that expected in the general population. Thus, it is unlikely that mitochondrial mutations alone are responsible for an insufficiency to produce cortisol. More research is needed into the pathogenesis of adrenal disease in these individuals.
... FGD is also caused by mutations in the MRAP (melanocortin 2 receptor accessory protein) gene [73,74], which is required for proper function and transport of the MC2R protein to the cell membrane location [32]. Mrap −/− knockout mice have been found to mimic the human FGD phenotype [73,75]. Individuals with FGD typically present with symptoms of adrenal insufficiency, including fatigue, weakness, hypoglycaemia, poor weight gain, and potentially life-threatening adrenal crises [32]. ...
Chapter
Full-text available
Steroidogenesis, the process by which steroids are synthesized, involves a complex cascade of enzymatic reactions that ultimately produce hormones, such as cortisol and aldosterone. Cortisol is a steroid hormone that plays a critical role in the regulation of various physiological processes, including metabolism, immune response, and stress response. Aldosterone is responsible for blood pressure and water balance. The biosynthesis of cortisol and aldosterone occurs primarily in the adrenal cortex and is processed by a series of enzymatic reactions that convert cholesterol into cortisol and aldosterone. Enzymes include CYP11A1, 3β-hydroxysteroid dehydrogenase 2, CYP11B1, CYP11B2, CYP17A1, and 21-hydroxylase. Mutations or defects in these enzymes can lead to impaired cortisol and aldosterone biosynthesis, thereby resulting in various disorders such as congenital adrenal hyperplasia, adrenal hypoplasia congenita, and familial glucocorticoid deficiency. Endocrine disruptors, such as phthalates, bisphenols, and pesticides, affect adrenal cortex development or steroidogenesis, thereby causing adrenal cortex dysfunction. Understanding the complex process of steroidogenesis involved in cortisol and aldosterone biosynthesis can provide crucial insights into the pathophysiology of adrenal disorders and inform the development of targeted therapies to alleviate the associated symptoms.
... On the one hand, the WNT/Rspondin/β-catenin pathway is necessary for the maintenance of progenitor pools and the acquisition of zG identity [4][5][6] . On the other hand, cAMP/PKA signalling, stimulated by pituitary ACTH, triggers the recruitment of progenitors by inducing the transdifferentiation of zG cells into zF cells and stimulates glucocorticoids production 7,8 . We and others have previously shown that these two signalling pathways antagonise each other by modulating various actors such as WNT4, PDE2A or CCDC80 3,6,9 . ...
Article
Full-text available
SUMOylation is a dynamic posttranslational modification, that provides fine-tuning of protein function involved in the cellular response to stress, differentiation, and tissue development. In the adrenal cortex, an emblematic endocrine organ that mediates adaptation to physiological demands, the SUMOylation gradient is inversely correlated with the gradient of cellular differentiation raising important questions about its role in functional zonation and the response to stress. Considering that SUMO-specific protease 2 (SENP2), a deSUMOylating enzyme, is upregulated by Adrenocorticotropic Hormone (ACTH)/cAMP-dependent Protein Kinase (PKA) signalling within the zona fasciculata , we generated mice with adrenal-specific Senp2 loss to address these questions. Disruption of SENP2 activity in steroidogenic cells leads to specific hypoplasia of the zona fasciculata , a blunted reponse to ACTH and isolated glucocorticoid deficiency. Mechanistically, overSUMOylation resulting from SENP2 loss shifts the balance between ACTH/PKA and WNT/β-catenin signalling leading to repression of PKA activity and ectopic activation of β-catenin. At the cellular level, this blocks transdifferentiation of β-catenin-positive zona glomerulosa cells into fasciculata cells and sensitises them to premature apoptosis. Our findings indicate that the SUMO pathway is critical for adrenal homeostasis and stress responsiveness.
... MRAP1 was first identified as the specific chaperone for MC2R, essential for MC2R forward trafficking [28,29,33,34]. Human (h) MRAP1 mutations account for~20% of familial glucocorticoid deficiency cases [28,35]. Subsequent studies suggest that MRAP1 possesses functions beyond regulating MC2R. ...
Article
Full-text available
The neural melanocortin receptors (MCRs), melanocortin-3 and -4 receptors (MC3R and MC4R), have crucial roles in regulating energy homeostasis. The melanocortin-2 receptor accessory proteins (MRAPs, MRAP1 and MRAP2) have been shown to regulate neural MCRs in a species-specific manner. The potential effects of MRAP1 and MRAP2 on canine neural MCRs have not been investigated before. Herein, we cloned canine (c) MC3R and identified one canine MRAP2 splice variant, MRAP2b, with N-terminal extension of cMRAP2a. Canine MC3R showed higher maximal responses to five agonists than those of human MC3R. We further investigated the modulation of cMRAP1, cMRAP2a, and cMRAP2b, on cMC3R and cMC4R pharmacology. For the cMC3R, all MRAPs had no effect on trafficking; cMRAP1 significantly decreased Bmax whereas cMRAP2a and cMRAP2b significantly increased Bmax. Both MRAP1 and MRAP2a decreased Rmaxs in response to α-MSH and ACTH; MRAP2b only decreased α-MSH-stimulated cAMP generation. For the MC4R, MRAP1 and MRAP2a increased cell surface expression, and MRAP1 and MRAP2a increased Bmaxs. All MRAPs had increased affinities to α-MSH and ACTH. MRAP2a increased ACTH-induced cAMP levels, whereas MRAP2b decreased α-MSH- and ACTH-stimulated cAMP production. These findings may lead to a better understanding of the regulation of neural MCRs by MRAP1 and MRAP2s.
... Only higher primates share a similar structure and function of the adrenal cortex with humans, while rodents show remarkable differences in both structure and function that need to be considered when they are investigated as models. Nevertheless, much of the current knowledge of fetal adrenal development derives from mice models [7]. Only more recently, spatial and temporal gene expression profiling using single-cell sequencing on human fetal material has provided additional insight into developmental pathways [8,9], and reprogramming of human induced pluripotent stem cells towards adrenal cell lines or construction of adrenal organoids are currently hot topics in the field [10,11]. ...
Article
Full-text available
The adrenal cortex undergoes multiple structural and functional rearrangements to satisfy the systemic needs for steroids during fetal life, postnatal development, and adulthood. A fully functional adrenal cortex relies on the proper subdivision in regions or ‘zones’ with distinct but interconnected functions, which evolve from the early embryonic stages to adulthood, and rely on a fine-tuned gene network. In particular, the steroidogenic activity of the fetal adrenal is instrumental in maintaining normal fetal development and growth. Here, we review and discuss the most recent advances in our understanding of embryonic and fetal adrenal development, including the known causes for adrenal dys-/agenesis, and the steroidogenic pathways that link the fetal adrenal with the hormone system of the mother through the fetal-placental unit. Finally, we discuss what we think are the major open questions in the field, including, among others, the impact of osteocalcin, thyroid hormone, and other hormone systems on adrenal development and function, and the reliability of rodents as models of adrenal pathophysiology.
... In vertebrates, corticotropin (ACTH), which is released from the anterior pituitary gland, plays important roles in stress response and other physiological activities, including steroid hormone biosynthesis and adrenal development [1][2][3]. As the critical part of the hypothalamus-pituitary-adrenal (HPA) axis, the biosynthesis and secretion of ACTH are controlled by both hypothalamic stimulatory factors and inhibitory factors (e.g., CRIF) [4][5][6]. ...
Article
Full-text available
Corticotropin (ACTH) is a pituitary hormone playing important roles in stress response within the hypothalamus–pituitary–adrenal (HPA) axis. The biosynthesis and secretion of ACTH are controlled by multiple factors, including corticotropin-releasing hormone (CRH). As a key hypothalamus-derived regulator, CRH binds to corticotropin-releasing hormone receptor 1 (CRHR1) in the anterior pituitary gland to regulate ACTH synthesis and release. Thus, CRH-binding protein (CRHBP), which binds CRH with high affinity to inhibit CRH-induced ACTH secretion from pituitary cells, draws wide attention. In contrast to the extensive investigation of CRHBP in mammals and other lower vertebrates, the gene structure, tissue expression and physiological functions of CRHBP in birds remain largely unknown. In the present study, using chicken (c-) as our animal model, we examined the gene structure, tissue expression and functionality of CRHBP. Our results showed that: (1) cCRHBP cDNA encodes a 345 amino acid precursor, which shares high sequence identity with that of mammals, reptiles, frogs and fish; (2) cCRHBP is abundantly expressed in the brain (cerebrum and hypothalamus), pituitary and ovary; (3) cCRHBP inhibits the signaling of cCRHRs induced by cCRH, thus reducing the cCRH-induced ACTH secretion from cultured chick pituitary cells; (4) stress mediators (e.g., glucocorticoids) and stress significantly upregulate CRHBP mRNA expression in chickens, supporting its role as a negative feedback regulator in the HPA axis. The present study enriches our understanding of the conserved roles of CRHBP across vertebrates. In addition, chicken is an important poultry animal with multiple economic traits which are tightly controlled by the HPA axis. The characterization of the chicken CRHBP gene helps to reveal the molecular basis of the chicken HPA axis and is thus beneficial to the poultry industry.
Article
Full-text available
Spatiotemporal control of Wnt signaling is essential for the development and homeostasis of many tissues. The transmembrane E3 ubiquitin ligases ZNRF3 (zinc and ring finger 3) and RNF43 (ring finger protein 43) antagonize Wnt signaling by promoting degradation of frizzled receptors. ZNRF3 and RNF43 are frequently inactivated in human cancer, but the molecular and therapeutic implications remain unclear. Here, we demonstrate that adrenocortical-specific loss of ZNRF3, but not RNF43, results in adrenal hyperplasia that depends on Porcupine-mediated Wnt ligand secretion. Furthermore, we discovered a Wnt/β-catenin signaling gradient in the adrenal cortex that is disrupted upon loss of ZNRF3. Unlike β-catenin gain-of-function models, which induce high Wnt/β-catenin activation and expansion of the peripheral cortex, ZNRF3 loss triggers activation of moderate-level Wnt/β-catenin signaling that drives proliferative expansion of only the histologically and functionally distinct inner cortex. Genetically reducing β-catenin dosage significantly reverses the ZNRF3-deficient phenotype. Thus, homeostatic maintenance of the adrenal cortex is dependent on varying levels of Wnt/β-catenin activation, which is regulated by ZNRF3. © 2019 Basham et al.; Published by Cold Spring Harbor Laboratory Press.
Article
Full-text available
Adrenal cortex steroids are essential for body homeostasis, and adrenal insufficiency is a life-threatening condition. Adrenal endocrine activity is maintained through recruitment of subcapsular progenitor cells that follow a unidirectional differentiation path from zona glomerulosa to zona fasciculata (zF). Here, we show that this unidirectionality is ensured by the histone methyltransferase EZH2. Indeed, we demonstrate that EZH2 maintains adrenal steroidogenic cell differentiation by preventing expression of GATA4 and WT1 that cause abnormal dedifferentiation to a progenitor-like state in Ezh2 KO adrenals. EZH2 further ensures normal cortical differentiation by programming cells for optimal response to adrenocorticotrophic hormone (ACTH)/PKA signaling. This is achieved by repression of phosphodiesterases PDE1B, 3A, and 7A and of PRKAR1B. Consequently, EZH2 ablation results in blunted zF differentiation and primary glucocorticoid insufficiency. These data demonstrate an all-encompassing role for EZH2 in programming steroidogenic cells for optimal response to differentiation signals and in maintaining their differentiated state.
Article
Full-text available
Melanocortin 2 receptor accessory protein (MRAP) is a single transmembrane domain accessory protein and a critical component of the hypothamo-pituitary-adrenal axis. MRAP is highly expressed in the adrenal gland and is essential for adrenocorticotropin hormone (ACTH) receptor expression and function. Human loss-of-function mutations in MRAP cause familial glucocorticoid (GC) deficiency (FGD) type 2 (FGD2), whereby the adrenal gland fails to respond to ACTH and to produce cortisol. In this study, we generated Mrap-null mice to study the function of MRAP in vivo. We found that the vast majority of Mrap-/- mice died at birth but could be rescued by administration of corticosterone to pregnant dams. Surviving Mrap-/- mice developed isolated GC deficiency with normal mineralocorticoid and catecholamine production, recapitulating FGD2. The adrenal glands of adult Mrap-/- mice were small, with grossly impaired adrenal capsular morphology and cortex zonation. Progenitor cell differentiation was significantly impaired, with dysregulation of WNT4/β-catenin and sonic hedgehog pathways. These data demonstrate the roles of MRAP in both steroidogenesis and the regulation of adrenal cortex zonation. This is the first mouse model of isolated GC deficiency and reveals the role of MRAP in adrenal progenitor cell regulation and cortex zonation.-Novoselova, T. V., Hussain, M., King, P. J., Guasti, L., Metherell, L. A., Charalambous, M., Clark, A. J. L., Chan, L. F. MRAP deficiency impairs adrenal progenitor cell differentiation and gland zonation.
Article
Full-text available
The adrenal cortex is characterized by three histologically and functionally distinct zones: the outermost zona glomerulosa (zG), the intermediate zona fasciculata, and the innermost zona reticularis. Important aspects of the physiology and maintenance of the adrenocortical stem/progenitor cells have emerged in the last few years. Studies have shown that the adrenocortical cells descend from a pool of progenitors that are localized in the subcapsular region of the zG. These cells continually undergo a process of centripetal displacement and differentiation, which is orchestrated by several paracrine and endocrine cues, including the pituitary-derived adrenocorticotrophic hormone, and angiotensin II. However, while several roles of the endocrine axes on adrenocortical function are well established, the mechanisms coordinating the maintenance of an undifferentiated progenitor cell pool with self-renewal capacity are poorly understood. Local factors, such as the composition of the extracellular matrix (ECM) with embedded signaling molecules, and the activity of major paracrine effectors, including ligands of the sonic hedgehog and Wnt signaling pathways, are thought to play a major role. Particularly, the composition of the ECM, which exhibits substantial differences within each of the three histologically distinct concentric zones, has been shown to influence the differentiation status of adrenocortical cells. New data from other organ systems and different experimental paradigms strongly support the conclusion that the interactions of ECM components with cell-surface receptors and secreted factors are key determinants of cell fate. In this review, we summarize established and emerging data on the paracrine and autocrine regulatory loops that regulate the biology of the progenitor cell niche and propose a role for bioengineered ECM models in further elucidating this biology in the adrenal.
Article
Pro-opiomelanocortin (POMC) is the archetypal polypeptide precursor of hormones and neuropeptides. In this review, we examine the variability in the individual peptides produced in different tissues and the impact of the simultaneous presence of their precursors or fragments. We also discuss the problems inherent in accurately measuring which of the precursors and their derived peptides are present in biological samples. We address how not being able to measure all the combinations of precursors and fragments quantitatively has affected our understanding of the pathophysiology associated with POMC processing. To understand how different ratios of peptides arise, we describe the role of the pro-hormone convertases (PCs) and their tissue specificities and consider the cellular processing pathways which enable regulated secretion of different peptides that play crucial roles in integrating a range of vital physiological functions. In the pituitary, correct processing of POMC peptides is essential to maintain the hypothalamic-pituitary-adrenal axis, and this processing can be disrupted in POMC-expressing tumors. In hypothalamic neurons expressing POMC, abnormalities in processing critically impact on the regulation of appetite, energy homeostasis, and body composition. More work is needed to understand whether expression of the POMC gene in a tissue equates to release of bioactive peptides. We suggest that this comprehensive view of POMC processing, with a focus on gaining a better understanding of the combination of peptides produced and their relative bioactivity, is a necessity for all involved in studying this fascinating physiological regulatory phenomenon.
Article
The melanocortin receptors (MCRs) and their accessory proteins (MRAPs) are involved in regulation of a diverse range of endocrine pathways. Genetic variants of these components result in phenotypic variation and disease. The MC1R is expressed in skin and variants in the MC1R gene are associated with ginger hair color. The MC2R mediates the action of ACTH in the adrenal gland to stimulate glucocorticoid production and MC2R mutations result in familial glucocorticoid deficiency (FGD). MC3R and MC4R are involved in metabolic regulation and their gene variants are associated with severe pediatric obesity, whereas the function of MC5R remains to be fully elucidated. MRAPs have been shown to modulate the function of MCRs and genetic variants in MRAPs are associated with diseases including FGD type 2 and potentially early onset obesity. This review provides an insight into recent advances in MCRs and MRAPs physiology, focusing on the disorders associated with their dysfunction.
Article
The adrenal cortex undergoes remodeling during fetal and postnatal life. How zona reticularis emerges in the postnatal gland to support adrenarche, a process whereby higher primates increase prepubertal androgen secretion, is unknown. Using cell-fate mapping and gene deletion studies in mice, we show that activation of PKA has no effect on the fetal cortex, while it accelerates regeneration of the adult cortex, triggers zona fasciculata differentiation that is subsequently converted into a functional reticularis-like zone, and drives hypersecretion syndromes. Remarkably, PKA effects are influenced by sex. Indeed, testicular androgens increase WNT signaling that antagonizes PKA, leading to slower adrenocortical cell turnover and delayed phenotype whereas gonadectomy sensitizes males to hypercorticism and reticularis-like formation. Thus, reticularis results from ultimate centripetal conversion of adult cortex under the combined effects of PKA and cell turnover that dictate organ size. We show that PKA-induced progenitor recruitment is sexually dimorphic and may provide a paradigm for overrepresentation of women in adrenal diseases.
Article
MRAP is highly expressed in adrenal gland and adipose tissue. In adrenal cells, MRAP is essential for ACTH-induced activation of the cAMP/PKA pathway by melanocortin 2 receptor (MC2R), leading to glucocorticoid production and secretion. Although ACTH was known to stimulate PKA-dependent lipolysis, the functional involvement of MRAP in adipocyte metabolism remains incompletely defined. Herein, we found that knockdown or overexpression of MRAP in 3T3-L1 adipocytes reduced or increased ACTH-induced lipolysis, respectively. Moreover, an unbiased proteomics screen and coimmunoprecipitation analysis identified Gαs as a novel interacting partner of MRAP. A MRAP mutant disabled in Gαs association failed to augment the activation of PKA and lipolytic response to ACTH. Furthermore, compared to wild type mice, transgenic mice (aP2-MRAP) overexpressing MRAP fat specifically exhibited increased lipolytic response to ACTH. When fed a high-fat diet (HFD), the transgenic mice displayed a significant decrease in the gain of adiposity and body weight as well as an improvement in glucose and insulin tolerance. These phenotypes were accompanied by increased adipose expression of genes for mitochondrial fatty acid oxidation and thermogenesis, and overall energy expenditure. Collectively, our data strongly suggest that MRAP plays a critical role in the regulation of ACTH-induced adipose lipolysis and whole-body energy balance.
Article
The atrophy and hypofunction of the adrenal cortex following long-term pharmacologic glucocorticoid therapy are a major health problem necessitating chronic glucocorticoid replacement that often prolongs the ultimate return of endogenous adrenocortical function. Underlying this functional recovery is anatomic regeneration, the cellular and molecular mechanisms of which are poorly understood. Investigating the lineage contribution of cortical Shh+ progenitor cells and the SHH-responsive capsular Gli1+ cells to the regenerating adrenal cortex, we observed a spatially and temporally bimodal contribution of both cell types to adrenocortical regeneration following cessation of glucocorticoid treatment. First, an early repopulation of the cortex is defined by a marked delamination and expansion of capsular Gli1+ cells, recapitulating the establishment of the capsular-cortical homeostatic niche during embryonic development. This rapid repopulation is promptly cleared from the cortical compartment only to be supplanted by repopulating cortical cells derived from the resident long-term-retained zG Shh+ progenitors. Pharmacologic and genetic dissection of Shh signaling further defines a SHH-dependent activation of WNT signaling that supports regeneration of the cortex following long-term glucocorticoid therapy. We define the signaling and lineage relationships that underlie the regeneration process.