Vous êtes sur la page 1sur 28

REVIEW

The Biology of Normal Zona Glomerulosa


and Aldosterone-Producing Adenoma:
Pathological Implications

Downloaded from https://academic.oup.com/edrv/article-abstract/39/6/1029/5051322 by Research 4 Life user on 22 November 2018


Teresa M. Seccia,1 Brasilina Caroccia,1 Elise P. Gomez-Sanchez,2 Celso E. Gomez-Sanchez,3,4
and Gian Paolo Rossi1

1
Department of Medicine-DIMED, University of Padua, 35122 Padua PD, Italy; 2Department of Pharmacology
and Toxicology, G.V. (Sonny) Montgomery VA Medical Center, Jackson, Mississippi 39216; 3Division of
Endocrinology, G.V. (Sonny) Montgomery VA Medical Center, Jackson, Mississippi 39216; and 4University of
Mississippi Medical Center, Jackson, Mississippi 39216

ABSTRACT The identification of several germline and somatic ion channel mutations in aldosterone-producing adenomas (APAs) and
detection of cell clusters that can be responsible for excess aldosterone production, as well as the isolation of autoantibodies activating the
angiotensin II type 1 receptor, have rapidly advanced the understanding of the biology of primary aldosteronism (PA), particularly that of
APA. Hence, the main purpose of this review is to discuss how discoveries of the last decade could affect histopathology analysis and clinical
practice. The structural remodeling through development and aging of the human adrenal cortex, particularly of the zona glomerulosa, and
the complex regulation of aldosterone, with emphasis on the concepts of zonation and channelopathies, will be addressed. Finally, the
diagnostic workup for PA and its subtyping to optimize treatment are reviewed. (Endocrine Reviews 39: 1029 – 1056, 2018)

A ldosterone, the main mineralocorticoid hor-


mone, is vital for maintaining body fluid
and electrolyte homeostasis, vascular resistance, and,
knowledge in hypertension. Multiple animal models
of hyperaldosteronism, including the knockout of
TWIK acid–sensitive potassium (TASK) channels
thereby, blood pressure under conditions of salt/water types  and , replicate the PA phenotype in mice ().
(volume) depletion. However, aldosterone levels are in- Aldosterone-producing adenomas (APAs) are re-
appropriately high in % to % of all hypertensive sponsible for about half of PA. Study of APAs has been
disorders, not only primary and secondary aldosteronism instrumental in recent advances in our understanding
but also overweight and obesity (, ) and the most severe of the regulation of normal and pathological aldo-
(stage II to III) or drug-resistant forms of hypertension (). sterone synthesis. Whole transcriptome analysis has
This indicates that aldosterone produced in excess with demonstrated consistent underexpression of the TASK-
respect to sodium status is a main determinant of high channels in APAs, and in vitro molecular blunting of
blood pressure, but it is clear from clinical studies from TASK- enhances aldosterone production (). This
around the world that inappropriately high aldosterone TASK- underexpression could be attributed to
concentrations produce prominent target organ dam- elevated miRNA  and  levels in ~% of the
age, thus contributing to an ominous prognosis (–). APAs and to functional genetic variants in the TASK-
Primary aldosteronism (PA) occurs in % to % of promoter in another quarter of the cases (), leaving
hypertensive patients (, ) but is often overlooked the mechanism for nearly half of the cases unex-
because patients are not screened for it. PA can mimic plained to date.
primary (essential) hypertension, and is still perceived Elevated serum parathyroid hormone levels have ISSN Print: 0163-769X
as an exceptionally rare condition necessitating a been noted in patients with PA, and type  parathyroid ISSN Online: 1945-7189
complex diagnostic workup, given the lack of known hormone receptor has been found in APAs (). The Printed: in USA
mechanisms and therefore of specific biomarkers. In demonstration that parathyroid hormone stimulates Copyright © 2018
Endocrine Society
fact, since its discovery in  the term primary has aldosterone secretion and potentiates the effect of
Received: 18 February 2018
been used to emphasize our ignorance of its causes. angiotensin II (Ang II) and K+ (, ) highlighted Accepted: 3 July 2018
In the last decade, multiple seminal discoveries hitherto unknown interactions between the para- First Published Online:
have made PA the paradigm of improved mechanistic thyroid gland and the adrenocortical zona glomerulosa. 10 July 2018

doi: 10.1210/er.2018-00060 https://academic.oup.com/edrv 1029


REVIEW

ESSENTIAL POINTS
· In this review we examine the biology of the normal zona glomerulosa through development and with aging and then
analyze the functional role of the major regulators of aldosterone secretion
· We discuss the theories that explain the transition of the normal zona glomerulosa into a multinodular disorder,
eventually leading to formation of an aldosterone-producing adenoma (APA), with particular focus on the activation of
signaling pathways such as wingless-related integration site/b-catenin and somatic mutations of ion channels leading to
zona glomerulosa cell depolarization and aldosterone production
· Clinical aspects, such as epidemiology and diagnosis of primary aldosteronism (PA), are briefly reviewed, and the potential

Downloaded from https://academic.oup.com/edrv/article-abstract/39/6/1029/5051322 by Research 4 Life user on 22 November 2018


role of circulating and tissue biomarkers of APA in subtyping PA is discussed

Agonistic autoantibodies for the type  angiotensin re- including that of ATPA, ATPB, CACNAD,
ceptor () were also found to increase aldosterone CACNAH, CTNNB (b-catenin), and CLCN
production (, ). chloride channel (–), thus providing compelling
In  the results of exome sequencing of  evidence that channelopathies and malfunctioning
APAs led Choi et al. () to uncover mutations in the signaling pathways are involved in the pathophysiology
selectivity filter of the Kir. (KCNJ) K+ channel. of many cases of PA (, ).
Subsequent studies demonstrated that KCNJ muta- Aldosterone plays a key role in causing not only
tions were present in approximately one-third of APAs arterial hypertension but also heart failure, where it
in a large European cohort () and between % and contributes substantively to cardiovascular and renal
% in a much larger meta-analysis of studies carried damage and events (–). However, our knowledge
out worldwide (). Germline KCNJ mutations were of the fundamental biology of the normal zona glo-
also found in rare familial forms of PA featuring drug- merulosa remains limited. The purpose of this review is
resistant hypertension and massive bilateral adrenal to update information about the complex regulation of
hyperplasia necessitating bilateral adrenalectomy (). aldosterone, with a particular emphasis on the clinical
This finding was soon followed by the discovery of and pathological implications of discoveries in the last
mutations in other genes affecting ion channel function, decade.

The Adrenal Cortex: Historical Perspective mineralocorticoid from an amorphous fraction of beef
adrenal extracts (), which they initially named
The earliest detailed description of the paired human electrocortin for its activity and, later, aldosterone, once
suprarenal or adrenal glands in European literature is it was recognized that the molecule exists as a tau-
ascribed to Bartolomeo Eustachi (Eustachius) in . tomer between an aldehyde at position  and forms a
His detailed drawings of the glands were hidden hemiacetal with the b hydroxyl (). Within
during the Inquisition and published  years later months, groups in America and Switzerland con-
by Giovanni Maria Lancisi as part of the Tabulae firmed these findings (). In , Lityńsky () in
Anatomicae (Anatomical Engravings) de Bartolomeo Poland and Jerome Conn, an endocrinologist at the
Eustachii (, ). Description of the structure of the University of Michigan, independently reported the
adrenal gland progressed little for the next three first two clinical cases of APA causing the syndrome of
centuries except for the recognition that it comprised a primary hyperaldosteronism, featuring hypertension,
distinct medulla and cortex (Cuvier in ) and hypokalemia due to abnormal potassium excretion,
description of the zonation of the cortex by Gottschau and hypomagnesemia, which were cured by adre-
et al. in  (). By the late s, most of the nalectomy ().
glucocorticoids produced by the adrenal cortex were
isolated and their structures defined, but all bioassays
developed to characterize adrenal cortical extracts Zonation of the Adrenal Cortex
failed to isolate the fraction containing mineralocor-
ticoid activity (), generating a -year debate over Anatomical and histological features
whether biologically relevant mineralocorticoids The adrenal gland may be roughly triangular or
existed, with the majority of researchers contending crescent-shaped to conform to the contour of the
that glucocorticoids were the major source of min- dorsal pole of the kidneys, as in humans, or spherical
eralocorticoid activity (). It was only in  that and held slightly apart from the kidney by fat and
Simpson and Tait in London, in collaboration with fibrous tissue, as in rats and mice. From the capsule
Reichstein from the University of Basel and the then encasing the adrenal gland, moving centripetally to-
Ciba Pharmaceutica Company, isolated an active ward the medulla, the cortex comprises the zona

1030 Seccia et al Biology of Zona Glomerulosa and APA Endocrine Reviews, December 2018, 39(6):1029–1056
REVIEW

glomerulosa, made of densely packed cells with com- -hydroxylase, corticosterone is the primary glucocor-
paratively little cytoplasm forming glomeruli (or balls); ticoid, and no androgens are produced in the smaller
the zona fasciculata, composed of cells with copious innermost zona fasciculata cells next to the medulla. Thus,
cytoplasm, containing numerous lipid droplets arranged these species do not have a true zona reticularis, although
in fascicles or columns; and, in some species, notably their zona fasciculata cells may have a reticular or netlike
humans, the zona reticularis, next to the medulla and appearance close to the medulla. The mouse fetal X-zone,
composed of cells slightly smaller than those of the zona which expresses a-hydroxysteroid dehydrogenase that
fasciculata, arranged to form a net (, ). The distinct catabolizes progesterone, regresses in the male at about
morphology, arrangement, and steroid production of the the time it is weaned but persists in the female until first
pregnancy (, ), and it should not be confused with a

Downloaded from https://academic.oup.com/edrv/article-abstract/39/6/1029/5051322 by Research 4 Life user on 22 November 2018


adrenocortical cells within each zone are phylogenetically
conserved (, ). zona reticularis.
Once antibodies were made that distinguished
Steroidogenesis and steroidogenic enzymes between highly homologous CYPB and CYPB
Cloning of the genes for the enzymes in the ste- enzymes of the rat and, relatively recently, the human,
roidogenic cascades for the synthesis of the adrenal it became clear that a narrow zona intermedia between
steroids confirmed the arduous enzymology tour de the zona glomerulosa and zona fasciculata, in which
force necessary to isolate the multiple enzymes and cells express neither CYPB nor CYPB, is evi-
their steroid substrates specific to each zona of the dent in the rat but is not clearly defined in humans
adrenal cortex. Enzymes for the conversion of cho- (–). Whether this is due in part to high chronic
lesterol to deoxycorticosterone (DOC), cytochrome P sodium (Na+) intake of humans or is a species dif-
side-chain cleavage (CYPA), b-hydroxysteroid ference is not clear. Though morphologically similar,
dehydrogenase- (HSDB), and -hydroxylase many zona glomerulosa cells of the adult human do not
(CYPA) are expressed in the zona glomerulosa express substantial amounts of CYPB protein.
and zona fasciculata. The human has two highly Some animals, notably cattle, pigs, and sheep, express
homologous HSDB enzymes; HSDB is expressed a-hydroxylase (CYP) in the zona fasciculata
primarily in the adrenal, and hydroxy-delta--steroid and zona reticularis but have a single cytochrome
dehydrogenase (HSDB) is expressed in extra-adrenal P–b hydroxylase, CYPB, in both the zona
tissues. The mouse has six different homologous iso- fasciculata and zona glomerulosa. In these species, the
“A narrow zona intermedia…is
enzymes. A recent study of mice with the clock genes CYPB acts as a sequential hydroxylase to syn- evident in the rat but is not
cry and cry deleted showed that development of thesize only aldosterone in the zona glomerulosa, a clearly defined in humans.”
hyperaldosteronism correlates with increased expres- function provided by CYPB in the human and rat
sion of the hsdb isozyme (corresponding to HSDB ().
in the human) in the zona glomerulosa (). One recent
study indicated that HSDB was also expressed in the Ontogeny
zona glomerulosa, whereas HSDB was expressed in The adrenal gland comprises tissues of two different
the zonae fasciculata and glomerulosa (). However, embryological origins with very different functions.
another study using highly specific antibodies against The adrenal medulla develops from neural crest cells,
both isozymes demonstrated that HSDB was progenitors of chromaffin cells, and the adrenal cortex
expressed at either very low or negligible levels in from the intermediate mesoderm (). The adrenal
the zona glomerulosa and confirmed that HSDB cortex, ovary, and testis arise from a common pro-
was highly expressed in the zonae glomerulosa and genitor, the adrenogonadal primordium, a specialized
fasciculata (). region of the celomic epithelium called the urogenital
Aldosterone synthase (CYPB), which converts ridge (, ) (Fig. ). The urogenital ridge is also the
DOC to aldosterone, is limited to the zona glomerulosa; origin for the kidneys and the progenitors of the
b-hydroxylase (CYPB), which converts DOC to definitive hematopoietic cells (, , ).
corticosterone, is in the zona fasciculata (Fig. ). In species, During the fourth to sixth week of gestation in the
including the human, that express a-hydroxylase/ human, and around day . in the mouse, there is a
,-lyase (CYPA), necessary for the synthesis of thickening of the celomic epithelium in the urogenital
a-hydroxypregnenolone and a-hydroxyprogester- ridge to form the adrenogonadal primordium. Shortly
one, CYPB converts -deoxycortisol to cortisol in the after at the eighth week in the human and . days in
zona fasciculata. The a--lyase activity of CYPA the mouse, the primordium divides into the dorso-
produces androstenedione in the zona fasciculata and medial portion to form the adrenal primordia and
dehydroepiandrosterone (DHEA) in the zona retic- the ventrolateral portion for the gonadal primordia
ularis, where HSDB is low. By volume, the primary (–). At this stage, use of in situ hybridization
corticosteroids in the human are cortisol, DHEA, and demonstrates that the adrenal gland expresses the
aldosterone synthesized in adrenal zonae fasciculata, transcription factors, steroidogenic factor- (SF; also
reticularis, and glomerulosa, respectively. In species, called ADBP or NRA) and dosage-sensitive sex
including mouse and rat, that do not express an adrenal reversal–adrenal hypoplasia congenital critical region

doi: 10.1210/er.2018-00060 https://academic.oup.com/edrv 1031


REVIEW

Figure 1. Steroidogenic
pathways in the human
adrenal cortex. Enzymes
localized in the mitochondria
are in yellow boxes.
[© 2018 Illustration
Presentation ENDOCRINE
SOCIETY]

Downloaded from https://academic.oup.com/edrv/article-abstract/39/6/1029/5051322 by Research 4 Life user on 22 November 2018


on the X chromosome (DAX; also known as NRB) poor during midgestation and appear to be the res-
(). Expression of SF exceeds that of DAX at all ervoir of typical proliferative cells, many of which are
embryonic stages studied (). The cells from the mitotically active. Midkine, a heparin-binding growth
neural crest invade the adrenal primordium around factor, selectively stimulates proliferation of the cells
weeks  to  of human gestation or day  in the from the definitive zone of the human fetal adrenal
mouse, before the adrenal primordium is surrounded cortex ().
by mesenchymal cells to form the capsule (). The As gestation advances, these cells form fingerlike
human and primate fetal adrenal cortex exhibits two columns of cells close to the outer rim of the fetal zone.
structurally defined areas, an inner “fetal zone” Before week , a third zone, named the transitional
comprising large polyhedral eosinophilic cells, which zone, appears between the definitive zone and the fetal
begins to regress during the fifth month of gestation zone in humans (–); this transitional zone is not
and disappears totally  year after birth, and a pe- as evident in mice. During the last trimester the
ripheral zone adjacent to the capsule comprised of transitional zone, which eventually becomes the zona
small basophilic cells that persists after birth and is fasciculata of the adult human adrenal cortex, expands
therefore called the definitive zone (, ). By the end and begins to produce cortisol under the regulation of
of gestation, the human fetal adrenal is nearly the size ACTH (). By the th week of gestation, the fetal
of the kidney, ~% of which is fetal zone (). The zone becomes rudimentary, and the definitive and
fetal zone has large steroidogenic cells that express the transitional zones become the zona glomerulosa and
CYPA enzyme, which has two activities—a- zona fasciculata, respectively.
hydroxylase and ,-lyase activities—that convert CYPB expression is apparent by embryonic day
pregnenolone into a-hydroxypregnenolone and  (E) in the rat adrenal gland and remains nearly
then DHEA. A substantial portion of the latter is constant until postpartum day . CYPB expression
sulfated to DHEA sulfate (DHEA-S), which in the is also observed at this time in single cells or small cell
placenta is converted to estrogens necessary for the clusters distributed throughout the adrenal ().
maintenance of a normal pregnancy. The smaller CYPB-positive cells scattered throughout the
definitive zone that becomes obvious at around the cortex decrease at E, and by E to E. there is an
eighth week of gestation between the capsule and the increase in the number of CYPB immunoreactive
fetal zone is composed of densely packed basophilic cells adjacent to the capsule, with only a few remaining
cells expressing SF. Definitive zone cells are lipid- in the area stained by the CYPB antibody. At E

1032 Seccia et al Biology of Zona Glomerulosa and APA Endocrine Reviews, December 2018, 39(6):1029–1056
REVIEW

there is a clear distinction between outer and inner typically mast cells, are present in the subcapsular area
cortex, with a small region in between where neither of the adrenal close to cells expressing mRNA for the
enzyme is expressed in the rat embryological adrenal HSDB and the CYPB enzymes, which appear by
cortex. The latter has been called the undifferentiated  and  weeks, respectively (). Tryptophan hy-
zone, where progenitor stem cells are located (, , droxylase and serotonin receptor type  expression
). Measurable aldosterone and corticosterone pro- occurred in the developing cortex by week , became
duction by rat fetal adrenals begins at about E (). limited to the definitive zone by week , and peaked
In the human immunoreactivity for steroidogenic between weeks  and , coinciding with the surge in
enzymes including steroidogenic acute regulatory CYPB expression (). This finding suggests a

Downloaded from https://academic.oup.com/edrv/article-abstract/39/6/1029/5051322 by Research 4 Life user on 22 November 2018


(StAR), CYPA, CYPA, P oxidoreductase, paracrine role for mast cell–derived serotonin in al-
and cytochrome b were detected in the fetal and dosterone secretion in the fetus, as has been dem-
transitional zones between  and  weeks’ gestation. onstrated in the adult zona glomerulosa ().
The HSDB enzyme necessary for the synthesis of The development of adrenal cortex zonation has
progesterone from pregnenolone was not detected also been addressed via cell fate mapping and gene
before the nd week of gestation and was detected deletion studies using a variety of techniques, in-
after the rd week only in the transitional and de- cluding specific zona glomerulosa Cre expression (,
finitive zones (). CYPA and DHEA sulfo- , ). Two models were classically proposed to
transferase were detected in the transitional and explain postnatal adrenal zonation: the zonal model of
definitive zones throughout gestation. lineage development and centripetal migration of cells
Immunohistochemical and steroid measurements from the subcapsular region. According to the zonal
show that the human adrenal produces DHEA in the model of lineage development, each zone develops and
transitional and fetal zone, but not in the definitive is maintained independently by progenitor cells em-
zone, as early as  weeks’ gestation (). Cortisol is bedded in the zone (). In the centripetal model,
produced in the transitional zone after the rd week undifferentiated progenitor cells in the capsule or
of gestation (). Mast cells and their secretory subscapular region differentiate into zona glomerulosa
products appear to play a role in the maturation of cells capable of producing aldosterone when stimu-
zona glomerulosa cells and their synthesis of aldoste- lated. These cells migrate centripetally and undergo
rone (). Studies performed between the th and lineage conversion to cortisol- or corticosterone-
st weeks of gestation with quantitative reverse producing cells, depending on the species, and even-
transcription PCR and immunohistochemistry dem- tually undergo apoptosis in the innermost part of the
onstrated that by the th week tryptase-positive cells, cortex, next to the adrenal medulla (). Lineage tracing

Figure 2. Ontogeny of the human adrenal and time during gestation at which steroidogenic genes are first expressed and steroids
synthesized. CYB5R3, cytochrome b5 reductase; HSD3B, 3b-hydroxysteroid dehydrogenase; SULT1A, sulfotransferase. [© 2018 Illustration
Presentation ENDOCRINE SOCIETY]

doi: 10.1210/er.2018-00060 https://academic.oup.com/edrv 1033


REVIEW

using Shh and Gli-expressing progenitor cells to adrenalectomy and transplantation, whereas trans-
map the fate of cells in the undifferentiated zone planted zona fasciculata cells only restore corticoste-
demonstrated radial stripes of cells that seemed to rone production (). This finding suggests that zona
migrate from the zona glomerulosa into the zona glomerulosa cells comprise progenitor cells that can
fasciculata, thus supporting the centripetal migration give rise to cells with the function of either zone,
model (). Cell fate mapping and gene deletion whereas zona fasciculata cells are able only to pro-
studies using zona glomerulosa–specific Cre expression liferate and maintain their differentiated function,
were used to test this model and localize progenitors of again lending support to the centripetal model. Results
zona glomerulosa and fasciculata cells (, ). In a of cell fate mapping and gene deletion studies (, )

Downloaded from https://academic.oup.com/edrv/article-abstract/39/6/1029/5051322 by Research 4 Life user on 22 November 2018


Cypb-Cre mouse model, Cypb-expressing cells suggest one of two possibilities. The most likely is
were permanently marked with green fluorescent that a single cell line developed from the adrenogo-
protein (GFP). Lineage tracing showed that the entire nadal primordium to generate all zones of the adrenal
zona glomerulosa and the majority of the zona fas- cortex. Temporal expression of transcriptional factors
ciculata were labeled within  months, indicating that produces the development of the fetal, definitive, and
all or most cortical cells descend from the Cypb+ transitional zones from these cells, and some of the
cell lineage during normal tissue replacement (). progenitor cells of the zona glomerulosa that differ-
Costaining with Sf and GFP also shows an exten- entiate into zona fasciculata cells retain the ability to
sive overlap between the GFP-labeled population and regenerate zona fasciculata cells but cannot rediffer-
the cells of the steroidogenic cortex (). Mice entiate into zona glomerulosa cells. However, cell fate
undergoing Cypb-Cre mediated Sf deletion, mapping techniques and gene deletion studies (, ,
which can no longer give rise to lineage-marked zona ) do not exclude the possibility of development of a
fasciculata cells, still have an entirely normal zona second cell line very early in differentiation that
fasciculata. This finding suggests that Cypb-Cre– generates only zona fasciculata cells, because zona
mediated Sf deletion occurs late enough to allow glomerulosa deletion of SF prevents lineage conver-
some progenitor cells to escape deletion of the Sf to sion of zona glomerulosa into zona fasciculata, but the
differentiate into fasciculata cells (, ). Signaling zona fasciculata develops normally from the other
molecules involved in adrenal cell renewal and zo- potential cell line. An unusual clinical case supports
nation are expressed from E. onward in mesenchymal this second developmental hypothesis: superselective
cells surrounding the developing adrenal. Members of adrenal vein sampling in a patient with bilateral
the R-spondin gene family, Rspo and Rspo, signaling hyperaldosteronism demonstrated an area of the ad-
molecules that bind leucine-rich repeat–containing renal with much lower production of aldosterone that
receptors and modulate the b-catenin pathway, are was spared during subtotal bilateral adrenalectomy.
maintained throughout development and adulthood Study of the excised tissue revealed that the both
(). b-catenin is highly expressed in the zona glo- adrenals harbored the same somatic KCNJ mutation
merulosa, and Rspo is expressed throughout the (GR) throughout the cortex, with one area of much
capsule. Its role in adrenal development is supported lower representation of the mutation. This finding
by the finding that tamoxifen-induced deletion of suggested that the mutation occurred early in the
Rspo at E. resulted in smaller adrenals (). Two development of the zona glomerulosa cell line (which
different pools of b-catenin exist: a cytoskeletal pool, followed the lineal conversion to zona fasciculata) and
which controls interaction with adherens junctions, that a second cell line lacking the mutation was also
and a cytosolic pool, which participates in canonical present (). This intriguing hypothesis clearly de-
wingless-related integration site (Wnt) signaling and serves further research.
acts as a coactivator of the TCF/LEF transcription The normal human neonate has a transient
factors (). Activated b-catenin is expressed in the physiological renal resistance to aldosterone and
adrenogonadal primordium, adrenal primordium, higher levels of plasma aldosterone than its mother
and subcapsular cells. Deficiency of Wnt alters (, ), associated with a low renal mineralocorticoid
adrenal function with a decrease in zona glomer- receptor expression (). Low aldosterone secretion in
ulosa function and very low aldosterone levels (). very preterm infants compounds the problem of low
Constitutive activation of b-catenin in the mouse receptor expression in the kidney (). Aldosterone
zona fasciculata causes increased Cypb expres- secretion rates per body surface area are higher in
sion in this zone with hyperaldosteronism and younger infants than in older infants, who are similar
development of adrenal tumors (, ); further- to adults (, ).
more, activated b-catenin is expressed in APAs and The fundamental enzyme for aldosterone synthesis
adrenal carcinomas (, ). is aldosterone synthase, or the CYPB enzyme. Its
Transplantation of rat zona glomerulosa and zona expression identifies cells that synthesize aldosterone,
fasciculata cells isolated by Percoll gradient indicate but its expression is regulated by aldosterone secre-
that zona glomerulosa cells completely restore pro- tagogues, so not all zona glomerulosa cells express
duction of aldosterone and corticosterone  days after CYPB. Immunohistochemistry of the adrenal of a

1034 Seccia et al Biology of Zona Glomerulosa and APA Endocrine Reviews, December 2018, 39(6):1029–1056
REVIEW

very young child shows that cells expressing CYPB movement, thus increasing cytosolic Ca+ concentra-
form a continuous band within the zona glomerulosa. tions, leading to activation of CaMK II complexes (,
With increasing age, expression of CYPB becomes ). CaMK II complexes mediate the transcription of
less continuous, so in the adult human CYPB- nuclear receptor–related  protein (also known as
expressing cells are scattered throughout the sub- NRA), activating transcription factors  and , and
capsular cortex among typical zona glomerulosa cells CREB, which initiate transcription of CYPB. The
that do not express the enzyme. These CYPB- major role for Ca+ in the regulation of aldosterone
expressing cells in some cases may be difficult to find biosynthesis is unambiguously supported by the blunted
(, , , ) except as clusters that extend from the Ang II–induced aldosterone secretion after inhibition of
Ca+ influx with Ca+ channel antagonists.

Downloaded from https://academic.oup.com/edrv/article-abstract/39/6/1029/5051322 by Research 4 Life user on 22 November 2018


subcapsular area and penetrate into the outer zona
fasciculata. The terms aldosterone-producing cell clusters Chronic Ang II stimulation maintains aldosterone
(APCCs) (), aldosterone-producing foci, and mega foci synthesis not only through increased CYPB
(, ), or just aldosterone-producing foci (, ), have transcription but also by increasing cholesterol uptake
been coined to identify these clusters; we will use APCCs by glomerulosa cells from high-, low- and very low-
herein. The number of APCCs increases with age (). density lipoproteins and increasing StAR protein,
Renin activity decreases in older adults, with no con- necessary for cholesterol transport into the mito-
comitant decline in plasma or urinary aldosterone (), chondria (, ). Activation of the renin-angiotensin-
suggesting an age-related autonomous aldosteronism aldosterone system by low Na+ intake induces expression
that might be related to the higher number and ab- of CYPB without affecting CYPB, probably be-
normal aldosterone physiology of APCCs. cause of the greater expression of AT receptors in zona
glomerulosa than in zona fasciculata cells (, ).
Chronic (- to -day) Ang II administration in rats was
Functional Regulation of also found to increase expression of Ang II receptors
Aldosterone Production subtypes A and B, an effect that was blocked by the
AT receptor antagonist losartan, suggesting that during
Under physiological conditions aldosterone secretion chronic stimulation Ang II increases aldosterone pro-
is regulated mainly by Ang II and K+, and to a lesser duction also by upregulating its own receptors (). As “The two-pore domain
extent by other secretagogues such as ACTH, endo- might be expected from its trophic effects in other cell channels TASK-1 and TASK-2
thelin  (ET-), estrogens, and urotensin II. Aldo- types, chronic Ang II stimulation also increases zona are highly expressed in the
sterone production can be upregulated acutely (within glomerulosa cell size (). normal human adrenal cortex
minutes) through increased expression and phos- and generate background K1
currents.”
phorylation of the StAR protein or chronically (over Potassium
hours to days) by augmented expression of the ste- Extracellular K+ is a potent regulator of aldosterone
roidogenic enzymes, mostly CYPB. synthesis. K+ channels, mostly the leak K+ channels
TASK-, TASK-, and TASK- (coded by Kcnk,
Ang II KcnK, and Kcnk genes), the TWIK-related potas-
Ang II is a potent activator of Ca+ influx, which sium channel , and the G protein inward rectifying
regulates all biosynthetic steps leading to aldosterone potassium channel Kir. (KCNJ), maintain adre-
secretion (). Upon binding to the angiotensin II nocortical cells hyperpolarized under resting condi-
type  (AT) receptor subtype in cells of zona glo- tions. The two-pore domain channels TASK- and
merulosa, Ang II activates phosphoinositide-specific TASK- are highly expressed in the normal human
phospholipase C, which hydrolyzes phosphatidyli- adrenal cortex and generate background K+ currents,
nositol ,-bisphosphate, which generates the sec- blunting aldosterone production. Inhibition of TASK
ondary messengers inositol ,,-trisphosphate (IP) channels by Ang II results in depolarization, and mice
and diacylglycerol (). IP triggers aldosterone secre- with deletions of TASK-, TASK-, or both genes have
tion by eliciting a transient increase in cytosolic calcium hyperaldosteronism (–). Increases in extracel-
(Ca+) concentration and activating Ca+/calmodulin- lular K+ depolarize the plasma membrane and activate
dependent protein kinases (CaMKs), whereas diac- the voltage-dependent calcium channels, leading to
ylglycerol stimulates protein kinase C, which regulates Ca+ influx and the signaling mechanisms described
StAR levels via cAMP response element binding above. Inhibition of Ca+ influx abolishes K+-stimulated
(CREB) and activator protein- (). Thus, activation aldosterone secretion. A role of K+ in zona glomerulosa
of the AT receptor stimulates both the early and late cell growth is also suggested by the increased thickness
regulatory steps of aldosterone synthesis, which in- of zona glomerulosa found in rats with a chronically high
clude phosphorylation of StAR and expression of the K+ intake.
steroidogenic enzymes, respectively.
Ang II regulates both Ca+ influx into the zona ACTH
glomerulosa cell via voltage-transient T- and long- ACTH binds to the transmembrane receptor mela-
lasting L-type Ca+ channels and intracytoplasmic Ca+ nocortin receptor  in the cells of zona glomerulosa and

doi: 10.1210/er.2018-00060 https://academic.oup.com/edrv 1035


REVIEW

zona fasciculata and exerts its downstream effects by II on an equimolar basis, and synergizes with Ang II
activating adenylate cyclase, thus generating cAMP, and ACTH as an aldosterone secretagogue on zona
which in turn activates protein kinase A (PKA). PKA glomerulosa cells (, ). ET- acts via ETB in rat
phosphorylates and activates StAR to increase cho- and via both ETA and ETB in human zona glomerulosa
lesterol delivery to the inner mitochondrial membrane cells by downstream activation of phospholipase C and
and activates members of the CREB family tran- protein kinase C pathways, rising intracellular Ca+,
scription factors. ACTH also promotes Ca+ influx and extracellular signal-regulated kinase/ribosomal S
through PKA-mediated phosphorylation of L-type kinase/CREB phosphorylation activation (, ).
calcium channels, further enhancing aldosterone ET-(-), which is formed in the adrenal cortex
by a chymase acting at the Tyr-Gly, acts through

Downloaded from https://academic.oup.com/edrv/article-abstract/39/6/1029/5051322 by Research 4 Life user on 22 November 2018


secretion.
Acute ACTH administration stimulates aldoste- ETA receptors to stimulate aldosterone secretion. ET-
rone secretion transiently, followed by decreased al- (-) is a weaker aldosterone secretagogue than ET-
dosterone synthesis within few days. The escape (-) in dispersed human adrenocortical cells, but it
from ACTH stimulation is restricted to aldosterone, has a much stronger effect on DNA synthesis and cell
because cortisol synthesis continues during chronic proliferation (). Therefore, it has been suggested
ACTH stimulation. Putative mechanisms for the loss that in the human adrenal cortex, depending on local
of aldosterone response to ACTH include melano- needs, the alternative cleavage of bigET- to ET-
cortin receptor  desensitization and internalization (-) by endothelin-converting enzyme- , or to
through a PKA-dependent mechanism; increased ET-(-) by chymase, may lead to the production of
a-hydroxylase (Pc) and b-hydroxylase either peptide, the former mainly stimulating ste-
(Pc) activity, resulting in enhanced production roidogenesis via both ETA and ETB receptors, the latter
of DOC and precursor steroids; blunting of the late mainly exerting growth-promoting effects via ETA
(CYPB-mediated) step of aldosterone synthesis; receptors (). How this process is regulated deserves
transformation of zona glomerulosa cells into zona further research.
fasciculata–like cells featuring round mitochon-
dria with ovoid cristae; suppression of the renin- Serotonin
angiotensin-aldosterone system; desensitization of Serotonin released by perivascular mast cells in the
zona glomerulosa cells to Ang II via downregulation of subcapsular region of the adrenal cortex activates the
the AT receptor; and a rapid increase in zona fas- -hydroxytryptamine receptor  (-HT) in zona
ciculata thickness alongside a decrease in zona glo- glomerulosa cells, thus stimulating aldosterone pro-
merulosa width [for a review see Gallo-Paynet ()]. duction. Some APAs overexpress -HT receptors,
Moreover, ACTH stimulation leads to an increase in and others have been described as having a high
-oxocortisol formation. The activities of CYPB density of infiltrating mast cells (–), suggesting
and CYP are necessary for the synthesis of that overexpression of -HT receptors or over-
-oxocortisol, either by coexpression in the same cells production of serotonin contributes to the aldoste-
or by a paracrine mechanism in which cortisol, se- ronism in subsets of APA.
creted by cells expressing CYPB and CYP, is
taken up by cells expressing CYPB (–). The Urotensin II
underlying mechanism is not clear because CYPB The potent vasoactive peptide urotensin II is ubiq-
and CYP are not expressed in the same cells in the uitously expressed in the vascular tissue and is in-
normal adrenal but are coexpressed in many adeno- volved in blood pressure regulation. It is also expressed
mas. A paracrine mechanism is also doubtful because in both the medulla and cortex of normal human
blood flow is centripetal in the adrenal, moving adrenal glands. Pheochromocytomas express higher
substrate from the zona fasciculata away from urotensin II levels, whereas APAs have lower levels
CYPB, and adrenal cells express a high level of the than the normal adrenal gland. In contrast, the op-
p-glycoprotein that pumps out polar steroids such as posite was found for the urotensin II receptor,
cortisol (). suggesting a urotensin II–mediated paracrine in-
teraction between the cortex and the medulla ().
ET-1 Infusion of urotensin II in normal rats for  week
ET- is a paracrine hormone that activates ETA and potently increased both CYPB expression and
ETB receptors mediating contraction, cell proliferation, plasma aldosterone levels () through activation of a
and hypertrophy in vascular smooth muscle, and ETB specific urotensin receptor, and these effects were
receptors, which stimulates prostacyclin and nitric abrogated by the urotensin receptor antagonist pal-
oxide production in endothelial cells, causing vaso- osuran (). Because the mild increase in CYPB in
relaxation and inhibition of sodium transport in renal these adrenals, possibly due to stress, was unaffected by
tubules. In freshly isolated human adrenocortical cells palosuran, the secretagogue effect of urotensin II on
ET- acts as an aldosterone secretagogue through both aldosterone seems to be specific and thus can be
ETA and ETB receptors, with a potency similar to Ang physiologically relevant. Of further note, rats chronically

1036 Seccia et al Biology of Zona Glomerulosa and APA Endocrine Reviews, December 2018, 39(6):1029–1056
REVIEW

infused with urotensin II and exposed to different Na+ secretion, whereas activation of D stimulates aldo-
intakes were recently described to show an increase not sterone secretion and thus counteracts D-mediated
only of aldosterone but also of renin release, suggesting inhibition (). A microarray comparison of genes
that under conditions of salt and volume overload the from normal human zona fasciculata and zona glo-
peptide can mediate the inappropriately high secretion merulosa and from zona fasciculata–like and zona
of both hormones (). glomerulosa–like APAs led researchers to identify the
expression of the neurofilament medium polypeptide
17b-Estradiol (NEFM) in normal zona glomerulosa cells, small zona
In human zona glomerulosa cells, estradiol (E) can act glomerulosa–like APAs, and the human adrenocortical

Downloaded from https://academic.oup.com/edrv/article-abstract/39/6/1029/5051322 by Research 4 Life user on 22 November 2018


as secretagogue or antisecretagogue, depending on the cell HR, but not in normal zona fasciculata and in
estrogen receptor subtype activated (). In the large APAs, including those that express KCNJ
normal human adult adrenal cortex and APA tissues, mutations (). Silencing of NEFM in HR cells
ERb is far more abundantly expressed than ERa and increased basal aldosterone production and cell
its variant ERa, whereas the G protein–coupled proliferation, as well as aldosterone production in
receptor- (GPER-) is highly expressed in the normal response to D agonist fenoldopam; moreover,
human adrenocortical zona glomerulosa and faintly expression of a mutant KCNJ gene in these cells
detectable in zonae fasciculata and reticularis (). E decreased NEFM expression and raised basal al-
tonically inhibits aldosterone synthesis via ERb, as dosterone production, suggesting that NEFM is a
demonstrated by the increased aldosterone synthesis negative modulator of aldosterone acting via the D
after ERb blockade or silencing, whereas the secre- receptor ().
tagogue effect of E on aldosterone involves GPER-
and protein kinase A and cAMP signaling () and is Atrial natriuretic peptide
unmasked when ERb is blocked. In APAs, GPER- is The blunting of aldosterone secretion was described as
the predominant ER subtype, which suggests that it one of the multiple actions of the natriuretic peptide
can be relevant for the pathophysiology of aldoste- (). In line with this effect, binding studies have
ronism (, ). Given the broad use of selective revealed a single class of high-affinity saturable sites in
estrogen receptor modulators, some of which act as HR cell membranes, which were thereafter char-
“A decrease in serum ionized
GPER- modulators, in women with hormone- acterized as NPRA receptors coupled to the particulate
Ca21 levels is the main
dependent forms of cancer, better knowledge of the guanylate cyclase (). These receptors antagonize stimulus for PTH secretion by
effect of selective estrogen receptor modulator treat- Ang II–stimulated aldosterone secretion in HR the chief cells of the
ment on aldosterone secretion would be an important cells and murine adrenals (, ) and also inhibit parathyroid gland.”
goal. However, this is an area where investigative ef- ACTH-stimulated aldosterone secretion via cyclic
forts are clearly necessary. guanosine monophosphate–dependent activation of
phosphodiesterase  and hydrolysis of cAMP in mice
Dopamine ().
Dopamine is thought to tonically inhibit aldosterone
secretion because the selective D receptor antago- Parathyroid hormone
nist metoclopramide induces aldosterone release in Although other factors, such as calcitriol, phosphate,
humans (, ). Dopamine blunts Ca+ influx and magnesium, and the FGF/klotho system, intervene
protein kinase C phosphorylation (), suggesting in a complex manner, a decrease in serum ionized
that its inhibitory effect involves the early steps of Ca+ levels is the main stimulus for PTH secretion by
steroidogenesis. However, dopamine or dopamine the chief cells of the parathyroid gland. PTH is a single-
agonists, such as bromocriptine, did not modify basal chain –amino acid peptide sharing sequence ho-
plasma aldosterone levels (), indicating maximal mology with ACTH (). Like ACTH, PTH also
tonic dopaminergic inhibition of endogenous aldo- activates cellular adenylate cyclase/cAMP-dependent
sterone production. protein kinase, phospholipase C/protein kinase C–
Dopamine, Ang II, and salt intake interact in a dependent, and cAMP-dependent signaling cascades.
complex way in regulating aldosterone secretion. PTH also promotes cytosolic Ca+ entry into the
Dopamine inhibits Ang II-stimulated aldosterone mitochondrial matrix, an essential step for triggering
secretion in Na+-depleted subjects (), but it is steroidogenesis, and particularly for its late step in-
ineffective in preventing basal or ACTH- or Ang II- volving CYPB, an enzyme that is Ca+-activated
stimulated aldosterone production in Na+-repleted and cAMP-activated, as discussed in the next section.
subjects (, ). Of the five known dopamine re- A bidirectional link between aldosterone and PTH
ceptor subtypes, four (D, D, D, and D) are secretion in humans has been proposed based on the
expressed in the normal human adrenal gland, sug- findings that PTH (and also the cancer-derived PTH-
gesting their role in the modulation of steroidogenesis related peptide) stimulates aldosterone secretion in a
(). Activation of the D subtype, the predomi- concentration-dependent fashion; APA cells express
nant D-like receptor, tonically inhibits aldosterone the type  PTH receptor; parathyroid cells express the

doi: 10.1210/er.2018-00060 https://academic.oup.com/edrv 1037


REVIEW

mineralocorticoid receptor; and patients with PA permissive window occurs at lower voltages. Ac-
(), and particularly those with the florid form of PA cordingly, they activate upon depolarization from
usually due to APA, have elevated serum PTH levels, resting hyperpolarized potentials, allowing a surge of
which are corrected by adrenalectomy (). Thus, the Ca+ influx at the beginning of an action potential
enhanced cardiovascular damage observed in both (when the electrochemical gradient is highly favorable
primary hyperparathyroidism and PA can derive from for cation entry). Because only a small fraction of channels
an interplay between PTH and aldosterone (, ). open upon modest membrane depolarizations, the
resultant current per unit time is small compared
Calcium ions with the maximal current that can be elicited by a
Ca+ affects adrenocortical steroidogenesis in multiple

Downloaded from https://academic.oup.com/edrv/article-abstract/39/6/1029/5051322 by Research 4 Life user on 22 November 2018


strong depolarization or a putative action potential
ways: by increasing the activity of cholesterol ester when more channels open. However, because channel
hydrolase, the enzyme that de-esterifies cholesterol, activation can be sustained for minutes (because of a
allowing its release from cytoplasmic storage and use lack of complete inactivation at hyperpolarized volt-
in aldosterone synthesis; by promoting cytoskeletal ages), Ca+ accumulation within the cell during this
delivery of cholesterol to the outer mitochondrial time period can still be large. Therefore, these channels
membrane and the transcription and translation of are well suited for responding to the small membrane
StAR, which increases transfer to the inner membrane; depolarizations from rest induced by physiological zona
by augmenting the mitochondrial oxidative metab- glomerulosa cell agonists and conveying Ca+ into the
olism and Ca+-dependent formation of reduced cell at the right place for the control of steroido-
nicotinamide adenine dinucleotide phosphate, a co- genesis. This explains why physiological concentra-
factor necessary for the activity of P cytochromes tions of Ang II and K+ may preferentially activate
including CYPA and CYPB; and by increasing Cav. and not Cav. channels, to allow Ca+ influx
the binding of Ca+ to calmodulin-activated Ca+/ into the cytoplasm.
calmodulin-dependent kinases, which target steroido- In contrast, the high-threshold Cav. (L-type)
genic factors, including CYPB, which catalyzes the Ca+ channels are normally activated by much
conversion of -DOC to aldosterone. stronger cell depolarization from rest; thus, they
Under resting conditions Ca+ concentration is probably play a minor role in the tonic regulation of
,-fold lower [about  to  nM ()] in the aldosterone secretion. Accordingly, the dihydropyr-
cytosol of zona glomerulosa cells than in the extra- idine (L-type) calcium antagonists are very weak in-
cellular space ( to  mM). Thus, upon depolarization- hibitors of aldosterone secretion, both in vivo and in
induced opening of voltage-gated Ca+ channels there vitro, supporting the view that the two types of
is a large concentration gradient for Ca+ influx (). channels may serve different functions in the regu-
Sustained Ca+ entry into zona glomerulosa cells is lation of steroidogenesis.
possible because voltage-operated calcium channels Ca+ cytosol concentration in the zona glomerulosa
have a permissive voltage window through which they cells, as well as ACTH-induced cAMP signaling and
conduct steady-state current (). PKA activation, are key for induction and activation of
Voltage-dependent activation and inactivation StAR (). They are also crucial for activation of
curves have been constructed for calcium channels in calmodulin-dependent kinases I and IV, which are
zona glomerulosa cells. Curves for activation predict crucial for StAR transcription ().
the proportion of channels that open at each voltage, Cytosolic Ca+ concentration is also controlled by
whereas those for inactivation predict the proportion the mitochondria, which represent ~% of zona
of channels that are available to open that have not glomerulosa cell cytoplasm volume. The high density of
inactivated. When considered together, these re- these organelles allows them to act as Ca+ buffers:
lationships define a permissive voltage window in they accumulate Ca+ during the rising phase of cy-
which there is steady-state opening of calcium tosolic Ca+ transients and release Ca+ during the
channels, enabling a sustained flux of calcium into decaying phase (). Even small increases in mito-
zona glomerulosa cells [Fig. (a)]. chondrial Ca+ levels have measurable effects, in-
Two types of voltage-gated Ca+ channels have cluding reduction of pyridine nucleotide nicotinamide
been described in mammalian zona glomerulosa cells: adenosine dinucleotide phosphate, enhanced steroido-
low-voltage activated T-type (Cav.), and high- genesis, particularly aldosterone biosynthesis, and gen-
voltage activated L-type (Cav.), which differ in the eration of ATP essential for maintaining cell homeostasis
voltage dependency of their opening and inactivation (). Reduction of nicotinamide adenine dinucleotide
properties. Knowing their features and permissive phosphate produced in the mitochondria is necessary for
window is key for understanding the impact of the ion a and -hydroxylations and subsequent scission of
channel mutations that have been identified in PA. cholesterol and also for b-hydroxylation of -DOC
The low-threshold Cav. channels activate and followed by -hydroxylation and -methyl oxidation
inactivate at lower voltages than high-threshold L-type of corticosterone, two crucial steps in aldosterone
Ca+ channels (Cav.), which means that their biosynthesis ().

1038 Seccia et al Biology of Zona Glomerulosa and APA Endocrine Reviews, December 2018, 39(6):1029–1056
REVIEW

Figure 3. Activation of wild-type and mutated channels. (a) Under physiological conditions the threshold of voltage-operated T-type
calcium channels Cav3.2 (blue) is lower than that of L-type Cav1.3 channels (green), leading to activation (and deactivation) at lower
voltages (permissive window of voltage), even though the voltage dependence [i.e., the fractional opening per unit increase in voltage
(slope factor)] does not differ greatly between channels. The pink rectangle indicates the range of resting membrane potentials of
normal glomerulosa cells. The y-axis indicates the amplitude of currents, expressed as normalized steady-state current. (b) Both somatic
and germline KCNJ5 mutations cause loss of ion selectivity with ensuing increased Na+ influx and therefore a shift of the resting
potential toward less polarized voltages (striped rectangle) that, in turn, causes opening of the voltage-dependent T-type Ca2+ channels
[see Choi et al. (19)]. (c) Germline CLCN2 mutations, by causing efflux of chloride ion, also determine a shift of the resting potential
toward less polarized voltages [see Scholl et al. (23) and Fernandes-Rosa et al. (24)]. (d) A similar shift of the resting potential toward less
polarized voltages can occur in cells with mutations of ATP1A1 that cause loss of function or with mutations of ATP2B3 associated with
impaired clearance of cytoplasmic Ca2+ ions [see Beuschlein et al. (138)]. (e) Ile770Met CACNA1D mutation (brown dotted line) causes

Downloaded from https://academic.oup.com/edrv/article-abstract/39/6/1029/5051322 by Research 4 Life user on 22 November 2018


a curve shift toward the left, leading to activation of the mutated channel at lower voltages than those needed to activate the wild-type
Cav1.3 channel (green) [see Scholl et al. (139)]. (f) Mutations of CACNA, as germline CACNA1H M1549V mutation, cause increased
constitutive Ca2+ influx at potentials close to the resting potential of zona glomerulosa cells (more rapid activation) and delayed
inactivation, with a resulting larger permissive voltage window [see Scholl et al. (140)]. For details on cell type and species in which these
curves were generated, see the original references. [© 2018 Illustration Presentation ENDOCRINE SOCIETY]

doi: 10.1210/er.2018-00060 https://academic.oup.com/edrv 1039


REVIEW

Biology of APA Formation ATPA () and ATPB (), cause a loss of
function. Overall, they are thought to promote
The multiple hypothetical mechanisms that may CYPB expression and aldosterone biosynthesis via
contribute to the development of APAs are depicted in an increase in intracellular Ca+ levels, albeit through
Fig. . For the sake of clarity, these will be itemized different mechanisms ().
().
Somatic mutations
Mutations in ion channels Very soon after PA-related KCNJ gene mutations
Somatic or germline mutations in the genes encoding were first described (), several others were identified
in APAs from patients throughout the world (, ,

Downloaded from https://academic.oup.com/edrv/article-abstract/39/6/1029/5051322 by Research 4 Life user on 22 November 2018


ion channels and proteins that regulate the membrane
potential of zona glomerulosa cells are common in ) (Table ). The KCNJ gene encodes the G
APAs, suggesting that they play a key role in the protein–activated inward rectifier potassium channel
biology of these tumors. Some mutations, such as (GIRK, also known as Kir .) that, under normal
KCNJ () and CACNAD () and CCl (, ) conditions, allows the selective efflux of K+ through the
lead to a gain of function, whereas others, such as channel pore, thereby maintaining zona glomerulosa

Figure 4. Mechanisms mostly deemed to contribute to the development of APAs. (a) Somatic mutations in the genes KCNJ5, ATP1A1,
and CLCN2 coding for Kir3.4, Na+/K+ ATPase, and ClC-2 channels cause membrane depolarization of zona glomerulosa, with ensuing
increased influx of Ca2+ into the cells, whereas mutations in CACNA and ATP2B3 genes, which code for Cav1.3 and plasma membrane
calcium-transporting ATPase, directly cause an increase in intracellular Ca2+ levels. In both cases the final result is enhanced CYP11B2
expression and increased aldosterone production. For a list of mutations, see Table 1. (b) Wnts are secreted proteins that control
growth and stem cell renewal, acting via canonical and noncanonical Wnt pathways. The canonical Wnt signaling is activated by the
binding of Wnt to a serpentine Frizzled receptor (Fzd) and the coreceptor LRP5/6, which lead to recruitment of Dishevelled protein Dsh
and disassembly of the b-catenin destruction complex. b-catenin, free to move toward the nucleus, binds to the TCF/LEF1 proteins,
triggering transcription of genes involved in growth. When activated by the noncanonical Wnt pathway, Wnt first binds to Fzd and
recruits Dsh to form a complex that activates the Rho-associated kinase pathway or phospholipase C, finally releasing Ca2+ from
intracellular stores. The increased Ca2+ can drive cell growth or aldosterone synthesis. (c) Hypomethylation of the promoter region of
CYP11B2 activates gene transcription, thereby promoting aldosterone secretion. (d) Elevated levels of miRNA 23 (miR23) and miRNA 34
(miR34) can downregulate KCNJ5 gene expression, blunting TASK2 synthesis, and finally enhancing aldosterone production. [© 2018
Illustration Presentation ENDOCRINE SOCIETY]

1040 Seccia et al Biology of Zona Glomerulosa and APA Endocrine Reviews, December 2018, 39(6):1029–1056
REVIEW

Table 1. Somatic and Germline Mutations Causing Excess Aldosterone Production


Gene and Mutation Channel and Protein Transmission Effect Phenotype and Clinical Disorder

KCNJ5 GIRK4 (also known as Kir 3.4) Somatic ↑ Na conductance


+
More frequent in women and younger patients;
larger APA with zona fasciculata–like cells;
Cell membrane depolarization higher plasma aldosterone levels and ARR at
↑ Ca2+ influx into the cell diagnosis

ATP1A1 a1 Subunit of the Na+/K+ ATPase Somatic Cell membrane depolarization More common in older men; smaller APA with

Downloaded from https://academic.oup.com/edrv/article-abstract/39/6/1029/5051322 by Research 4 Life user on 22 November 2018


zona glomerulosa–like cells; higher plasma
↑ Ca 2+
influx into the cell aldosterone levels and lower K+ at diagnosis

CACNA1D a1 Subunit of the Cav1.3 voltage- Somatic ↑ Ca2+ influx activation More common in older men; smaller APA with
dependent L-type calcium zona glomerulosa–like cells

ATP2B3 Plasma membrane calcium- Somatic ↑ Ca2+ influx into the cell More common in older men; smaller APA with
transporting ATPase 3 zona glomerulosa–like cells; higher plasma
aldosterone levels and lower K+ at diagnosis

CTNNB1 b-Catenin Somatic Activation of canonical or More common in women and older patients;
noncanonical Wnt pathways larger APA

CYP11B2/CYP11B1 CYP11B2 Germline CYP11B2 under control of CYP11B1 Early and severe hypertension; higher plasma
chimeric promoter levels of 18-hydroxycortisol and 18-
oxocortisol; bilateral hyperplasia or adrenal
nodules

FH-I or GRA

Gene unknown in Unknown Germline ↑ Aldosterone production No specific phenotype; diagnosis is made after
chromosome 7p22 exclusion of other known familiar forms

FH-II

KCNJ5 GIRK4 (also known as Kir 3.4) Germline ↑ Na+ conductance Early and severe hypertension and hypokalemia;
higher levels of aldosterone, 18-
hydroxycortisol, and 18-oxocortisol; bilateral
hyperplasia

Cell membrane depolarization FH-III

↑ Ca2+ influx into the cell

CACNA1H ↑ Ca2+ influx activation Germline ↑ CYP11B2 expression Early onset of PA and hypertension,
developmental delay, and attention deficit

FH-IV
2
CLCN2 ClC-2 Germline ↑ Cl conductance Detected in young patients

Cell membrane depolarization

Abbreviations: ARR, aldosterone-to-renin ratio; FH, familial hyperaldosteronism; GRA, glucocorticoid-remediable aldosteronism.

cells hyperpolarized (, ). These mutations are fasciculata–like cells, higher expression of CYPB, and
located in exon  and cause amino acid changes near, lower expression of CYPB than zona glomerulosa–like
or within, a highly conserved TXGYGFR motif of the APAs that tend to be smaller (–). A differential
GIRK selectivity filter, which result in loss of ion gene expression profile between zona fasciculata–like
selectivity (). The ensuing increased Na+ influx and smaller zona glomerulosa–like APAs identified a
determines cell membrane depolarization, opening of consistent expression of NPNT in zona glomerulosa–
voltage-dependent T-type Ca+ channels [Fig. (b)], like APAs and its lack in zona fasciculata–like APAs
and activation of the Na+/Ca+ exchanger in the re- (). NPNT is a Wnt target gene that encodes the
verse mode (e.g., Na+ out and Ca+ in) (), thus extracellular matrix protein nephronectin (). Ac-
increasing cytosolic Ca+ levels and ultimately trig- cordingly, nephronectin was immunochemically dem-
gering aldosterone synthesis. onstrated in zona glomerulosa–like APAs and normal
Tumors with KCNJ mutations have been de- human zona glomerulosa but not in zona fasciculata–like
scribed to be larger, with a predominance of zona APAs and in the normal human zona fasciculata ().

doi: 10.1210/er.2018-00060 https://academic.oup.com/edrv 1041


REVIEW

Table 2. Prevalence of Mutations in K+ or Ca2+ Channels in Either APCC or APA


APCC

a Mutation Without Hypertension (93, 154) With Hypertensiona (155, 156) APA (155, 156)
Available data are from
hypertensive patients with PA. KCNJ5 0% 0%–8% 38%–63%

CACNA1D 23%–26% 0%–65% 9%

ATP1A1 3%–9% 0%–4% 5%

Downloaded from https://academic.oup.com/edrv/article-abstract/39/6/1029/5051322 by Research 4 Life user on 22 November 2018


ATP2B3 0%–5% 0%–4% 1%

CACNA1D + ATP2B3 0%–3% 0%° 0%

Both nephronectin and NEFM, as discussed earlier as of CYPB, as well as NRA and NRA genes
regards dopamine, are expressed in the normal zona encoding transcription factors NURR and NOR,
glomerulosa but not in zona fasciculata cells (, ). respectively (, ). Somatic mutations in ATPA,
This leads to two hypotheses to be tested. Might either ATPB, and CACNAD, but not in KCNJ so far,
or both factors be involved in adrenocortical cell have also been detected in APCCs from normal ad-
proliferation and replenishment? Does their absence renal glands (, ) (Table ).
allow a zona fasciculata–like phenotype? Although it is The effects of these mutations on cell proliferation
not yet clear why KCNJ mutations result in loss of remain unclear (). The KCNJ TA mutation
NEFM expression, it might at least partially explain the was associated with massive bilateral hyperplasia,
zona fasciculata–like phenotype of APA with KCNJ suggesting a growth-promoting effect (). In contrast
mutations (). with this contention, studies performed in the labo-
The CACNAD gene codes for the a subunit of ratory of some of us (C.E.G.-S. and E.P.G.-S.) showed
the Cav. voltage-dependent L-type calcium channel, that cells transfected with this mutant GIRK
which increases Ca+ influx in response to de- exhibited a lower proliferation rate than cells trans-
polarization (, ). The a-subunit is composed of fected with wild-type GIRK or even apoptosis (),
four repeated domains, each containing six trans- indicating that other mechanisms are involved in
membrane segments, and forms the channel pore. abnormal cell growth.
Mutations in hotspot areas of CACNAD cause a gain Of note, despite an explosion of research, the
of function of CaV. in zona glomerulosa cells due to factors leading to somatic mutations in APA are
opening of the Cav. channel at membrane potentials unknown. Because the rate of KCNJ mutations varies
more hyperpolarized than under physiological con- widely (), ranging from % in Europe () to %
ditions, thus increasing Ca+ influx and CYPB to % in Asia and Japan (, ), whereas mutations
expression () [Fig. (f)]. in ATPA, ATPB, and CACNAD seem to be
ATPA and ATPB are members of the P-type uncommon in the Asian cohorts, some of us (G.P.R.,
ATPase gene family, which encode the a subunit of the T.M.S.) have suggested that the occurrence of these
Na+/K+ ATPase and the plasma membrane calcium- mutations depends on ethnic or environmental factors,
transporting ATPase , respectively (). At rest, including salt intake and screening policies imple-
ATPA maintains the negative cell membrane potential mented in the different countries (). Moreover, there
by exchanging two extracellular K+ ions for three cy- are APAs associated with hyperplasia or smaller
toplasmic Na+ ions with ATP consumption. Somatic aldosterone-producing nodules in the adjacent zona
mutations in ATPA are found in ~% of APAs. They glomerulosa, and different somatic mutations have been
involve the transmembrane helices M and M of the found in the same adrenal gland (, , ). The
protein and cause loss of pump activity, reduced affinity hypothesis that one trigger may induce different mu-
for K+, and Na+ leak, leading to cell membrane de- tations has been suggested, although no solid evidence
polarization [Fig. (d)] and increased Ca+ influx and, for this contention has been provided to date.
thereby, increased aldosterone production (). It remains unknown how somatic mutations in-
Mutations of ATPB, found in .% of APAs, duce the transformation of normal adrenocortical cells
involve the M transmembrane helix, which is re- into APA cells. One hypothesis is that somatic mu-
sponsible for Ca+ transport and binding of ATP and tations induce formation of CYPB-expressing
phosphate (). They distort the Ca+ ion-binding APCCs from mutated zona glomerulosa cells, lead-
motifs, thus increasing intracellular Ca+ and priming ing to renin-independent aldosterone production and,
aldosterone production. eventually, to an APA. Lending support to this hy-
Transfection of different mutant channels into pothesis are the findings by Nishimoto et al. () that
adrenocortical cells resulted in increased transcription APCCs comprise mainly small subcapsular zona

1042 Seccia et al Biology of Zona Glomerulosa and APA Endocrine Reviews, December 2018, 39(6):1029–1056
REVIEW

glomerulosa–like cells surrounding large lipid-rich occurring even in the same codon were described to
zona fasciculata–like cells and that the transcriptome cause altogether different clinical phenotypes and
profiles of APCCs are similar to that of zona glo- responsiveness to drug treatment, thus leading to the
merulosa cells with high expression of CYPB, and division of FH-III into subtypes A and B. FH-IIIA
finally that some APCCs harbor cells with ATPA features stage III, drug-resistant hypertension neces-
and CACNAD mutations. However, how zona glo- sitating bilateral adrenalectomy. FH-IIIB results in a
merulosa cells are induced to form nests of cells (i.e., much milder clinical phenotype best treated with
APCCs) remains to be established. Moreover, it is antihypertensive agents () (Fig. ).
unknown why KCNJ mutations do not occur in

Downloaded from https://academic.oup.com/edrv/article-abstract/39/6/1029/5051322 by Research 4 Life user on 22 November 2018


APCCs. The hypothesis that APCCs with KCNJ FH type IV
mutations rapidly progress to APA, though conceiv- Germline mutations in CACNAH cause different
able (), remains to be proven. phenotypic presentations of PA [reviewed in ()]. The
T-type calcium channel blocker mibefradil reduces the
Germline mutations excess aldosterone production in transfected Cav.
Germline mutations cause ,% of PA cases and have Val cells, suggesting that inhibitors of this channel
been recently reviewed (, –), therefore, they could be the treatment of choice for patients with
will be examined only briefly herein (Table ). A CACNAH gain-of-function mutations (). Un-
classification proposed recently by one of us (G.P.R.), fortunately, mibefradil has been withdrawn from the
which considers not only the mutations but also the market because of increased risk of death in patients
clinical and imaging features and the response to drug concomitantly receiving other antiarrhythmic drugs,
treatment, will be followed (). mostly amiodarone.
The finding of somatic or germline mutations in K+
Familial hyperaldosteronism type I and Ca+ channels in a large percentage of APAs
(glucocorticoid-remediable aldosteronism) implicated these mutations in oncogenesis and sug-
Familial hyperaldosteronism (FH) type I is an auto- gested that they might be driver mutations conferring
somal dominant disease is caused by an unequal growth advantages to the neoplastic cells ().
crossing-over of the homologous genes coding for However, the heterogeneity of adrenal morphology
CYPB and b-hydroxylase (CYPB). This re- found in patients with PA with these germline mu-
sults in a chimeric gene comprising the CYPB tations argues against this theory. If these were driver
coding sequences under control of the CYPB mutations, they would be expected to invariably lead
promoter and thus controlled by ACTH (). This to neoplastic growth. In contrast, patients with FH-III
explains why in FH-I the clinical and biochemical germline KCNJ mutations exhibit adrenal pheno-
signs of PA can be antagonized by suppressing ACTH types ranging from normal adrenal to massive bilateral
with low-dose dexamethasone (). Measurements of
-hydroxycortisol and -oxocortisol levels suggest
the diagnosis, but it is confirmed with long PCR, a
more widely available technique ().

FH type II
Clinically heterogeneous, FH-II comprises non–
glucocorticoid-remediable forms of familial PA inherited
with an autosomal dominant pattern (–).
Although an association of a locus on the region
p has been found in some FH-II families (,
), the genetic basis remains unknown; the di-
agnosis of FH-II is made after exclusion of the other
Mendelian forms. The finding of new mutations in
families of patients previously classified as having FH-II
separates them from this group. Recently two groups
independently reported a family with the chloride
channel CLCN (, ). Figure 5. Development of APA or multinodular cortical hyperplasia from zona glomerulosa. In FH-I
the hybrid chimeric gene comprising the CYP11B2 coding sequences under control of the CYP11B1
FH type III (FH-IIIA and FH-IIIB) promoter induces hyperproduction of aldosterone controlled by ACTH. Growth and proliferation
of stem cells cause multinodular cortical hyperplasia or multinodular lesions with tumors. In the
Different mutations in the KCNJ gene located in or
only FH-I pedigree [see Jeunemaitre et al. (163) and Pascoe et al. (172)], two patients had a tumor but
near the selectivity filter cause FH-III. The molecular did not lateralize on adrenal venous sampling (AVS), suggesting that these tumors were not APAs. In
mechanisms derived from these mutations resemble FH-III the excess production of aldosterone is caused by mutations in the KCNJ5 gene, which are located
those found in somatic KCNJ mutations found in or near the selectivity filter. Whether growth and proliferation of stem cells cause development of
in APAs (, , , ); however, mutations APAs remains to be investigated. [© 2018 Illustration Presentation ENDOCRINE SOCIETY]

doi: 10.1210/er.2018-00060 https://academic.oup.com/edrv 1043


REVIEW

adrenal hyperplasia or nodules (, , ), and development of APAs. Although the underlying mech-
adrenals of patients with germline CACNAH mu- anisms remain unclear, one hypothesis is that Wnt ac-
tations were found to have an APA, hyperplasia, or tivation results from decreased expression of the secreted
even a normal phenotype (, ). Therefore, it negative Wnt regulator Frizzled-related protein II ().
seems reasonable to hypothesize that K+ and Ca+
channel mutations, though favoring inappropriate Hypomethylation of CYP11B2 and genes
aldosterone production, have little or no impact on the controlling CYP11B2 transcription
cell growth. Other abnormalities, such as those in- DNA methylation or demethylation by methyl-
volving the Wnt/b-catenin signaling pathway or transferase and demethyltransferase enzymes are key

Downloaded from https://academic.oup.com/edrv/article-abstract/39/6/1029/5051322 by Research 4 Life user on 22 November 2018


hypomethylation of CYPB, may be involved in mechanisms of DNA damage repair, cell survival, and,
triggering cell growth in the zona glomerulosa. when dysregulated, tumor progression (). When
Three gain-of-function missense mutations methylation affects the promoter region, it blunts gene
(GD, IM, and VL) in CACNAD were transcription and hormone secretion, whereas hypo-
found to be associated with extra-adrenal signs, such as methylation has the opposite effect (). By using
seizures, cerebral palsy, and other neurologic abnor- DNA methylation array analysis in  APAs, Yoshii
malities (PASNA, OMIM: ) (, ). The et al. () showed that seven of eight putative
mutations cause channel activation at membrane methylation sites of the CYPB gene, four of which
potentials close to the resting potential of the zona are located in the promoter region, were hypo-
glomerulosa cells, leading to an increased Ca+ influx methylated in APAs. However, methylation levels
and stimulation of aldosterone production [Fig. (d)]. showed no correlation with CYPB transcription
Very recently, six gain-of-function mutations have levels in APAs (), suggesting the contribution of
been identified in the CLCN gene via whole-exome other mechanisms to excess aldosterone production in
sequencing of germline DNA from young-onset hy- APAs. More recently, Purkinje cell protein  (PCP), a
pertension and PA diagnosed before  years of age calmodulin-binding factor that drives intracellular
(, ). CLCN gene encodes the voltage-gated ClC- calcium from the cytoplasm to the nucleus, has been
chloride channel, whose opening causes depolarization identified as one of the most hypomethylated genes in
of zona glomerulosa cells and enhanced expression of APAs (). The PCP gene not only is more highly
CYPB [Fig. (c)]. These mutant channels provide expressed in APAs than in the normal adrenal cortex,
higher probabilities of channel opening at the glo- but it also has a site in its promoter region that binds
merulosa resting potential and ensuing abnormally the transcription factor CCAAT/enhancer binding
high aldosterone production. protein b. Thus, it has been suggested that hypo-
methylation of PCP DNA in APAs allows increased
Wnt/b-catenin signaling pathway PCP expression, which in turn leads to CYPB
Wnts are secreted proteins that control growth and stem upregulation (). Of note, APAs with underlying
cell renewal through activation of highly conserved KCNJ mutations have not been found to have altered
signaling pathways, known as canonical and non- PCP methylation ().
canonical (). The canonical Wnt signaling is activated
by the binding of Wnt to a serpentine Frizzled re- Origin of APA cells from zona glomerulosa or
ceptor and the coreceptor LRP/, which leads to zona fasciculata
recruitment of Dishevelled protein Dsh and disas- Although aldosterone is synthetized by zona glomer-
sembly of the b-catenin destruction complex () ulosa cells, paradoxically, most APAs are composed of
Once b-catenin is free to move toward to the nucleus, cells with a high cytoplasm-to-nucleus ratio re-
it binds to the TCF/LEF proteins and triggers tran- sembling zona fasciculata cells (e.g., zona fasciculata–
scription of genes involved in cell growth (). The like cells). Despite their morphological appearance,
noncanonical Wnt pathways mostly regulate intracellular zona fasciculata–like cells of these APAs express
Ca+, cell polarity, and migration. By binding to the CYPB, DAB, and CD, putative markers of zona
Frizzled receptor and recruiting Dsh, Wnt activates the glomerulosa cells. Different theories have been pro-
Rho-associated kinase pathway or phospholipase C, with vided to explain the occurrence of zona fasciculata–like
release of Ca+ from intracellular stores (). cells in the APA. One contends that APA cells derive
In the adrenocortical tissue only zona glomerulosa cells from zona glomerulosa cells and that an injury causes
display active Wnt signaling. As mentioned earlier, dis- the morphological change (); another suggests that a
ruption of the Wnt pathway results in abnormal adrenal somatic mutation in zona fasciculata cells causes
development and blunting of aldosterone synthesis. In CYPB overexpression (, ) (Fig. ). To date
mice, mutations in the b-catenin gene (CTNNB) cause there is no clear evidence supporting the superiority of
development of PA and tumors, which can become one theory over the other, leaving this question open.
malignant (, ). Only % of APAs harbor CTNNB A different opinion is that zona fasciculata–like
mutations (), but #% of the tumors exhibit active cells are clear and large because they accumulate
Wnt signaling, suggesting a role of active Wnt in the cholesterol in their cytoplasm, thus displaying lipid

1044 Seccia et al Biology of Zona Glomerulosa and APA Endocrine Reviews, December 2018, 39(6):1029–1056
REVIEW

droplets and morphological ballooning, because they source of autonomous aldosterone production that
are hormonally inactive (). may evolve into an APA. The term APCC-to-APA
The detection of hybrid cell types that coexpress transitional lesion recapitulates this recent hypothesis
the enzymes needed for cortisol [CYPB and cy- (, ) that, though supported by some experts,
tochrome P A (CYP)] and aldosterone remains to be conclusively proven. Moreover, even
synthesis (CYPB) adds further complexity to the though mutations were found in APCCs in in two
issue (). Nakamura et al. () found different types series of adrenal glands from normotensive subjects or
of hybrid cells in APAs characterized by the coex- kidney donors (, ), they were never detected in
pression of CYPB and CYPB, CYPB and APCCs of patients with APAs (, ) (Table ).

Downloaded from https://academic.oup.com/edrv/article-abstract/39/6/1029/5051322 by Research 4 Life user on 22 November 2018


CYP, or CYPB and CYP. Because in an in
vitro model CYPB was found to convert DOC to The two-hit theory
corticosterone and corticosterone to -OH cortico- The two-hit hypothesis was originally conceived in
sterone (), and because immature CYPB/  by Alfred G. Knudson, based on Poisson statistics,
CYPB double-positive cells were found in the to explain onset of nonhereditary retinoblastoma ().
subcapsular zone of murine adrenal glands (, ), it
was suggested that CYPB expression in APAs
could be involved in mineralocorticoid synthesis and
that CYPB/CYPB hybrid cells could represent
an undifferentiated cell phenotype associated with
tumorigenesis (). The CYPB/CYP and
CYPB/CYP hybrid cells were suggested to make
cortisol, but functional evidence is still lacking. The
finding of large cells coexpressing CYPB,
CYPB, and CYP in APAs was even more in-
triguing, because these cells are unlikely to make
steroids efficiently given the divergent direction of the
pathways mediated by CYPB and CYPB/
CYP. Whether the coexpression of all three en-
zymes is a marker of tumorigenesis remains unknown
(). No association was found between such hybrid
cells and somatic KCNJ and ATPase mutations,
leaving uncertain the role played by these mutations in
the genesis of the hybrid cells ().

Origin of APA cells from APCCs


Development of specific antibodies against CYB
and CYPB allowed unequivocal identification of
cortisol- and aldosterone-producing cells and unveiled
different patterns of CYPB and CYPB ex-
pression that underlie lateralized, and thus surgically Figure 6. Steroid synthesis in APA cells and origin of APA cells from zona glomerulosa or
zona fasciculata. [(a), left] Staining of an APA with hematoxylin and eosin identifies zona
curable, human PA (). These findings led us to
glomerulosa–like cells (visualized as pink cells) and zona fasciculata–like cells (yellow). However,
question the classic view of APA as a single, well- hematoxylin and eosin cannot provide functional information about steroidogenetic capability.
demarcated or encapsulated nodule exclusively con- Note the discontinuous layer of zona glomerulosa cells under the capsule. [(a), right]
stituted by cells producing excess aldosterone (). Immunohistochemistry with antibodies against CYP11B1 and CYP11B2 allows identification of
The antibodies showed that islets of CYPB-positive cortisol-producing (green) and aldosterone-producing (violet) cells in the APA, as well as
cells, arranged in cuneiform or trapezoid clusters, aldosterone-producing (violet) cells in the subcapsular clusters. Both CYP11B1- and CYP11B2-
named APCCs, can be present in the zona glomerulosa positive cells can be detected in the APA, suggesting that there is no perfect matching between
zona fasciculata–like cells. Moreover, the APA may also contain CYP17 positive cells or even double
and outer zona fasciculata of normal glands and in
(CYP17 and CYP11B2 or CYP17 and CYP11B1) or triple (CYP17 and CYP11B2 and CYP11B1)
hyperproducing aldosterone glands (). APCCs are positive cells, intermingled with cells that do not show any immunoreaction. (b) Illustration of the
often heterogeneous, with zona glomerulosa cells two-hit theory with its variants. First, an injury or mutation (first hit) in a CYP11B2-positive cell of
intermingled with zona fasciculata cells expressing zona glomerulosa causes hyperproduction of aldosterone and transforms the zona glomerulosa cell
CYPB in the APCC adjacent to the zona fasciculata into a zona fasciculata–like cell; then, activation of a pathway involved in cell growth (e.g., Wnt;
(, ). The findings that APCC cells have higher second hit) causes abnormal proliferation with the tumor mass (APA). Second, the injury or
mutation (first hit) occurs in a CYP11B1-positive cell of zona fasciculata, with transformation of the
CYPB expression levels than the adjacent zona
cell into a cell producing aldosterone; the second hit causes abnormal proliferation with the tumor
glomerulosa cells, some APCCs carry APA-associated mass (APA). Finally, the injury or mutation can associate with expression of other enzymes involved
somatic mutations (, , ), and APCCs resemble in steroidogenesis, as CYP17. To date there are no evidence supporting the superiority of one
APAs in that they harbor zona glomerulosa and zona theory over the other. Note that the colors used to visualize the cells have been chosen arbitrarily.
fasciculata cells suggested that the APCC may be a [© 2018 Illustration Presentation ENDOCRINE SOCIETY]

doi: 10.1210/er.2018-00060 https://academic.oup.com/edrv 1045


REVIEW

In contrast to inherited cancer that can be caused by example by selecting the mutations or by promoting cell
one mutation, sporadic tumors would result from two growth (). Moreover, factors released by neighboring
or more accumulated mutations. This theory has been cells may prevent or promote the transition from normal
proposed also for APAs: the first hit would be a so- to hyperplastic or adenomatous cells. The presence of tiny
matic gene mutation causing excess aldosterone se- nodules in the tissue adjacent to APAs () supports the
cretion, and the second hit would be a novel mutation, view of an altered microenvironment in the entire adrenal
or activation of a system (e.g., Wnt), which induces an gland that harbors the APA. However, whether these
abnormal balance between cell proliferation and ap- nodules in the apparent normal tissue are the precursors of
optosis, leading to nodule formation () (Fig. ). An an APA or favor transition from normal to hyperplastic or

Downloaded from https://academic.oup.com/edrv/article-abstract/39/6/1029/5051322 by Research 4 Life user on 22 November 2018


alternative view is that mutations as KCNJ are the adenomatous cells remains to be clarified.
“second hit,” which follows the “first hit” responsible
for proliferation ().
The two-hit or multiple-hit theory is consistent with Epidemiology of PA
the following: In vitro data show that KCNJ mutations
per se do not increase cell proliferation (), two somatic About a decade after the description of PA, Jerome
mutations were detected in different nodules of the same Conn contended that PA was not a rare disease be-
adrenal gland (, ), a germline APC mutation was cause he detected PA in .% of the hypertensive
found to concur with a somatic KCJN mutation in one patients he had screened, even in the absence of hy-
patient with macronodular adrenal PA (), and uni- pokalemia (). Because others could not confirm
lateral adrenocortical micronodules comprising an this experience, most clinicians continued to believe
outer subcapsular portion of cells expressing only that PA was exceedingly rare, and available estimates
CYPB and an inner portion of cells expressing of PA prevalence varied widely, from .% to %
both CYPB and CYPB and harboring KCNJ (median .%), probably because of differences in the
mutations were reported (). Collectively, these selection criteria for retrospective studies (). This
findings suggest that the onset of somatic or germline gap of knowledge was eventually filled by a large
mutations, possibly associated with some yet unknown prospective survey of consecutive newly diagnosed
mutations or injuries, can produce a spectrum of hypertensive patients referred to specialized hyper-
clinical and pathological phenotypes ranging from tension centers and studied with a predefined di-
APA to micro and macro nodules. agnostic protocol, the PA Prevalence in Hypertensives
(PAPY) study, in  (). This study provided
Abnormal electrical excitability of zona compelling evidence of a high prevalence (.%)
glomerulosa cells among referred hypertensive patients. Subsequently an
Ca+ is needed for aldosterone synthesis; therefore, even higher prevalence of PA was found in patients
prolonged Ca+ entry into zona glomerulosa cells with drug-resistant hypertension by Douma et al.
through voltage-gated calcium channels must be (). Evidence for a high prevalence of PA in patients
postulated when abnormal aldosterone production with hypertension seen in general practice was also
occurs (–). Because isolated zona glomerulosa provided by Olivieri et al. () and more recently
cells are hyperpolarized (2 mV), aldosterone se- confirmed (). Thus, available data support the idea
cretagogues should markedly depolarize the cell to that rather than being a rare disease, PA is the most
allow Ca+ entry, which is difficult to reconcile with the common, albeit overlooked, cause of endocrine
high sensitivity of zona glomerulosa cells to small hypertension.
changes (. mM) in extracellular K+. The demon- The following reasons can account for the
stration that mice zona glomerulosa cells have the underdiagnosis of PA. First, hypokalemia, previously
intrinsic capacity to behave as electrical oscillators that considered a hallmark of PA, is lacking in the ma-
depend on the Cav. provides an explanation for this jority of the cases (). The incidence of normoka-
sensitivity (). Spontaneous rhythmic oscillations of lemia in PA may be due to tissue potassium released
low periodicity in the membrane potential could by the common use of a tourniquet during blood
create a platform that generates a recurring Ca+ signal sampling (, ). Second, current guidelines for
controlled and amplified by aldosterone secretagogues, case detection of PA are seldom applied by general
including Ang II and extracellular K+ (, ). practitioners (). Finally, in most PA, aldosterone is
Mutations of the Ca+ or K+ channels have been within the normal range but with suppressed renin.
hypothesized to amplify these oscillating current sig- PA in these patients remains undiagnosed and often
nals, thereby triggering aldosterone synthesis, but to mislabeled as low-renin essential hypertension, as we
date evidence to support this contention is lacking. recently reported ().
The high prevalence of PA, the severity of car-
Abnormal microenvironment diovascular and renal damage caused by the excess
The microenvironment from which a neoplasm arises aldosterone, and a more favorable outcome of ad-
may play multiple roles in tumor development, for renalectomized patients with APAs compared with

1046 Seccia et al Biology of Zona Glomerulosa and APA Endocrine Reviews, December 2018, 39(6):1029–1056
REVIEW

patients treated pharmacologically necessitate the their positive predictive value, as clearly shown in a
early and accurate screening of hypertensive pa- PAPY study post hoc analysis (). By definition these
tients (, –). For this purpose, a simplified tests identify only the subset of PA cases that are
and more cost-effective strategy is briefly discussed unresponsive to salt or volume suppression of aldo-
below. sterone secretion, notably a minority of cases ().
Moreover, most studies supporting use of these tests
were affected by a tautology bias in that they attempted
Diagnosis of APAs to validate the confirmatory tests not against a gold
reference standard but against another confirmatory

Downloaded from https://academic.oup.com/edrv/article-abstract/39/6/1029/5051322 by Research 4 Life user on 22 November 2018


Patients with PA are characterized by higher cardio- test, based on the presumed but unproven autonomy
vascular morbidity and mortality than age- and sex- of aldosterone secretion from the renin-angiotensin
matched patients with essential hypertension and a system (). The largest study investigating one such
similar degree of blood pressure elevation (, , , test provided unambiguous evidence that when a solid
). Among the cardiovascular complications, atrial diagnosis of APA was used as reference index, neither
fibrillation, the most common arrhythmia worldwide, the ARR value nor the fall of plasma aldosterone
has a significantly higher relative incidence in patients concentration after captopril administration furnished
with PA compared with those with essential hyper- any diagnostic gain over baseline ARR values in two
tension (). This is not unexpected, given the cohorts of patients (). This study calls for a sim-
compelling evidence relating aldosterone to atrial fi- plification of the diagnostic algorithm to include only
brillation (). Thus, early identification of patients the determination of the ARR at baseline, a strategy that
with PA, followed by targeted treatment, translates should extend screening for PA to most, or even all,
into prevention of morbid events and reversal of hypertensive patients, even in municipalities with low
cardiovascular damage. Compelling evidence has been levels of access to specialized medical care.
recently provided by completion of the long-term Use of the cutoff values of ARR for diagnosis of PA
longitudinal phase of the PAPY Study, where adre- implies a definition of PA as a categorical disorder.
nalectomy was found to lower the risk of atrial fi- However, the available evidence suggests that the
brillation in patients with PA (). The Endocrine spectrum of autonomous aldosterone secretion extends
Society Guidelines suggest screening for PA in patients beyond cases featuring an overt phenotype of hyper- “PA mimicking “low-renin
with a high pretest probability for PA (Table ) (), tension or hypokalemia. By analyzing a cohort of essential” hypertension could
but a broader systematic screening for PA in most or normotensive subjects, Baudrand et al. () found a be far more common than
all hypertensive patients is endorsed by other experts, continuous spectrum of autonomous aldosterone se- currently perceived.”
based on the high prevalence rate of PA and the high cretion, ranging from subtle to overtly dysregulated
rate of cardiovascular damage (). Unfortunately, aldosterone secretion, defined as suppressed PRA
because of the economic burden, wider screening is (,. ng/mL/h) and elevated aldosterone excretion
not feasible in many municipalities and several de- rate ($ mg with a urinary sodium excretion
veloped countries. rate . mmol/d). Together with data showing a %
The diagnosis of PA requires demonstration of prevalence of PA in normotensive subjects from a
excessive aldosterone secretion in relative autonomy general population survey (, ) and the demon-
of the renin-angiotensin aldosterone system (). stration of APAs in normoaldosteronemic patients
This is commonly done with concomitant mea- (), this evidence suggests that autonomous aldoste-
surements of plasma aldosterone levels and renin rone secretion may begin early in normotensive subjects
activity or concentration and calculation of the and then may gradually transform into low-renin “es-
aldosterone-to-renin ratio (ARR) (). Direct renin sential” hypertension, then to more or less florid PA,
concentration has replaced plasma renin activity finally evolving into stage II to III or drug-resistant
(PRA) in many laboratories, rendering measure- hypertension. Therefore, PA mimicking “low-renin es-
ments of renin simpler, quicker, and more accurate sential” hypertension could be far more common than
(). Different cutoff values of ARR, using either currently perceived. Combined measurement of the
PRA or direct renin concentration, have been ARR and -hour urinary sodium excretion would allow
proposed; only those obtained from large pro- early identification of these patients. Although animal
spective studies are accepted for the diagnosis of PA studies support a direct interaction between high salt
(, ). intake and aldosterone-induced increase in blood
A number of so-called confirmatory tests, in- pressure and related target organ damage (, ),
cluding the captopril challenge test, are still used in whether institution of lifestyle intervention based on low
some centers to confirm the diagnosis of PA. With the sodium and high potassium intake is a valuable strategy
prevalence rate of PA between % and % in pa- to prevent transition from “low-renin essential” hyper-
tients referred to specialized centers, these tests tension to overt PA remains to be proven.
function as exclusionary rather than confirmatory tests Adrenal CT is useful to detect a large mass that may
because their negative predictive value largely exceeds represent adrenocortical carcinoma and to assist the

doi: 10.1210/er.2018-00060 https://academic.oup.com/edrv 1047


REVIEW

Table 3. Conditions That Should Make the Search for PA Mandatory According to the Endocrine Society Guidelines
Data from Funder JW, Carey
RM, Mantero F, et al. The 1. Unexplained hypokalemia (spontaneous or diuretic-induced)
management of primary
aldosteronism: case detection, 2. Hypertension (BP .140/90 mm Hg) resistant to 3 antihypertensive drugs (including a diuretic)
diagnosis, and treatment. An
3. Controlled BP (.140/90 mm Hg) on $4 antihypertensive drugs
Endocrine Society Clinical
Practice Guideline. J Clin 4. Hypertension and spontaneous or diuretic-induced hypokalemia
Endocrinol Metab. 2016;101(5):
1889–1916. 5. Hypertension and adrenal incidentaloma

Downloaded from https://academic.oup.com/edrv/article-abstract/39/6/1029/5051322 by Research 4 Life user on 22 November 2018


6. Obstructive sleep apnea syndrome

7. Hypertension and a family history of early-onset hypertension or cerebrovascular accident at a young age (,40 y)

8. All hypertensive first-degree relatives of patients with PA

9. Incidentally discovered, apparently nonfunctioning adrenal mass (“incidentaloma”)

10. Evidence of organ damage that is disproportionate for the severity of hypertension

interventional radiologist and surgeon if the patient phenotype, comprising spontaneous hypokalemia,
needs further treatment (, ). MRI has no ad- marked aldosterone excess, suppressed plasma renin, a
vantage over CT in subtype evaluation of PA; in addition unilateral adrenal lesion with radiologic features
to being more expensive, it has less spatial resolution consistent with a cortical adenoma, and a contralateral
than CT (). However, very small APAs (, mm) or normal gland on CT (). However, even for young
adrenal zona glomerulosa hyperplasia cannot be visual- patients, bilateral aldosterone secretion cannot be
ized with either CT or MRI (). excluded without AVS (). An in-depth review of
The most common forms of PA are unilateral the use of AVS has been recently published ().
APAs, which can be treated with unilateral adrenal- Demonstration of a CYPB-positive adenoma at
ectomy, and bilateral idiopathic hyperplasia [idio- pathology provides a conclusive diagnosis of the PA
pathic hyperaldosteronism (IHA)], which requires subtype, which in lateralized forms of PA can be an APA
lifelong mineralocorticoid receptor antagonists. The or unilateral multinodular adrenocortical hyperplasia (,
distinction between APA and IHA is crucial to identify ). Thus, in the PAPY Study the “four corners” criteria
the appropriate treatment (, ). were introduced more than a decade ago for the diagnosis
Adrenal venous sampling (AVS) is currently the of APA (). With the availability of monoclonal anti-
only way to reliably discriminate between APA and bodies for human CYPB, these criteria must be
IHA, because CT and MRI have poor accuracy (). amended by the addition of immunohistochemical de-
Unilateral aldosterone excess from a small, CT- tection of CYPB in the resected adrenal (Table ) ().
undetectable APAs in the adrenal gland contralateral In patients with PA , years of age or with a
to a CT-detectable nonfunctioning adrenal mass cannot family history of PA or stroke at a young age (,
be reliably identified without AVS (). However, AVS years), the guidelines suggest genetic testing for FH-I
is a minimally invasive but technically difficult and [i.e., glucocorticoid-remediable aldosteronism (GRA)] and
expensive procedure, which is potentially affected by for germline mutations of KCNJ causing FH-III ().
several factors (, ). For these reasons it should be
performed only in centers endowed with a skilled
multidisciplinary team with extensive expertise and in The Biomarkers of APA
properly selected patients (). As a preliminary test
for adrenalectomy, AVS should be reserved for patients A relentless search for biomarkers for APA has
who are seeking long-term cure of PA with surgery and continued for decades, given the limited availability of
are reasonable candidates for general anesthesia and AVS. Unfortunately, this remains a challenging un-
adrenalectomy (, ). AVS should be performed resolved issue, as we recently reviewed in depth ()
after correction of hypokalemia, if present, and ad- and update here.
justment of antihypertensive medications to allow
correct interpretation of the AVS results (). The circulating markers of APA and the
The Endocrine Society guidelines suggest that steroid profiles
unilateral adrenalectomy should not be performed Regarding circulating biomarkers, one approach has
without prior demonstration of lateralized aldosterone been the measurement of -oxocortisol in peripheral
production by AVS. An exception to this rule may be venous plasma to distinguish patients with bilateral
young patients (age , years) with a florid PA adrenal hyperplasia from those with an APA (,

1048 Seccia et al Biology of Zona Glomerulosa and APA Endocrine Reviews, December 2018, 39(6):1029–1056
REVIEW

). Another approach has been to use liquid


chromatography–tandem mass spectrometry (LC-MS/ Table 4. “Five Corners” Criteria for the Diagnosis of APA
The “Five corners” criteria imply
MS) to determine the steroid profiles in urine and 1. Biochemical evidence of PA (e.g., an inappropriately high that, in a setting of PA, a nodule
peripheral venous plasma. Although LC-MS/MS aldosterone/renin ratio) can be defined as an
showed -oxocortisol levels in peripheral plasma to aldosterone-producing
2. Lateralized aldosterone secretion by AVS
be on average . times higher in patients with APAs adrenocortical adenoma after
evidence of CYP11B2
than IHA, a more recent study by Eisenhofer et al. () 3. Detection of a nodule by imaging (CT or MRI) and an immunoreactivity at
showed a marked overlap of values between PA sub- adenoma at pathology immunohistochemistry.
types, thus questioning the usefulness of this marker for 4. Biochemical correction of PA after adrenalectomy

Downloaded from https://academic.oup.com/edrv/article-abstract/39/6/1029/5051322 by Research 4 Life user on 22 November 2018


discrimination purposes. This led to the proposal of
using principal component or discriminant analysis of 5. Detection of a CYP11B2-positive adenoma in the resected
adrenal cortex at immunohistochemistry with a
the profile of all  adrenal steroids identified with LC-
monoclonal antibody for human CYP11B2
MS/MS, which was reported to achieve correct classi-
fication of % of cases of APAs. Of potential relevance,
in one study steroid profiles in peripheral samples
identified % of the  patients who did not benefit The tissue markers of APA
from adrenalectomy or in whom AVS indicated IHA, Not only has the search for the circulating markers
suggesting that LC-MS/MS could be an approach to been difficult, but reliable cellular biomarkers for APA
avoid AVS in such patients. However, because LC-MS/ cells have been a major hurdle. For decades, the ex-
MS and complex statistical analysis entail techniques pression of CYPB was thought to be the signature
unavailable in most centers, this proposal to replace AVS of zona glomerulosa and APA cells, but this turned out
with an even less available strategy seems premature. to be untrue. Different laboratories have identified
LC-MS/MS can be useful to detect excess lateralized genes and proteins overexpressed or underexpressed
secretion of aldosterone at AVS: LC-MS/MS-derived in zona glomerulosa and APA cells that could be in-
lateralization ratios of aldosterone normalized to cor- volved in steroidogenesis or cell growth, but each
tisol and to the hybrid steroids -oxocortisol and candidate marker raised questions that still await
-hydroxycortisol were significantly higher than answers [for a review, see ()]. The most promising
immunoassay-derived ratios (), but LC-MS/MS- markers for aldosterone-producing cells are CD
derived lateralized secretion of the hybrid steroids (NM_) and Dab (NM_), which are
was found only in % and %, respectively, of patients both markedly expressed in zona glomerulosa and
with APAs. Nonetheless, by identifying steroids such as APA, with the former also expressed in some zona
aOH progesterone and, even more so, androstene- fasciculata cells and the latter missing in some zona
dione that have a much larger step difference between glomerulosa cells. Because CD is expressed at the cell
the adrenal vein blood and the inferior vena cava blood, membrane level, it is successfully being used to isolate
LC-MS/MS has been instrumental in improving the aldosterone-producing cells for research purposes
indexes for assessing selectivity and lateralization (). (). We recently proposed that the combination of
Regarding the measurement of steroids in the urine, the two markers would allow identification of all
gas chromatography–mass spectrometry has been used to aldosterone-producing cells, but studies designed to
characterize the urine steroid metabolome of patients with investigate this issue are lacking thus far. After the
PA (). A small but statistically significant increase in publication of our recent review on the markers of
excretion of cortisol and total glucocorticoid metabolites zona glomerulosa, two markers have been proposed.
was found in patients with PA who had no sign of CXC chemokine receptor type  (CXCR), which
glucocorticoid excess, suggesting that a mild glucocorticoid binds its ligand CXC chemokine CXCL (stromal
excess could be a feature of some patients with PA. Al- cell-derived factor-), putatively involved in cell migration,
though of potential relevance, both strategies should be was found to be overexpressed in aldosterone-producing
validated in larger samples with the gold reference stan- tissue of normal adrenals and in about two-thirds of
dard of the diagnosis of APA as defined above (Table ). APAs compared with nonfunctional adenomas ().
Finally, autoimmune mechanisms have been suggested Therefore, CXCR has been proposed as a marker of
as a cause of human PA. Autoantibodies against type- APA. Limited ex vivo autoradiography and in vivo
angiotensin II receptor (ATAAs) were isolated from the positron emission tomography imaging studies, using
plasma of patients with PA at levels comparable to those the CXCR ligand Ga-pentixafor- (GaCPCR.)
found in preeclamptic women (, ). Moreover, cir- as tracer, claimed specificity for aldosterone-producing
culating ATAA levels discriminated between APAs and tissues, pointing to positron emission tomography
IHA, suggesting that they might be useful not only for imaging as a promising approach for noninvasive
diagnosing PA but also for differentiating APA from non- characterization of adrenal lesions in PA. However,
APA conditions (). Whether ATAAs, because of their because CXCR is underexpressed in about one-third
agonistic activity, have an etiologic role in the development of APAs and results were heterogeneous even in a very
of APA remains to be clarified (, ). small series of highly selected patients with APA,

doi: 10.1210/er.2018-00060 https://academic.oup.com/edrv 1049


REVIEW

caution is warranted in drawing conclusions at this of PA in the last decade eclipses that of all other
stage (). endocrine causes of arterial hypertension. The
Transcriptome analysis recently revealed that advancements involve molecular mechanisms and
among the genes encoding Ca+-binding proteins in immunophenotyping, as well as the diagnostic
APA, the CALN gene encoding calneuron  showed the workup of patients from screening to subtyping.
strongest correlation with CYPB (). Calneuron  is Nonetheless, little doubt exists that PA is a
located in the endoplasmic reticulum, where it binds markedly underdiagnosed condition, despite the
cytosolic Ca+ and enhances Ca+ storage. Upon IP fact that its high prevalence was identified by
receptor signaling it releases Ca+, thus promoting Jerome Conn . years ago. Major factors con-

Downloaded from https://academic.oup.com/edrv/article-abstract/39/6/1029/5051322 by Research 4 Life user on 22 November 2018


CYPB transcription. Moreover, aldosterone pro- tributing to neglecting PA as the most important
duction in HAC cells was potentiated by CALN ex- cause of secondary hypertension are misconcep-
pression, and, conversely, CALN silencing decreased Ca+ tions, particularly the belief that PA is an excep-
in the endoplasmic reticulum and abrogated Ang II- and tional cause of arterial hypertension and that the
KCNJ TA–mediated aldosterone production. Thus, diagnosis is complex and out of reach for many
elevated CALN could be a marker of excess aldosterone doctors. An overwhelming amount of data sup-
production (). However, because what matters for ports exactly the opposite; PA is the most common
activation of aldosterone biosynthesis is mitochondrial curable cause of hypertension, and its diagnosis is
Ca+ rather than endoplasmic reticulum Ca+ (), how simple and should not discourage anyone. Over-
the latter is transferred to the mitochondria and whether looking PA translates into missing the opportunity
the calcium uniporter () plays a role in this process are for a cure or optimal treatment of many patients.
fundamental questions that await an answer. Accordingly, in our view opinion leaders have an
ethical obligation to spread the message and work
to improve the education of all primary care
Conclusions providers. The updated information provided in
this review can be a small step to bridge the gap
There is no doubt that the progress made in our between new knowledge and its implementation in
understanding of the mechanisms of the pathogenesis clinical practice.

References
1. Bentley-Lewis R, Adler GK, Perlstein T, Seely EW, 5. Nanba K, Vaidya A, Williams GH, Zheng I, Else T, aldosteronism in 1,125 hypertensive patients. J Am
Hopkins PN, Williams GH, Garg R. Body mass index Rainey WE. Age-related autonomous aldosteron- Coll Cardiol. 2006;48(11):2293–2300.
predicts aldosterone production in normotensive ism. Circulation. 2017;136(4):347–355. 11. Aragao-Santiago L, Gomez-Sanchez CE, Mulatero P,
adults on a high-salt diet. J Clin Endocrinol Metab. 6. Brown JM, Robinson-Cohen C, Luque-Fernandez Spyroglou A, Reincke M, Williams TA. Mouse
2007;92(11):4472–4475. MA, Allison MA, Baudrand R, Ix JH, Kestenbaum models of primary aldosteronism: from physiology
2. Rossi GP, Belfiore A, Bernini G, Fabris B, Caridi G, B, de Boer IH, Vaidya A. The spectrum of subclinical to pathophysiology. Endocrinology. 2017;158(12):
Ferri C, Giacchetti G, Letizia C, Maccario M, primary aldosteronism and incident hypertension: 4129–4138.
Mannelli M, Palumbo G, Patalano A, Rizzoni D, a cohort study. Ann Intern Med. 2017;167(9): 12. Lenzini L, Caroccia B, Campos AG, Fassina A, Belloni
Rossi E, Pessina AC, Mantero F; Primary Aldoste- 630–641. AS, Seccia TM, Kuppusamy M, Ferraro S, Skander G,
ronism Prevalence in Hypertension Study In- 7. Baudrand R, Guarda FJ, Fardella C, Hundemer G, Bader M, Rainey WE, Rossi GP. Lower expression of
vestigators. Body mass index predicts plasma Brown J, Williams G, Vaidya A. Continuum of renin- the TWIK-related acid-sensitive K+ channel 2
aldosterone concentrations in overweight–obese independent aldosteronism in normotension. Hy- (TASK-2) gene is a hallmark of aldosterone-
primary hypertensive patients. J Clin Endocrinol pertension. 2017;69(5):950–956. producing adenoma causing human primary al-
Metab. 2008;93(7):2566–2571. 8. Brown JM, Underwood PC, Ferri C, Hopkins PN, dosteronism. J Clin Endocrinol Metab. 2014;99(4):
3. Williams B, MacDonald TM, Morant S, Webb DJ, Williams GH, Adler GK, Vaidya A. Aldosterone E674–E682.
Sever P, McInnes G, Ford I, Cruickshank JK, dysregulation with aging predicts renal vascular 13. Rossi GP, Ragazzo F, Seccia TM, Maniero C, Barisa M,
Caulfield MJ, Salsbury J, Mackenzie I, Padmanabhan function and cardiovascular risk. Hypertension. 2014; Calò LA, Frigo AC, Fassina A, Pessina AC. Hyper-
S, Brown MJ; British Hypertension Society’s 63(6):1205–1211. parathyroidism can be useful in the identification of
PATHWAY Studies Group. Spironolactone versus 9. Monticone S, Burrello J, Tizzani D, Bertello C, Viola primary aldosteronism due to aldosterone-
placebo, bisoprolol, and doxazosin to determine A, Buffolo F, Gabetti L, Mengozzi G, Williams TA, producing adenoma. Hypertension. 2012;60(2):
the optimal treatment for drug-resistant hyper- Rabbia F, Veglio F, Mulatero P. Prevalence and 431–436.
tension (PATHWAY-2): a randomised, double- clinical manifestations of primary aldosteronism 14. Olgaard K, Lewin E, Bro S, Daugaard H, Egfjord M,
blind, crossover trial. Lancet. 2015;386(10008): encountered in primary care practice. J Am Coll Pless V. Enhancement of the stimulatory effect of
2059–2068. Cardiol. 2017;69(14):1811–1820. calcium on aldosterone secretion by parathyroid
4. Newton-Cheh C, Guo CY, Gona P, Larson MG, 10. Rossi GP, Bernini G, Caliumi C, Desideri G, Fabris B, hormone. Miner Electrolyte Metab. 1994;20(5):
Benjamin EJ, Wang TJ, Kathiresan S, O’Donnell CJ, Ferri C, Ganzaroli C, Giacchetti G, Letizia C, 309–314.
Musone SL, Camargo AL, Drake JA, Levy D, Maccario M, Mallamaci F, Mannelli M, Mattarello 15. Isales CM, Barrett PQ, Brines M, Bollag W,
Hirschhorn JN, Vasan RS. Clinical and genetic cor- M-J, Moretti A, Palumbo G, Parenti G, Porteri E, Rasmussen H. Parathyroid hormone modulates
relates of aldosterone-to-renin ratio and relations to Semplicini A, Rizzoni D, Rossi E, Boscaro M, Pessina angiotensin II–induced aldosterone secretion from
blood pressure in a community sample. Hyperten- AC, Mantero F; PAPY Study Investigators. A pro- the adrenal glomerulosa cell. Endocrinology. 1991;
sion. 2007;49(4):846–856. spective study of the prevalence of primary 129(1):489–495.

1050 Seccia et al Biology of Zona Glomerulosa and APA Endocrine Reviews, December 2018, 39(6):1029–1056
REVIEW

16. Rossitto G, Regolisti G, Rossi E, Negro A, Nicoli D, 28. Rossi GP, Sacchetto A, Pavan E, Palatini P, Graniero K, Ito S, Sasano H, Okamura H. Isoform-specific
Casali B, Toniato A, Caroccia B, Seccia TM, Walther GR, Canali C, Pessina AC. Remodeling of the left monoclonal antibodies against 3b-hydroxyste-
T, Rossi GP. Elevation of angiotensin-II type-1-receptor ventricle in primary aldosteronism due to Conn’s roid dehydrogenase/isomerase family provide
autoantibodies titer in primary aldosteronism as a adenoma. Circulation. 1997;95(6):1471–1478. markers for subclassification of human primary
result of aldosterone-producing adenoma. Hyperten- 29. Pitt B, Zannad F, Remme WJ, Cody R, Castaigne A, aldosteronism. J Clin Endocrinol Metab. 2014;99(2):
sion. 2013;61(2):526–533. Perez A, Palensky J, Wittes J; Randomized Aldactone E257–E262.
17. Kem DC, Li H, Velarde-Miranda C, Liles C, Evaluation Study Investigators. The effect of spi- 44. Gomez-Sanchez CE, Lewis M, Nanba K, Rainey WE,
Vanderlinde-Wood M, Galloway A, Khan M, Zillner ronolactone on morbidity and mortality in patients Kuppusamy M, Gomez-Sanchez EP. Development
C, Benbrook A, Rao V, Gomez-Sanchez CE, with severe heart failure. N Engl J Med. 1999;341(10): of monoclonal antibodies against the human 3b-
Cunningham MW, Yu X. Autoimmune mecha- 709–717. hydroxysteroid dehydrogenase/isomerase isozymes.
nisms activating the angiotensin AT1 receptor in 30. Pitt B, Remme W, Zannad F, Neaton J, Martinez F, Steroids. 2017;127(August):56–61.
“primary” aldosteronism. J Clin Endocrinol Metab. Roniker B, Bittman R, Hurley S, Kleiman J, Gatlin M; 45. Zubair M, Ishihara S, Oka S, Okumura K, Morohashi

Downloaded from https://academic.oup.com/edrv/article-abstract/39/6/1029/5051322 by Research 4 Life user on 22 November 2018


2014;99(5):1790–1797. Eplerenone Post-Acute Myocardial Infarction Heart K. Two-step regulation of Ad4BP/SF-1 gene tran-
18. Li H, Yu X, Cicala MV, Mantero F, Benbrook A, Failure Efficacy and Survival Study Investigators. scription during fetal adrenal development: initia-
Veitla V, Cunningham MW, Kem DC. Prevalence of Eplerenone, a selective aldosterone blocker, in
tion by a Hox-Pbx1-Prep1 complex and maintenance
angiotensin II type 1 receptor (AT1R)-activating patients with left ventricular dysfunction after
via autoregulation by Ad4BP/SF-1. Mol Cell Biol. 2006;
autoantibodies in primary aldosteronism. J Am myocardial infarction. N Engl J Med. 2003;348(14):
26(11):4111–4121.
Soc Hypertens. 2015;9(1):15–20. 1309–1321.
46. Hershkovitz L, Beuschlein F, Klammer S, Krup M,
19. Choi M, Scholl UI, Yue P, Björklund P, Zhao B, 31. Pfeffer MA, Claggett B, Assmann SF, Boineau R,
Weinstein Y. Adrenal 20alpha-hydroxysteroid de-
Nelson-Williams C, Ji W, Cho Y, Patel A, Men CJ, Anand IS, Clausell N, Desai AS, Diaz R, Fleg JL,
hydrogenase in the mouse catabolizes pro-
Lolis E, Wisgerhof MV, Geller DS, Mane S, Hellman Gordeev I, Heitner JF, Lewis EF, O’Meara E, Rouleau
J-L, Probstfield JL, Shaburishvili T, Shah SJ, Solomon gesterone and 11-deoxycorticosterone and is
P, Westin G, Åkerström G, Wang W, Carling T, restricted to the X-zone. Endocrinology. 2007;148(3):
Lifton RP. K+ channel mutations in adrenal SD, Sweitzer NK, McKinlay SM, Pitt B. Regional
variation in patients and outcomes in the Treat- 976–988.
aldosterone-producing adenomas and hereditary 47. Mitani F, Suzuki H, Hata J, Ogishima T, Shimada H,
hypertension. Science. 2011;331(6018):768–772. ment of Preserved Cardiac Function Heart Failure
With an Aldosterone Antagonist (TOPCAT) trial. Ishimura Y. A novel cell layer without corticosteroid-
20. Boulkroun S, Beuschlein F, Rossi GP, Golib-Dzib JF, synthesizing enzymes in rat adrenal cortex: histo-
Fischer E, Amar L, Mulatero P, Samson-Couterie B, Circulation. 2015;131(1):34–42.
32. Zannad F, McMurray JJV, Krum H, van Veldhuisen chemical detection and possible physiological role.
Hahner S, Quinkler M, Fallo F, Letizia C, Allolio B,
DJ, Swedberg K, Shi H, Vincent J, Pocock SJ, Pitt B; Endocrinology. 1994;135(1):431–438.
Ceolotto G, Cicala MV, Lang K, Lefebvre H, Lenzini
EMPHASIS-HF Study Group. Eplerenone in patients 48. Romero DG, Yanes LL, de Rodriguez AF, Plonczynski
L, Maniero C, Monticone S, Perrocheau M, Pilon C,
with systolic heart failure and mild symptoms. MW, Welsh BL, Reckelhoff JF, Gomez-Sanchez EP,
Plouin PF, Rayes N, Seccia TM, Veglio F, Williams TA,
N Engl J Med. 2011;364(1):11–21. Gomez-Sanchez CE. Disabled-2 is expressed in
Zinnamosca L, Mantero F, Benecke A, Jeunemaitre
33. Miller WL. A brief history of adrenal research: adrenal zona glomerulosa and is involved in al-
X, Reincke M, Zennaro MC. Prevalence, clinical, and
steroidogenesis—the soul of the adrenal. Mol Cell dosterone secretion. Endocrinology. 2007;148(6):
molecular correlates of KCNJ5 mutations in primary
Endocrinol. 2013;371(1–2):5–14. 2644–2652.
aldosteronism. Hypertension. 2012;59(3):592–598.
34. Malendowicz LK. 100th anniversary of the discovery 49. Gomez-Sanchez CE, Qi X, Velarde-Miranda C,
21. Lenzini L, Rossitto G, Maiolino G, Letizia C, Funder
of the human adrenal fetal zone by Stella Starkel Plonczynski MW, Parker CR, Rainey W, Satoh F,
JW, Rossi GP. A meta-analysis of somatic KCNJ5
and Lesław We˛grzynowski: how far have we come? Maekawa T, Nakamura Y, Sasano H, Gomez-
K(+) channel mutations in 1636 patients with an
Folia Histochem Cytobiol. 2010;48(4):491–506. Sanchez EP. Development of monoclonal anti-
aldosterone-producing adenoma. J Clin Endocrinol
35. Williams JS, Williams GH. 50th anniversary of al- bodies against human CYP11B1 and CYP11B2. Mol
Metab. 2015;100(8):E1089–E1095.
dosterone. J Clin Endocrinol Metab. 2003;88(6): Cell Endocrinol. 2014;383(1-2):111–117.
22. Wu V-C, Wang S-M, Chueh SJ, Yang S-Y, Huang
2364–2372. 50. Nishimoto K, Koga M, Seki T, Oki K, Gomez-
K-H, Lin Y-H, Wang J-J, Connolly R, Hu Y-H, Gomez-
36. Tait SAS, Tait JF, Coghlan JP. The discovery, isolation Sanchez EP, Gomez-Sanchez CE, Naruse M,
Sanchez CE, Peng K-Y, Wu K-D. The prevalence of and identification of aldosterone: reflections on
CTNNB1 mutations in primary aldosteronism and Sakaguchi T, Morita S, Kosaka T, Oya M, Ogishima
emerging regulation and function. Mol Cell Endo- T, Yasuda M, Suematsu M, Kabe Y, Omura M,
consequences for clinical outcomes. Sci Rep. 2017;7: crinol. 2004;217(1-2):1–21.
39121. Nishikawa T, Mukai K. Immunohistochemistry of
37. Litynski M. [Hypertension caused by tumors of the aldosterone synthase leads the way to the path-
23. Scholl UI, Stölting G, Schewe J, Thiel A, Tan H, adrenal cortex]. Pol Tyg Lek (Wars). 1953;8(6):
Nelson-Williams C, Vichot AA, Jin SC, Loring E, ogenesis of primary aldosteronism. Mol Cell Endo-
204–208. crinol. 2017;441:124–133.
Untiet V, Yoo T, Choi J, Xu S, Wu A, Kirchner M, 38. Gottschau M. Struktur und embryonale Entwick-
Mertins P, Rump LC, Onder AM, Gamble C, 51. Boon WC, Coghlan JP, McDougall JG. Late steps of
lung der Nebennieren bei Säugetieren. Arch Anat
McKenney D, Lash RW, Jones DP, Chune G, aldosterone biosynthesis: sheep are not rats. Clin
Physiol. 1883;9:412–458.
Gagliardi P, Choi M, Gordon R, Stowasser M, Fahlke Exp Pharmacol Physiol Suppl. 1998;25(S1):S21–S27.
39. Pignatti E, Leng S, Carlone DL, Breault DT. Regu-
C, Lifton RP. CLCN2 chloride channel mutations in 52. Gruenwald P. Embryonic and postnatal develop-
lation of zonation and homeostasis in the adrenal
familial hyperaldosteronism type II. Nat Genet. 2018; ment of the adrenal cortex, particularly the zona
cortex. Mol Cell Endocrinol. 2017;441:146–155.
50(3):349–354. glomerulosa and accessory nodules. Anat Rec. 1946;
40. Mazzocchi G, Rebuffat P, Belloni AS, Robba C,
24. Fernandes-Rosa FL, Daniil G, Orozco IJ, Göppner C, Nussdorfer GG. An ultrastructural stereologic study 95(4):391–421.
El Zein R, Jain V, Boulkroun S, Jeunemaitre X, Amar 53. Kim AC, Barlaskar FM, Heaton JH, Else T, Kelly VR,
of the effects of angiotensin II on the zona glo-
L, Lefebvre H, Schwarzmayr T, Strom TM, Jentsch TJ, merulosa of rat adrenal cortex. Acta Endocrinol Krill KT, Scheys JO, Simon DP, Trovato A, Yang
Zennaro M-C. A gain-of-function mutation in the (Copenh). 1980;95(4):523–527. W-H, Hammer GD. In search of adrenocortical
CLCN2 chloride channel gene causes primary al- 41. Nussdorfer GG, Mazzocchi G, Rebuffat P. An ul- stem and progenitor cells. Endocr Rev. 2009;30(3):
dosteronism. Nat Genet. 2018;50(3):355–361. trastructural stereologic study of the effects of 241–263.
25. Åkerström T, Maharjan R, Sven Willenberg H, 39,59-cyclic nucleotides on the zona glomerulosa of 54. Pihlajoki M, Dörner J, Cochran RS, Heikinheimo M,
Cupisti K, Ip J, Moser A, Stålberg P, Robinson B, rat adrenal cortex. Endocrinology. 1974;94(2): Wilson DB. Adrenocortical zonation, renewal, and
Alexander Iwen K, Dralle H, Walz MK, Lehnert H, 602–607. remodeling. Front Endocrinol (Lausanne). 2015;6:27.
Sidhu S, Gomez-Sanchez C, Hellman P, Björklund P. 42. Doi M, Takahashi Y, Komatsu R, Yamazaki F, 55. Hammer GD, Ingraham HA. Steroidogenic factor-1:
Activating mutations in CTNNB1 in aldosterone Yamada H, Haraguchi S, Emoto N, Okuno Y, its role in endocrine organ development and dif-
producing adenomas. Sci Rep. 2016;6(1):19546. Tsujimoto G, Kanematsu A, Ogawa O, Todo T, ferentiation. Front Neuroendocrinol. 1999;20(3):
26. Fernandes-Rosa FL, Boulkroun S, Zennaro M-C. Tsutsui K, van der Horst GTJ, Okamura H. Salt- 199–223.
Somatic and inherited mutations in primary al- sensitive hypertension in circadian clock–deficient 56. Hammer GD, Parker KL, Schimmer BP. Minireview:
dosteronism. J Mol Endocrinol. 2017;59(1):R47–R63. Cry-null mice involves dysregulated adrenal transcriptional regulation of adrenocortical devel-
27. Lenzini L, Rossi GP. The molecular basis of primary Hsd3b6. Nat Med. 2010;16(1):67–74. opment. Endocrinology. 2005;146(3):1018–1024.
aldosteronism: from chimeric gene to channelop- 43. Doi M, Satoh F, Maekawa T, Nakamura Y, Fustin J-M, 57. Morohashi K. The ontogenesis of the steroidogenic
athy. Curr Opin Pharmacol. 2015;21:35–42. Tainaka M, Hotta Y, Takahashi Y, Morimoto R, Takase tissues. Genes Cells. 1997;2(2):95–106.

doi: 10.1210/er.2018-00060 https://academic.oup.com/edrv 1051


REVIEW

58. Mesiano S, Jaffe RB. Developmental and functional capsule is a signaling center controlling cell renewal study. J Clin Endocrinol Metab. 2015;100(11):
biology of the primate fetal adrenal cortex. Endocr and zonation through Rspo3. Genes Dev. 2016; 4074–4081.
Rev. 1997;18(3):378–403. 30(12):1389–1394. 88. Kowarski A, Katz H, Migeon CJ. Plasma aldosterone
59. Xing Y, Lerario AM, Rainey W, Hammer GD. De- 75. Deane HW, Greep RO. The morphology and his- concentration in normal subjects from infancy to
velopment of adrenal cortex zonation. Endocrinol tochemistry of the adrenal cortex after hypophy- adulthood. J Clin Endocrinol Metab. 1974;38(3):
Metab Clin North Am. 2015;44(2):243–274. sectomy. Anat Rec. 1946;94:412. 489–491.
60. Lerario AM, Finco I, LaPensee C, Hammer GD. 76. King P, Paul A, Laufer E. Shh signaling regulates 89. Weldon VV, Kowarski A, Migeon CJ. Aldosterone
Molecular mechanisms of stem/progenitor cell adrenocortical development and identifies pro- secretion rates in normal subjects from infancy to
maintenance in the adrenal cortex. Front Endocrinol genitors of steroidogenic lineages. Proc Natl Acad adulthood. Pediatrics. 1967;39(5):713–723.
(Lausanne). 2017;8:52. Sci USA. 2009;106(50):21185–21190. 90. Nishimoto K, Nakagawa K, Li D, Kosaka T, Oya M,
61. Hanley NA, Rainey WE, Wilson DI, Ball SG, Parker KL. 77. Heikkilä M, Peltoketo H, Leppäluoto J, Ilves M, Mikami S, Shibata H, Itoh H, Mitani F, Yamazaki T,
Expression profiles of SF-1, DAX1, and CYP17 in Vuolteenaho O, Vainio S. Wnt-4 deficiency alters Ogishima T, Suematsu M, Mukai K. Adrenocortical

Downloaded from https://academic.oup.com/edrv/article-abstract/39/6/1029/5051322 by Research 4 Life user on 22 November 2018


the human fetal adrenal gland: potential inter- mouse adrenal cortex function, reducing aldoste- zonation in humans under normal and pathological
actions in gene regulation. Mol Endocrinol. 2001; rone production. Endocrinology. 2002;143(11): conditions. J Clin Endocrinol Metab. 2010;95(5):
15(1):57–68. 4358–4365. 2296–2305.
62. Keegan CE, Hammer GD. Recent insights into or- 78. Drelon C, Berthon A, Mathieu M, Martinez A, Val P. 91. Aiba M, Fujibayashi M. Alteration of subcapsular
ganogenesis of the adrenal cortex. Trends Endocrinol Adrenal cortex tissue homeostasis and zonation: adrenocortical zonation in humans with aging: the
Metab. 2002;13(5):200–208. a WNT perspective. Mol Cell Endocrinol. 2015;408: progenitor zone predominates over the previously
63. Ishimoto H, Jaffe RB. Development and function of 156–164. well-developed zona glomerulosa after 40 years of
the human fetal adrenal cortex: a key component in 79. Berthon A, Sahut-Barnola I, Lambert-Langlais S, de age. J Histochem Cytochem. 2011;59(5):557–564.
the feto-placental unit. Endocr Rev. 2011;32(3): Joussineau C, Damon-Soubeyrand C, Louiset E, 92. Boulkroun S, Samson-Couterie B, Golib-Dzib J-F,
317–355. Taketo MM, Tissier F, Bertherat J, Lefrançois- Amar L, Plouin P-F, Sibony M, Lefebvre H, Louiset E,
64. Johannisson E. The foetal adrenal cortex in the Martinez A-M, Martinez A, Val P. Constitutive Jeunemaitre X, Meatchi T, Benecke A, Lalli E,
human. Its ultrastructure at different stages of beta-catenin activation induces adrenal hyperplasia Zennaro M-C. Aldosterone-producing adenoma
development and in different functional states. and promotes adrenal cancer development. Hum formation in the adrenal cortex involves expression
Acta Endocrinol (Copenh). 1968;58(suppl 130):7. Mol Genet. 2010;19(8):1561–1576. of stem/progenitor cell markers. Endocrinology.
65. Ishimoto H, Muench MO, Higuchi T, Minegishi K, 80. Berthon A, Drelon C, Ragazzon B, Boulkroun S, 2011;152(12):4753–4763.
Tanaka M, Yoshimura Y, Jaffe RB. Midkine, a Tissier F, Amar L, Samson-Couterie B, Zennaro M-C, 93. Nishimoto K, Tomlins SA, Kuick R, Cani AK,
heparin-binding growth factor, selectively stimu- Plouin P-F, Skah S, Plateroti M, Lefèbvre H, Sahut- Giordano TJ, Hovelson DH, Liu C-J, Sanjanwala AR,
lates proliferation of definitive zone cells of the Barnola I, Batisse-Lignier M, Assié G, Lefrançois- Edwards MA, Gomez-Sanchez CE, Nanba K, Rainey
human fetal adrenal gland. J Clin Endocrinol Metab. Martinez A-M, Bertherat J, Martinez A, Val P. WE. Aldosterone-stimulating somatic gene muta-
2006;91(10):4050–4056.
WNT/b-catenin signalling is activated in aldosterone- tions are common in normal adrenal glands. Proc
66. Mesiano S, Coulter CL, Jaffe RB. Localization of
producing adenomas and controls aldosterone pro- Natl Acad Sci USA. 2015;112(33):E4591–E4599.
cytochrome P450 cholesterol side-chain cleavage,
duction. Hum Mol Genet. 2014;23(4):889–905. 94. Guagliardo NA, Yao J, Hu C, Barrett PQ. Minireview:
cytochrome P450 17 alpha-hydroxylase/17, 20-lyase,
81. Boulkroun S, Samson-Couterie B, Dzib JF, Lefebvre aldosterone biosynthesis: electrically gated for our
and 3 beta-hydroxysteroid dehydrogenase isom-
H, Louiset E, Amar L, Plouin PF, Lalli E, Jeunemaitre protection. Endocrinology. 2012;153(8):3579–3586.
erase steroidogenic enzymes in human and rhesus
X, Benecke A, Meatchi T, Zennaro MC. Adrenal 95. Hattangady NG, Olala LO, Bollag WB, Rainey WE.
monkey fetal adrenal glands: reappraisal of func-
cortex remodeling and functional zona glomerulosa Acute and chronic regulation of aldosterone pro-
tional zonation. J Clin Endocrinol Metab. 1993;77(5):
hyperplasia in primary aldosteronism. Hypertension. duction. Mol Cell Endocrinol. 2012;350(2):151–162.
1184–1189.
2010;56(5):885–892. 96. Barrett PQ, Lu HK, Colbran R, Czernik A, Pancrazio JJ.
67. McNutt NS, Jones AL. Observations on the ultra-
82. Teebken OE, Scheumann GF. Differentiated corti- Stimulation of unitary T-type Ca(2+) channel
structure of cytodifferentiation in the human fetal
costeroid production and regeneration after se- currents by calmodulin-dependent protein kinase II.
adrenal cortex. Lab Invest. 1970;22(6):513–527.
lective transplantation of cultured and noncultured Am J Physiol Cell Physiol. 2000;279(6):C1694–C1703.
68. Sucheston ME, Cannon MS. Development of
adrenocortical cells in the adrenalectomized rat. 97. Fern RJ, Hahm MS, Lu HK, Liu LP, Gorelick FS, Barrett
zonular patterns in the human adrenal gland.
J Morphol. 1968;126(4):477–491. Transplantation. 2000;70(5):836–843. PQ. Ca2+/calmodulin-dependent protein kinase II
69. Wotus C, Levay-Young BK, Rogers LM, Gomez- 83. Tamura A, Nishimoto K, Seki T, Matsuzawa Y, Saito activation and regulation of adrenal glomerulosa
Sanchez CE, Engeland WC. Development of adre- J, Omura M, Gomez-Sanchez CE, Makita K, Matsui Ca2+ signaling. Am J Physiol. 1995;269(6 Pt 2):
nal zonation in fetal rats defined by expression of S, Moriya N, Inoue A, Nagata M, Sasano H, F751–F760.
aldosterone synthase and 11beta-hydroxylase. En- Nakamura Y, Yamazaki Y, Kabe Y, Mukai K, Kosaka 98. Nogueira EF, Rainey WE. Regulation of aldosterone
docrinology. 1998;139(10):4397–4403. T, Oya M, Suematsu S, Nishikawa T. Somatic synthase by activator transcription factor/cAMP
70. Mitani F, Mukai K, Miyamoto H, Ishimura Y. Lo- KCNJ5 mutation occurring early in adrenal de- response element-binding protein family mem-
calization of replicating cells in rat adrenal cortex velopment may cause a novel form of juvenile bers. Endocrinology. 2010;151(3):1060–1070.
during the late gestational and early postnatal primary aldosteronism. Mol Cell Endocrinol. 2017; 99. Romero DG, Plonczynski MW, Welsh BL, Gomez-
stages. Endocr Res. 1998;24(3-4):983–986. 441:134–139. Sanchez CE, Zhou MY, Gomez-Sanchez EP. Gene
71. Narasaka T, Suzuki T, Moriya T, Sasano H. Temporal 84. Martinerie L, Munier M, Le Menuet D, Meduri G, expression profile in rat adrenal zona glomerulosa
and spatial distribution of corticosteroidogenic en- Viengchareun S, Lombès M. The mineralocorticoid cells stimulated with aldosterone secretagogues.
zymes immunoreactivity in developing human ad- signaling pathway throughout development: ex- Physiol Genomics. 2007;32(1):117–127.
renal. Mol Cell Endocrinol. 2001;174(1–2):111–120. pression, regulation and pathophysiological impli- 100. Breault L, Lehoux JG, Gallo-Payet N. Angiotensin II
72. Naccache A, Louiset E, Duparc C, Laquerrière A, cations. Biochimie. 2013;95(2):148–157. receptors in the human adrenal gland. Endocr Res.
Patrier S, Renouf S, Gomez-Sanchez CE, Mukai K, 85. Martinerie L, Pussard E, Foix-L’Hélias L, Petit F, 1996;22(4):355–361.
Lefebvre H, Castanet M. Temporal and spatial Cosson C, Boileau P, Lombès M. Physiological partial 101. Vanderriele P, Caroccia B, Lenzini L, Gioco F, Fassina
distribution of mast cells and steroidogenic en- aldosterone resistance in human newborns. Pediatr A, Seccia TM, Rossi GPD. Angiotensin-(1–7) peptide
zymes in the human fetal adrenal. Mol Cell Endo- Res. 2009;66(3):323–328. and Mas receptor and angiotensin II–type 2 re-
crinol. 2016;434:69–80. 86. Martinerie L, Viengchareun S, Delezoide A-L, Jaubert ceptor are involved in aldosterone and cortisol
73. Freedman BD, Kempna PB, Carlone DL, Shah M, F, Sinico M, Prevot S, Boileau P, Meduri G, Lombès production in human adrenocortical cell line (NCI-
Guagliardo NA, Barrett PQ, Gomez-Sanchez CE, M. Low renal mineralocorticoid receptor expression H295). Acta Physiol. 2016;217:39.
Majzoub JA, Breault DT. Adrenocortical zonation at birth contributes to partial aldosterone re- 102. Giuliani L, Lenzini L, Antonello M, Aldighieri E,
results from lineage conversion of differentiated sistance in neonates. Endocrinology. 2009;150(9): Belloni AS, Fassina A, Gomez-Sanchez C, Rossi GP.
zona glomerulosa cells. Dev Cell. 2013;26(6): 4414–4424. Expression and functional role of urotensin-II and its
666–673. 87. Martinerie L, Pussard E, Yousef N, Cosson C, Lema I, receptor in the adrenal cortex and medulla: novel
74. Vidal V, Sacco S, Rocha AS, da Silva F, Panzolini C, Husseini K, Mur S, Lombès M, Boileau P. insights for the pathophysiology of primary aldo-
Dumontet T, Doan TMP, Shan J, Rak-Raszewska A, Aldosterone-signaling defect exacerbates sodium steronism. J Clin Endocrinol Metab. 2009;94(2):
Bird T, Vainio S, Martinez A, Schedl A. The adrenal wasting in very preterm neonates: the Premaldo 684–690.

1052 Seccia et al Biology of Zona Glomerulosa and APA Endocrine Reviews, December 2018, 39(6):1029–1056
REVIEW

103. Yao J, McHedlishvili D, McIntire WE, Guagliardo NA, receptors including for glucose-dependent insuli- 131. Spiessberger B, Bernhard D, Herrmann S, Feil S,
Erisir A, Coburn CA, Santarelli VP, Bayliss DA, Barrett notropic peptide in a patient with an aldoster- Werner C, Luppa PB, Hofmann F. cGMP-dependent
PQ. Functional TASK-3-like channels in mito- onoma. J Clin Endocrinol Metab. 2009;94(3):750–756. protein kinase II and aldosterone secretion. FEBS J.
chondria of aldosterone-producing zona glomer- 117. Lefebvre H, Cartier D, Duparc C, Lihrmann I, 2009;276(4):1007–1013.
ulosa cells. Hypertension. 2017;70(2):347–356. Contesse V, Delarue C, Godin M, Fischmeister R, 132. Maniero C, Fassina A, Seccia TM, Toniato A,
104. Davies LA, Hu C, Guagliardo NA, Sen N, Chen X, Vaudry H, Kuhn J-M. Characterization of serotonin(4) Iacobone M, Plebani M, De Caro R, Calò LA, Pessina
Talley EM, Carey RM, Bayliss DA, Barrett PQ. TASK receptors in adrenocortical aldosterone-producing AC, Rossi GP. Mild hyperparathyroidism: a novel
channel deletion in mice causes primary hyper- adenomas: in vivo and in vitro studies. J Clin Endo- surgically correctable feature of primary aldoste-
aldosteronism [published correction appears in crinol Metab. 2002;87(3):1211–1216. ronism. J Hypertens. 2012;30(2):390–395.
Proc Natl Acad Sci USA. 2008;105(36):13696]. Proc 118. Caroccia B, Menegolo M, Seccia TM, Petrelli L, 133. Tomaschitz A, Ritz E, Pieske B, Fahrleitner-Pammer
Natl Acad Sci USA. 2008;105(6):2203–2208. Antonello M, Limena A, Porzionato A, De Caro R, A, Kienreich K, Horina JH, Drechsler C, März W,
105. Penton D, Bandulik S, Schweda F, Haubs S, Tauber P, Poglitsch M, Rossi GP. Urotensin II exerts pressor Ofner M, Pieber TR, Pilz S. Aldosterone and para-

Downloaded from https://academic.oup.com/edrv/article-abstract/39/6/1029/5051322 by Research 4 Life user on 22 November 2018


Reichold M, Cong LD, El Wakil A, Budde T, Lesage F, effects by stimulating renin and aldosterone syn- thyroid hormone: a precarious couple for cardio-
Lalli E, Zennaro MC, Warth R, Barhanin J. Task3 thase gene expression. Sci Rep. 2017;7(1):13876. vascular disease. Cardiovasc Res. 2012;94(1):10–19.
potassium channel gene invalidation causes low 119. Caroccia B, Seccia TM, Campos AG, Gioco F, 134. Rossi GP. Hyperparathyroidism, arterial hyperten-
renin and salt-sensitive arterial hypertension. En- Kuppusamy M, Ceolotto G, Guerzoni E, Simonato F, sion and aortic stiffness: a possible bidirectional link
docrinology. 2012;153(10):4740–4748. Mareso S, Lenzini L, Fassina A, Rossi GP. GPER-1 and between the adrenal cortex and the parathyroid
106. Heitzmann D, Derand R, Jungbauer S, Bandulik S, estrogen receptor-b ligands modulate aldosterone glands that causes vascular damage? Hypertens Res.
Sterner C, Schweda F, El Wakil A, Lalli E, Guy N, synthesis. Endocrinology. 2014;155(11):4296–4304. 2011;34(3):286–288.
Mengual R, Reichold M, Tegtmeier I, Bendahhou S, 120. Caroccia B, Seccia TM, Barton M, Rossi GP. Estrogen 135. Spät A, Fülöp L, Szanda G. The role of mitochondrial
Gomez-Sanchez CE, Aller MI, Wisden W, Weber A, signaling in the adrenal cortex: implications for Ca(2+) and NAD(P)H in the control of aldosterone
Lesage F, Warth R, Barhanin J. Invalidation of TASK1 blood pressure sex differences. Hypertension. 2016; secretion. Cell Calcium. 2012;52(1):64–72.
potassium channels disrupts adrenal gland zonation 68(4):840–848. 136. Spät A, Szanda G. Special features of mitochondrial
and mineralocorticoid homeostasis. EMBO J. 2008; 121. Carey RM, Thorner MO, Ortt EM. Dopaminergic Ca2+ signalling in adrenal glomerulosa cells. Pflugers
27(1):179–187. inhibition of metoclopramide-induced aldosterone Arch. 2012;464(1):43–50.
107. Bayliss DA, Barrett PQ. Emerging roles for two-pore- secretion in man. Dissociation of responses to 137. Rossier MF. T-type calcium channel: a privileged
domain potassium channels and their potential dopamine and bromocriptine. J Clin Invest. 1980; gate for calcium entry and control of adrenal ste-
therapeutic impact. Trends Pharmacol Sci. 2008; 66(1):10–18. roidogenesis. Front Endocrinol (Lausanne). 2016;
29(11):566–575. 122. Seccia TM, Miotto D, De Toni R, Gallina V, Vincenzi 7(May):43.
108. Gallo-Payet N. 60 years of POMC: adrenal and extra- M, Pessina AC, Rossi GP. Subtyping of primary al- 138. Beuschlein F, Boulkroun S, Osswald A, Wieland T,
adrenal functions of ACTH. J Mol Endocrinol. 2016; dosteronism by adrenal vein sampling: effect of Nielsen HN, Lichtenauer UD, Penton D, Schack VR,
56(4):T135–T156. acute D(2) receptor dopaminergic blockade on Amar L, Fischer E, Walther A, Tauber P,
109. Yamakita N, Gomez-Sanchez CE, Mune T, Yoshida adrenal vein cortisol and chromogranin A levels. Eur Schwarzmayr T, Diener S, Graf E, Allolio B, Samson-
H, Miyazaki S, Yasuda K, Nakai T. Regulation of 18- J Endocrinol. 2011;165(1):85–90. Couterie B, Benecke A, Quinkler M, Fallo F, Plouin P-
oxocortisol and 18-hydroxycortisol by the renin- 123. Missale C, Memo M, Liberini P, Spano P. Dopamine F, Mantero F, Meitinger T, Mulatero P, Jeunemaitre
angiotensin system and ACTH in man. J Steroid selectively inhibits angiotensin II–induced aldoste- X, Warth R, Vilsen B, Zennaro M-C, Strom TM,
Biochem Mol Biol. 1993;46(3):395–399. rone secretion by interacting with D-2 receptors. Reincke M. Somatic mutations in ATP1A1 and
110. Gomez-Sanchez CE, Clore JN, Estep HL, Watlington J Pharmacol Exp Ther. 1988;246(3):1137–1143. ATP2B3 lead to aldosterone-producing adenomas
CO. Effect of chronic adrenocorticotropin stimu- 124. Drake CR Jr, Ragsdale NV, Kaiser DL, Carey RM. and secondary hypertension. Nat Genet. 2013;45(4):
lation on the excretion of 18-hydroxycortisol and Dopaminergic suppression of angiotensin II– 440–444, e1–e2.
18-oxocortisol. J Clin Endocrinol Metab. 1988;67(2): induced aldosterone secretion in man: differential 139. Scholl UI, Goh G, Stölting G, de Oliveira RC, Choi M,
322–326. responses during sodium loading and depletion. Overton JD, Fonseca AL, Korah R, Starker LF,
111. Lenders JWM, Williams TA, Reincke M, Gomez- Metabolism. 1984;33(8):696–702. Kunstman JW, Prasad ML, Hartung EA, Mauras N,
Sanchez CE. Diagnosis of endocrine disease: 18- 125. Malchoff CD, Hughes J, Sen S, Jackson S, Carey RM. Benson MR, Brady T, Shapiro JR, Loring E, Nelson-
oxocortisol and 18-hydroxycortisol: is there clinical Dopamine inhibits the aldosterone response to Williams C, Libutti SK, Mane S, Hellman P, Westin G,
utility of these steroids? Eur J Endocrinol. 2018; upright posture. J Clin Endocrinol Metab. 1986;63(1): Åkerström G, Björklund P, Carling T, Fahlke C,
178(1):R1–R9. 197–201. Hidalgo P, Lifton RP. Somatic and germline CAC-
112. Bello-Reuss E, Ernest S, Holland OB, Hellmich MR. 126. Pivonello R, Ferone D, de Herder WW, de Krijger RR, NA1D calcium channel mutations in aldosterone-
Role of multidrug resistance P-glycoprotein in the Waaijers M, Mooij DM, van Koetsveld PM, Barreca producing adenomas and primary aldosteronism.
secretion of aldosterone by human adrenal NCI- A, De Caro ML del B, Lombardi G, Colao A, Nat Genet. 2013;45(9):1050–1054.
H295 cells. Am J Physiol Cell Physiol. 2000;278(6): Lamberts SWJ, Hofland LJ. Dopamine receptor 140. Scholl UI, Stölting G, Nelson-Williams C, Vichot AA,
C1256–C1265. expression and function in human normal adrenal Choi M, Loring E, Prasad ML, Goh G, Carling T,
113. Rossi GP, Andreis PG, Colonna S, Albertin G, gland and adrenal tumors. J Clin Endocrinol Metab. Juhlin CC, Quack I, Rump LC, Thiel A, Lande M,
Aragona F, Belloni AS, Nussdorfer GG. Endothelin- 2004;89(9):4493–4502. Frazier BG, Rasoulpour M, Bowlin DL, Sethna CB,
1[1-31]: a novel autocrine-paracrine regulator of 127. Maniero C, Garg S, Zhao W, Johnson TI, Zhou J, Trachtman H, Fahlke C, Lifton RP. Recurrent gain of
human adrenal cortex secretion and growth. J Clin Gurnell M, Brown MJ. NEFM (neurofilament me- function mutation in calcium channel CACNA1H
Endocrinol Metab. 2002;87(1):322–328. dium) polypeptide, a marker for zona glomerulosa causes early-onset hypertension with primary al-
114. Rossi GP, Albertin G, Neri G, Andreis PG, Hofmann cells in human adrenal, inhibits D1R (dopamine D1 dosteronism. eLife. 2015;4:e06315.
S, Pessina AC, Nussdorfer GG. Endothelin-1 stim- receptor)-mediated secretion of aldosterone. Hy- 141. Gallo-Payet N, Battista MC. Steroidogenesis-adrenal
ulates steroid secretion of human adrenocortical pertension. 2017;70(2):357–364. cell signal transduction. Compr Physiol. 2014;4(3):
cells ex vivo via both ETA and ETB receptor 128. Anderson JV, Struthers AD, Payne NN, Slater JD, 889–964.
subtypes. J Clin Endocrinol Metab. 1997;82(10): Bloom SR. Atrial natriuretic peptide inhibits the 142. Seccia TM, Caroccia B, Gomez-Sanchez EP,
3445–3449. aldosterone response to angiotensin II in man. Clin Vanderriele P-E, Gomez-Sanchez CE, Rossi GP. Re-
115. Duparc C, Moreau L, Dzib JFG, Boyer H-G, Tetsi Sci (Lond). 1986;70(5):507–512. view of markers of zona glomerulosa and aldosterone-
Nomigni M, Boutelet I, Boulkroun S, Mukai K, 129. Bodart V, Rainey WE, Fournier A, Ong H, De Léan A. producing adenoma cells. Hypertension. 2017;70(5):
Benecke AG, Amar L, Gobet F, Meatchi T, Plouin P- The H295R human adrenocortical cell line contains 867–874.
F, Zennaro M-C, Louiset E, Lefebvre H. Mast cell functional atrial natriuretic peptide receptors that 143. Lenzini L, Prisco S, Caroccia B, Rossi GP. Saga of
hyperplasia is associated with aldosterone hyper- inhibit aldosterone biosynthesis. Mol Cell Endocrinol. familial hyperaldosteronism: yet a new channel.
secretion in a subset of aldosterone-producing 1996;118(1–2):137–144. Hypertension. 2018;71(6):1010–1014.
adenomas. J Clin Endocrinol Metab. 2015;100(4): 130. Zhao D, Vellaichamy E, Somanna NK, Pandey KN. 144. Taguchi R, Yamada M, Nakajima Y, Satoh T,
E550–E560. Guanylyl cyclase/natriuretic peptide receptor–A Hashimoto K, Shibusawa N, Ozawa A, Okada S,
116. Lampron A, Bourdeau I, Oble S, Godbout A, gene disruption causes increased adrenal angio- Rokutanda N, Takata D, Koibuchi Y, Horiguchi J,
Schürch W, Arjane P, Hamet P, Lacroix A. Regula- tensin II and aldosterone levels. Am J Physiol Renal Oyama T, Takeyoshi I, Mori M. Expression and
tion of aldosterone secretion by several aberrant Physiol. 2007;293(1):F121–F127. mutations of KCNJ5 mRNA in Japanese patients

doi: 10.1210/er.2018-00060 https://academic.oup.com/edrv 1053


REVIEW

with aldosterone-producing adenomas. J Clin image-negative hyperaldosteronism. J Clin Endo- and adrenal tumors occurring in a single French
Endocrinol Metab. 2012;97(4):1311–1319. crinol Metab. 2017;102(4):1182–1192. pedigree. J Clin Invest. 1995;96(5):2236–2246.
145. Kuppusamy M, Caroccia B, Stindl J, Bandulik S, 157. Åkerström T, Carling T, Beuschlein F, Hellman P. 173. Reimer EN, Walenda G, Seidel E, Scholl UI. CAC-
Lenzini L, Gioco F, Fishman V, Zanotti G, Gomez- Genetics of adrenocortical tumours. J Intern Med. NA1H(M1549V) mutant calcium channel causes
Sanchez C, Bader M, Warth R, Rossi GP. A novel 2016;280(6):540–550. autonomous aldosterone production in HAC15
KCNJ5-insT149 somatic mutation close to, but 158. Nanba K, Chen AX, Omata K, Vinco M, Giordano TJ, cells and is inhibited by mibefradil. Endocrinology.
outside, the selectivity filter causes resistant hy- Else T, Hammer GD, Tomlins SA, Rainey WE. 2016;157(8):3016–3022.
pertension by loss of selectivity for potassium. J Clin Molecular heterogeneity in aldosterone-producing 174. Stratton MR, Campbell PJ, Futreal PA. The cancer
Endocrinol Metab. 2014;99(9):E1765–E1773. adenomas. J Clin Endocrinol Metab. 2016;101(3): genome. Nature. 2009;458(7239):719–724.
146. Azizan EAB, Lam BYH, Newhouse SJ, Zhou J, Kuc RE, 999–1007. 175. Mulatero P, Tauber P, Zennaro M-C, Monticone S,
Clarke J, Happerfield L, Marker A, Hoffman GJ, 159. Nanba K, Omata K, Tomlins SA, Giordano TJ, Lang K, Beuschlein F, Fischer E, Tizzani D, Pallauf A,
Brown MJ. Microarray, qPCR, and KCNJ5 se- Hammer GD, Rainey WE, Else T. Double adreno- Viola A, Amar L, Williams TA, Strom TM, Graf E,

Downloaded from https://academic.oup.com/edrv/article-abstract/39/6/1029/5051322 by Research 4 Life user on 22 November 2018


quencing of aldosterone-producing adenomas re- cortical adenomas harboring independent KCNJ5 Bandulik S, Penton D, Plouin P-F, Warth R, Allolio B,
veal differences in genotype and phenotype and PRKACA somatic mutations. Eur J Endocrinol. Jeunemaitre X, Veglio F, Reincke M. KCNJ5 muta-
between zona glomerulosa– and zona fasciculata– 2016;175(2):K1–K6. tions in European families with nonglucocorticoid
like tumors. J Clin Endocrinol Metab. 2012;97(5): 160. Rossi GP, Ceolotto G, Caroccia B, Lenzini L. Genetic remediable familial hyperaldosteronism. Hyperten-
E819–E829. screening in arterial hypertension. Nat Rev Endo- sion. 2012;59(2):235–240.
147. Monticone S, Castellano I, Versace K, Lucatello B, crinol. 2017;13(5):289–298. 176. Adachi M, Muroya K, Asakura Y, Sugiyama K,
Veglio F, Gomez-Sanchez CE, Williams TA, Mulatero 161. Azizan EAB, Brown MJ. Novel genetic determinants Homma K, Hasegawa T. Discordant genotype-
P. Immunohistochemical, genetic and clinical of adrenal aldosterone regulation. Curr Opin phenotype correlation in familial hyperaldosteronism
characterization of sporadic aldosterone-producing Endocrinol Diabetes Obes. 2016;23(3):209–217. type III with KCNJ5 gene mutation: a patient report
adenomas. Mol Cell Endocrinol. 2015;411:146–154. 162. Zennaro M-C, Boulkroun S, Fernandes-Rosa F. and review of the literature. Horm Res Paediatr. 2014;
148. Åkerström T, Crona J, Delgado Verdugo A, Starker Genetic causes of functional adrenocortical ade- 82(2):138–142.
LF, Cupisti K, Willenberg HS, Knoefel WT, Saeger W, nomas. Endocr Rev. 2017;38(6):516–537. 177. Daniil G, Fernandes-Rosa FL, Chemin J, Blesneac I,
Feller A, Ip J, Soon P, Anlauf M, Alesina PF, Schmid 163. Jeunemaitre X, Charru A, Pascoe L, Guyene TT, Beltrand J, Polak M, Jeunemaitre X, Boulkroun S,
KW, Decaussin M, Levillain P, Wängberg B, Peix J-L, Aupetit-Faisant B, Shackleton CH, Schambelan M, Amar L, Strom TM, Lory P, Zennaro M-C. CAC-
Robinson B, Zedenius J, Bäckdahl M, Caramuta S, Plouin PF, Corvol P. [Hyperaldosteronism sensitive NA1H mutations are associated with different
Iwen KA, Botling J, Stålberg P, Kraimps J-L, Dralle H, to dexamethasone with adrenal adenoma. Clinical, forms of primary aldosteronism. EBioMedicine. 2016;
Hellman P, Sidhu S, Westin G, Lehnert H, Walz MK, biological and genetic study]. Presse Med. 1995; 13:225–236.
Åkerström G, Carling T, Choi M, Lifton RP, 24(27):1243–1248. 178. Pinggera A, Mackenroth L, Rump A, Schallner J,
Björklund P. Comprehensive re-sequencing of ad- 164. Funder JW, Carey RM, Mantero F, Murad MH, Beleggia F, Wollnik B, Striessnig J. New gain-of-
renal aldosterone producing lesions reveal three Reincke M, Shibata H, Stowasser M, Young WF Jr. function mutation shows CACNA1D as re-
somatic mutations near the KCNJ5 potassium The management of primary aldosteronism: case currently mutated gene in autism spectrum
channel selectivity filter. PLoS One. 2012;7(7):e41926. detection, diagnosis, and treatment: an Endocrine disorders and epilepsy. Hum Mol Genet. 2017;
149. Dekkers T, ter Meer M, Lenders JWM, Hermus ARM, Society clinical practice guideline. J Clin Endocrinol 26(15):2923–2932.
Schultze Kool L, Langenhuijsen JF, Nishimoto K, Metab. 2016;101(5):1889–1916. 179. Langton PF, Kakugawa S, Vincent J-P. Making,
Ogishima T, Mukai K, Azizan EAB, Tops B, Deinum J, 165. Gordon RD, Stowasser M, Tunny TJ, Klemm SA, exporting, and modulating Wnts. Trends Cell Biol.
Küsters B. Adrenal nodularity and somatic muta- Finn WL, Krek AL. Clinical and pathological diversity 2016;26(10):756–765.
tions in primary aldosteronism: one node is the of primary aldosteronism, including a new familial 180. Heaton JH, Wood MA, Kim AC, Lima LO, Barlaskar
culprit? J Clin Endocrinol Metab. 2014;99(7): variety. Clin Exp Pharmacol Physiol. 1991;18(5): FM, Almeida MQ, Fragoso MCBV, Kuick R, Lerario
E1341–E1351. 283–286. AM, Simon DP, Soares IC, Starnes E, Thomas DG,
150. Teo AE, Garg S, Johnson TI, Zhao W, Zhou J, Gomez- 166. Mulatero P, Williams TA, Monticone S, Veglio F. Is Latronico AC, Giordano TJ, Hammer GD. Progres-
Sanchez CE, Gurnell M, Brown MJ. Physiological and familial hyperaldosteronism underdiagnosed in sion to adrenocortical tumorigenesis in mice and
pathological roles in human adrenal of the hypertensive children? Hypertension. 2011;57(6): humans through insulin-like growth factor 2 and
glomeruli-defining matrix protein NPNT (neph- 1053–1055. b-catenin. Am J Pathol. 2012;181(3):1017–1033.
ronectin). Hypertension. 2017;69(6):1207–1216. 167. Stowasser M, Gordon RD. Familial hyper- 181. Klutstein M, Nejman D, Greenfield R, Cedar H. DNA
151. Oki K, Plonczynski MW, Luis Lam M, Gomez- aldosteronism. J Steroid Biochem Mol Biol. 2001; methylation in cancer and aging. Cancer Res. 2016;
Sanchez EP, Gomez-Sanchez CE. Potassium chan- 78(3):215–229. 76(12):3446–3450.
nel mutant KCNJ5 T158A expression in HAC-15 168. Sukor N, Mulatero P, Gordon RD, So A, Duffy D, 182. Yoshii Y, Oki K, Gomez-Sanchez CE, Ohno H, Itcho
cells increases aldosterone synthesis. Endocrinology. Bertello C, Kelemen L, Jeske Y, Veglio F, Stowasser M. K, Kobuke K, Yoneda M. Hypomethylation of
2012;153(4):1774–1782. Further evidence for linkage of familial hyper- CYP11B2 in aldosterone-producing adenoma. Hy-
152. Omata K, Tomlins SA, Rainey WE. Aldosterone- aldosteronism type II at chromosome 7p22 in pertension. 2016;68(6):1432–1437.
producing cell clusters in normal and pathological Italian as well as Australian and South American 183. Kobuke K, Oki K, Gomez-Sanchez CE, Ohno H,
states. Horm Metab Res. 2017;49(12):951–956. families. J Hypertens. 2008;26(8):1577–1582. Itcho K, Yoshii Y, Yoneda M, Hattori N. Purkinje cell
153. Omata K, Yamazaki Y, Nakamura Y, Anand SK, 169. Lafferty AR, Torpy DJ, Stowasser M, Taymans SE, Lin protein 4 expression is associated with DNA
Barletta JA, Sasano H, Rainey WE, Tomlins SA, JP, Huggard P, Gordon RD, Stratakis CA. A novel methylation status in aldosterone-producing ade-
Vaidya A. Genetic and histopathologic intertumor genetic locus for low renin hypertension: familial noma. J Clin Endocrinol Metab. 2018103(3):965–971.
heterogeneity in primary aldosteronism. J Clin hyperaldosteronism type II maps to chromosome 7 184. Azizan EAB, Poulsen H, Tuluc P, Zhou J, Clausen MV,
Endocrinol Metab. 2017;102(6):1792–1796. (7p22). J Med Genet. 2000;37(11):831–835. Lieb A, Maniero C, Garg S, Bochukova EG, Zhao W,
154. Omata K, Anand SK, Hovelson DH, Liu C-J, 170. Tong A, Liu G, Wang F, Jiang J, Yan Z, Zhang D, Shaikh LH, Brighton CA, Teo AED, Davenport AP,
Yamazaki Y, Nakamura Y, Ito S, Satoh F, Sasano H, Zhang Y, Cai J. A novel phenotype of familial Dekkers T, Tops B, Küsters B, Ceral J, Yeo GSH, Neogi
Rainey WE, Tomlins SA. Aldosterone-producing cell hyperaldosteronism type III: concurrence of aldo- SG, McFarlane I, Rosenfeld N, Marass F, Hadfield J,
clusters frequently harbor somatic mutations and steronism and Cushing’s syndrome. J Clin Endocrinol Margas W, Chaggar K, Solar M, Deinum J, Dolphin
accumulate with age in normal adrenals. J Endocr Metab. 2016;101(11):4290–4297. AC, Farooqi IS, Striessnig J, Nissen P, Brown MJ.
Soc. 2017;1(7):787–799. 171. Charmandari E, Sertedaki A, Kino T, Merakou C, Somatic mutations in ATP1A1 and CACNA1D
155. Fernandes-Rosa FL, Giscos-Douriez I, Amar L, Hoffman DA, Hatch MM, Hurt DE, Lin L, Xekouki P, underlie a common subtype of adrenal hyperten-
Gomez-Sanchez CE, Meatchi T, Boulkroun S, Stratakis CA, Chrousos GP. A novel point mutation sion. Nat Genet. 2013;45(9):1055–1060.
Zennaro MC. Different somatic mutations in in the KCNJ5 gene causing primary hyper- 185. Nakamura Y, Kitada M, Satoh F, Maekawa T,
multinodular adrenals with aldosterone-producing aldosteronism and early-onset autosomal domi- Morimoto R, Yamazaki Y, Ise K, Gomez-Sanchez CE,
adenoma. Hypertension. 2015;66(5):1014–1022. nant hypertension. J Clin Endocrinol Metab. 2012; Ito S, Arai Y, Dezawa M, Sasano H. Intratumoral
156. Yamazaki Y, Nakamura Y, Omata K, Ise K, Tezuka Y, 97(8):E1532–E1539. heterogeneity of steroidogenesis in aldosterone-
Ono Y, Morimoto R, Nozawa Y, Gomez-Sanchez CE, 172. Pascoe L, Jeunemaitre X, Lebrethon MC, Curnow producing adenoma revealed by intensive double-
Tomlins SA, Rainey WE, Ito S, Satoh F, Sasano H. KM, Gomez-Sanchez CE, Gasc JM, Saez JM, Corvol P. and triple-immunostaining for CYP11B2/B1 and
Histopathological classification of cross-sectional Glucocorticoid-suppressible hyperaldosteronism CYP17. Mol Cell Endocrinol. 2016;422:57–63.

1054 Seccia et al Biology of Zona Glomerulosa and APA Endocrine Reviews, December 2018, 39(6):1029–1056
REVIEW

186. Zhou M, Xue D, Foecking MF, Gomez-Sanchez CE. new-onset diabetes mellitus in primary aldoste- inflammation and fibrosis in hypertensive rats.
Stable expression of rat cytochrome P450 11 beta- ronism: a population study over 5 years. J Hypertens. Kidney Int. 2003;63(5):1791–1800.
hydroxylase (CYP11B1) and aldosterone synthase 2017;35(8):1698–1708. 216. Young MJ, Funder JW. Mineralocorticoids, salt,
(CYP11B2) in MA-10 cells. J Steroid Biochem Mol 202. Hundemer GL, Curhan GC, Yozamp N, Wang M, hypertension: effects on the heart. Steroids. 1996;
Biol. 1995;52(6):523–528. Vaidya A. Cardiometabolic outcomes and mortality 61(4):233–235.
187. Gioco F, Seccia TM, Gomez-Sanchez EP, Rossi GP, in medically treated primary aldosteronism: a ret- 217. Seccia TM, Miotto D, Battistel M, Motta R, Barisa M,
Gomez-Sanchez CE. Adrenal histopathology in rospective cohort study. Lancet Diabetes Endocrinol. Maniero C, Pessina AC, Rossi GP. A stress reaction
primary aldosteronism: is it time for a change? 2018;6(1):51–59. affects assessment of selectivity of adrenal venous
Hypertension. 2015;66(4):724–730. 203. Rossi GP, Maiolino G, Flego A, Belfiore A, Bernini G, sampling and of lateralization of aldosterone excess
188. Nishimoto K, Seki T, Kurihara I, Yokota K, Omura M, Fabris B, Ferri C, Giacchetti G, Letizia C, Maccario M, in primary aldosteronism. Eur J Endocrinol. 2012;
Nishikawa T, Shibata H, Kosaka T, Oya M, Suematsu Mallamaci F, Muiesan ML, Mannelli M, Negro A, 166(5):869–875.
M, Mukai K. Case report: nodule development from Palumbo G, Parenti G, Rossi E, Mantero F. Adre- 218. Miotto D, De Toni R, Pitter G, Seccia TM, Motta R,

Downloaded from https://academic.oup.com/edrv/article-abstract/39/6/1029/5051322 by Research 4 Life user on 22 November 2018


subcapsular aldosterone-producing cell clusters nalectomy lowers incident atrial fibrillation in primary Vincenzi M, Feltrin G, Rossi GP. Impact of accessory
causes hyperaldosteronism. J Clin Endocrinol Metab. aldosteronism patients at long term. Hypertension. hepatic veins on adrenal vein sampling for identi-
2016;101(1):6–9. 2018;71(4):585–591. fication of surgically curable primary aldosteronism.
189. Knudson AG Jr. Mutation and cancer: statistical 204. Rossi GP, Sechi LA, Giacchetti G, Ronconi V, Hypertension. 2009;54(4):885–889.
study of retinoblastoma. Proc Natl Acad Sci USA. Strazzullo P, Funder JW. Primary aldosteronism: 219. Omura M, Sasano H, Fujiwara T, Yamaguchi K,
1971;68(4):820–823. cardiovascular, renal and metabolic implications. Nishikawa T. Unique cases of unilateral hyper-
190. Vouillarmet J, Fernandes-Rosa F, Graeppi-Dulac J, Trends Endocrinol Metab. 2008;19(3):88–90. aldosteronemia due to multiple adrenocortical
Lantelme P, Decaussin-Petrucci M, Thivolet C, Peix 205. Rossi GP, Bernini G, Desideri G, Fabris B, Ferri C, micronodules, which can only be detected by se-
J-L, Boulkroun S, Clauser E, Zennaro M-C. Giacchetti G, Letizia C, Maccario M, Mannelli M, lective adrenal venous sampling. Metabolism. 2002;
Aldosterone-producing adenoma with a somatic Matterello M-J, Montemurro D, Palumbo G, Rizzoni 51(3):350–355.
KCNJ5 mutation revealing APC-dependent familial D, Rossi E, Pessina AC, Mantero F; PAPY Study 220. Goh BKP, Tan YH, Chang KTE, Eng PHK, Yip SKH,
adenomatous polyposis. J Clin Endocrinol Metab. Participants. Renal damage in primary aldosteron- Cheng CWS. Primary hyperaldosteronism secondary
2016;101(11):3874–3878. ism: results of the PAPY Study. Hypertension. 2006; to unilateral adrenal hyperplasia: an unusual cause of
191. Yang T, Zhang H-L, Liang Q, Shi Y, Mei Y-A, Barrett 48(2):232–238. surgically correctable hypertension. A review of 30
PQ, Hu C. Small-conductance Ca2+-activated po- 206. Milliez P, Girerd X, Plouin P-F, Blacher J, Safar ME, cases. World J Surg. 2007;31(1):72–79.
tassium channels negatively regulate aldosterone Mourad J-J. Evidence for an increased rate of car- 221. Satoh F, Morimoto R, Ono Y, Iwakura Y, Omata K,
secretion in human adrenocortical cells. Hyperten- diovascular events in patients with primary aldo- Kudo M, Takase K, Seiji K, Sasamoto H, Honma S,
sion. 2016;68(3):785–795. Okuyama M, Yamashita K, Gomez-Sanchez CE,
steronism. J Am Coll Cardiol. 2005;45(8):1243–1248.
192. Barrett PQ, Guagliardo NA, Klein PM, Hu C, Breault Rainey WE, Arai Y, Sasano H, Nakamura Y, Ito S.
207. Seccia TM, Caroccia B, Adler GK, Maiolino G, Cesari
DT, Beenhakker MP. Role of voltage-gated calcium Measurement of peripheral plasma 18-oxocortisol
M, Rossi GP. Arterial hypertension, atrial fibrillation,
channels in the regulation of aldosterone pro- can discriminate unilateral adenoma from bilateral
and hyperaldosteronism: the triple trouble. Hyper-
duction from zona glomerulosa cells of the adrenal diseases in patients with primary aldosteronism.
tension. 2017;69(4):545–550.
cortex. J Physiol. 2016;594(20):5851–5860. Hypertension. 2015;65(5):1096–1102.
208. Rossi GP, Auchus RJ, Brown M, Lenders JWM,
193. Hu C, Rusin CG, Tan Z, Guagliardo NA, Barrett PQ. 222. Mulatero P, di Cella SM, Monticone S, Schiavone D,
Naruse M, Plouin PF, Satoh F, Young WF Jr. An
Zona glomerulosa cells of the mouse adrenal cortex Manzo M, Mengozzi G, Rabbia F, Terzolo M,
expert consensus statement on use of adrenal vein
are intrinsic electrical oscillators. J Clin Invest. 2012; Gomez-Sanchez EP, Gomez-Sanchez CE, Veglio F.
sampling for the subtyping of primary aldoste-
122(6):2046–2053. 18-hydroxycorticosterone, 18-hydroxycortisol, and
ronism. Hypertension. 2014;63(1):151–160.
194. Rossi GP, Seccia TM, Pessina AC. Primary aldos- 18-oxocortisol in the diagnosis of primary aldo-
209. Rossi GP, Ceolotto G, Rossitto G, Seccia TM,
teronism—part I: prevalence, screening, and selec- steronism and its subtypes. J Clin Endocrinol Metab.
Maiolino G, Berton C, Basso D, Plebani M. Prospective
tion of cases for adrenal vein sampling. J Nephrol. 2012;97(3):881–889.
validation of an automated chemiluminescence-
2008;21(4):447–454. 223. Eisenhofer G, Dekkers T, Peitzsch M, Dietz AS,
195. Douma S, Petidis K, Doumas M, Papaefthimiou P, based assay of renin and aldosterone for the work- Bidlingmaier M, Treitl M, Williams TA, Bornstein
Triantafyllou A, Kartali N, Papadopoulos N, up of arterial hypertension. Clin Chem Lab Med. 2016; SR, Haase M, Rump LC, Willenberg HS,
Vogiatzis K, Zamboulis C. Prevalence of primary 54(9):1441–1450. Beuschlein F, Deinum J, Lenders JWM, Reincke
hyperaldosteronism in resistant hypertension: a retro- 210. Rossi GP, Belfiore A, Bernini G, Desideri G, Fabris B, M. Mass spectrometry–based adrenal and pe-
spective observational study. Lancet. 2008;371(9628): Ferri C, Giacchetti G, Letizia C, Maccario M, ripheral venous steroid profiling for subtyping
1921–1926. Mallamaci F, Mannelli M, Palumbo G, Rizzoni D, primary aldosteronism. Clin Chem. 2016;62(3):
196. Olivieri O, Ciacciarelli A, Signorelli D, Pizzolo F, Rossi E, Agabiti-Rosei E, Pessina AC, Mantero F; 514–524.
Guarini P, Pavan C, Corgnati A, Falcone S, Corrocher Primary Aldosteronism Prevalence in Italy Study In- 224. Ceolotto G, Antonelli G, Maiolino G, Cesari M,
R, Micchi A, Cressoni C, Blengio G. Aldosterone to vestigators. Comparison of the captopril and the saline Rossitto G, Bisogni V, Plebani M, Rossi GP.
renin ratio in a primary care setting: the Bussolengo infusion test for excluding aldosterone-producing Androstenedione and 17-a-hydroxyprogesterone
study. J Clin Endocrinol Metab. 2004;89(9):4221–4226. adenoma. Hypertension. 2007;50(2):424–431. are better indicators of adrenal vein sampling se-
197. Abdelhamid S, Blomer R, Hommel G, Haack D, 211. Rossi GP, Seccia TM, Pessina AC. A diagnostic lectivity than cortisol. Hypertension. 2017;70(2):
Lewicka S, Fiegel P, Krumme B. Urinary tetrahy- algorithm—the holy grail of primary aldosteronism. 342–346.
droaldosterone as a screening method for primary Nat Rev Endocrinol. 2011;7(12):697–699. 225. Arlt W, Lang K, Sitch AJ, Dietz AS, Rhayem Y, Bancos
aldosteronism: a comparative study. Am J Hypertens. 212. Maiolino G, Rossitto G, Bisogni V, Cesari M, Seccia I, Feuchtinger A, Chortis V, Gilligan LC, Ludwig P,
2003;16(7):522–530. TM, Plebani M, Rossi GP; PAPY Study Investigators. Riester A, Asbach E, Hughes BA, O’Neil DM,
198. Brown JJ, Chinn RH, Davies DL, Fraser R, Lever AF, Quantitative value of aldosterone–renin ratio for Bidlingmaier M, Tomlinson JW, Hassan-Smith ZK,
Rae RJ, Robertson JIS. Falsely high plasma potassium detection of aldosterone-producing adenoma: the Rees DA, Adolf C, Hahner S, Quinkler M, Dekkers T,
values in patients with hyperaldosteronism. BMJ. Aldosterone–Renin Ratio for Primary Aldosteron- Deinum J, Biehl M, Keevil BG, Shackleton CHL,
1970;2(5700):18–20. ism (AQUARR) Study. J Am Heart Assoc. 2017;6(5): Deeks JJ, Walch AK, Beuschlein F, Reincke M. Steroid
199. Mulatero P, Monticone S, Burrello J, Veglio F, e005574. metabolome analysis reveals prevalent glucocorti-
Williams TA, Funder J. Guidelines for primary al- 213. Rossi GP. Does primary aldosteronism exist in coid excess in primary aldosteronism. JCI Insight.
dosteronism: uptake by primary care physicians in normotensive and mildly hypertensive patients, and 2017;2(8):e93136.
Europe. J Hypertens. 2016;34(11):2253–2257. should we look for it? Hypertens Res. 2011;34(1): 226. Caroccia B, Fassina A, Seccia TM, Recarti C, Petrelli L,
200. Rossi GP, Gioco F, Fassina A, Gomez-Sanchez CE. 43–46. Belloni AS, Pelizzo MR, Rossi GP. Isolation of human
Normoaldosteronemic aldosterone-producing ad- 214. Ito Y, Takeda R, Karashima S, Yamamoto Y, Yoneda adrenocortical aldosterone-producing cells by a
enoma: immunochemical characterization and di- T, Takeda Y. Prevalence of primary aldosteronism novel immunomagnetic beads method. Endocri-
agnostic implications. J Hypertens. 2015;33(12): among prehypertensive and stage 1 hypertensive nology. 2010;151(3):1375–1380.
2546–2549. subjects. Hypertens Res. 2011;34(1):98–102. 227. Heinze B, Fuss CT, Mulatero P, Beuschlein F, Reincke
201. Wu VC, Chueh SJ, Chen L, Chang CH, Hu YH, Lin 215. Blasi ER, Rocha R, Rudolph AE, Blomme EAG, Polly M, Mustafa M, Schirbel A, Deutschbein T, Williams
YH, Wu KD, Yang WS; TAIPAI Study Group. Risk of ML, McMahon EG. Aldosterone/salt induces renal TA, Rhayem Y, Quinkler M, Rayes N, Monticone S,

doi: 10.1210/er.2018-00060 https://academic.oup.com/edrv 1055


REVIEW

Wild V, Gomez-Sanchez CE, Reis A-C, Petersenn S, and from the University of Padova (DOR1625891/16, CYP11A1, cytochrome P450 side-chain cleavage enzyme;
Wester H-J, Kropf S, Fassnacht M, Lang K, Herrmann DOR1670784/16, BIRD163255/16) and by the Founda- CYP11B1, 11b-hydroxylase; CYP11B2, aldosterone synthase;
K, Buck AK, Bluemel C, Hahner S. Targeting CXCR4 tion for Advanced Research in Arterial Hypertension and CYP17, 17a hydroxylase; CYP17A1, 17a-hydroxylase/17,20-
(CXC chemokine receptor type 4) for molecular Cardiovascular Disease (FORICA). C.E.G.-S. was supported lyase; CYP21A2, 21-hydroxylase; DAX1, dosage-sensitive sex
imaging of aldosterone-producing adenoma. Hy- by National Heart, Lung, and Blood Institute Grant R01 reversal–adrenal hypoplasia congenital critical region on the
pertension. 2018;71(2):317–325. HL27255. X chromosome; DHEA, dehydroepiandrosterone; DHEA-S,
228. Kobuke K, Oki K, Gomez-Sanchez CE, Gomez- Correspondence and Reprint Requests: Gian Paolo dehydroepiandrosterone-sulfate; DOC, deoxycorticosterone;
Sanchez EP, Ohno H, Itcho K, Yoshii Y, Yoneda Rossi, MD, FACC, FAHA, Clinica dell’Ipertensione Arteriosa, E, embryonic day; E2, estradiol; ET-1, endothelin-1; FH, familial
M, Hattori N. Calneuron 1 increased Ca2+ in the Department of Medicine-DIMED, University Hospital, Via hyperaldosteronism; GFP, green fluorescent protein; GIRK4, G
endoplasmic reticulum and aldosterone pro- Giustiniani, 2, 35128 Padova PD, Italy. E-mail: gianpaolo.rossi@ protein–activated inward rectifier potassium channel; GPER-
duction in aldosterone-producing adenoma. Hy- unipd.it. 1, G protein–coupled receptor-1; GRA, glucocorticoid-
pertension. 2018;71(1):125–133. Disclosure Summary: The authors have nothing to remediable aldosteronism; HSD3B1, hydroxy-delta-5-steroid

Downloaded from https://academic.oup.com/edrv/article-abstract/39/6/1029/5051322 by Research 4 Life user on 22 November 2018


229. Mammucari C, Raffaello A, Vecellio Reane D, Rizzuto disclose. dehydrogenase; HSD3B2, 3b-hydroxysteroid dehydrogenase-
R. Molecular structure and pathophysiological roles 2; IHA, idiopathic hyperaldosteronism; IP3, inositol 1,4,5-tri-
of the Mitochondrial Calcium Uniporter. Biochim Abbreviations sphosphate; LC-MS/MS, liquid chromatography–tandem
Biophys Acta. 2016;1863(10):2457–2464. 5-HT4, 5-hydroxytryptamine receptor 4; Ang II, angiotensin II; mass spectrometry; NEFM, neurofilament medium poly-
APA, aldosterone-producing adenoma; APCC, aldosterone- peptide; PA, primary aldosteronism; PAPY, Primary Aldo-
producing cell cluster; ARR, aldosterone-to-renin ratio; AT1, steronism Prevalence in Hypertensives; PCP4, Purkinje cell
angiotensin II type 1; AT1AA, autoantibody against type 1 protein 4; PKA, protein kinase A; PRA, plasma renin activity;
Acknowledgments angiotensin II receptor; AVS, adrenal venous sampling; CaMK, SF1, steroidogenic factor-1; StAR, steroidogenic acute regu-
Financial Support: G.P.R. and T.M.S. were supported by calmodulin-dependent protein kinase; CREB, cAMP response latory; TASK, TWIK acid–sensitive potassium; Wnt, wingless-
grants from the Ministry of Health (RF2011-02352318) element binding; CXCR4, CXC chemokine receptor type 4; related integration site.

1056 Seccia et al Biology of Zona Glomerulosa and APA Endocrine Reviews, December 2018, 39(6):1029–1056

Vous aimerez peut-être aussi