You are on page 1of 9

Endocrinology, 2022, Vol. 163, No.

1, 1–9
https://doi.org/10.1210/endocr/bqab240
Mini-Review

Mini-Review

Downloaded from https://academic.oup.com/endo/article/163/1/bqab240/6433012 by SISTEMAS BIBLIOINFORMA S.A user on 31 May 2022


Primary Aldosteronism, Aldosterone, and
Extracellular Vesicles
Cristian  A.  Carvajal,1,2,3,* Alejandra  Tapia-Castillo,1,2,3,* Jorge  A.  Pérez,1,2,3
and Carlos E. Fardella1,2,3
1
Department of Endocrinology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago,
Chile; 2Millennium Institute of Immunology and Immunotherapy (IMII-ICM), Santiago, Chile; and 3Centro
Traslacional de Endocrinología UC (CETREN-UC), Pontificia Universidad Católica de Chile, Santiago, Chile
ORCiD numbers: 0000-0002-0668-412X (C. A. Carvajal); 0000-0002-6081-1468 (A. Tapia-Castillo); 0000-0002-5500-4434 (C. E.
Fardella).

*Authors with equal contribution.

Received: 14 October 2021; Editorial Decision: 15 November 2021; First Published Online: 22 November 2021; Corrected and
Typeset: 16 December 2021.

Abstract 
Primary aldosteronism (PA) is an endocrine related condition leading to arterial hyperten-
sion due to inappropriately high and unregulated aldosterone concentration. Recently,
a broad spectrum of PA has been recognized, which brings new challenges associated
with early identification of this condition that affect renal epithelial and extrarenal tis-
sues. Reports have shown the potential role of extracellular vesicles (EVs) and EV cargo
as novel and complementary biomarkers in diagnosis and prognosis of PA. In vivo and in
vitro studies have identified specific EV surface antigens, EV-proteins, and EV microRNAs
that can be useful to develop novel diagnostic algorithms to detect, confirm, or follow up
the PA. Moreover, the study of EVs in the field of PA provides further insight in the patho-
physiological mechanism of the PA disease.
Key Words: primary aldosteronism, aldosterone, extracellular vesicles, biomarkers

Arterial hypertension (AHT) is a public health problem Primary Aldosteronism


that affects to 1.39 billion people with a prevalence of PA is an endocrine disorder characterized by an inappro-
31.1% worldwide (1) and 27.6% in Chile. Primary al- priately high circulating aldosterone independent of known
dosteronism (PA), the most frequent form of secondary physiological regulators as renin, angiotensin II, potassium,
hypertension (2) is characterized by a higher rate of car- and sodium status (eg, high salt intake) (6). The conse-
diovascular (CV) events than essential hypertension (EH) quences of PA are AHT, hypokalemia, and metabolic alkal-
(3-5). In this respect, the aim of this review is to explore re- osis, along with an increase in plasma volume, suppression
cent literature related to PA, aldosterone, and extracellular of plasma renin, increased potassium excretion (7), and per-
vesicles through a search of articles in PubMed MEDLINE ipheral vascular resistance that results in resistant hyper-
database up to September 2021. tension (8). The main causes of primary aldosteronism are

ISSN Online 1945-7170


© The Author(s) 2021. Published by Oxford University Press on behalf of the Endocrine Society. All rights reserved.
For permissions, please e-mail: journals.permissions@oup.com
https://academic.oup.com/endo   1
2 Endocrinology, 2022, Vol. 163, No. 1

bilateral PA (~60%) (9) and aldosterone-producing aden- Primary Aldosteronism and the
omas (~30%), which lead to an overproduction of aldos- Aldosterone-to-Renin Ratio
terone (10). Less frequent causes include unilateral adrenal
Currently, the Endocrine Society Guidelines recommend
hyperplasia, aldosterone producing carcinoma, and familial
the screening for PA by determining the aldosterone-
hyperaldosteronism (8,11). Retrospective cohort studies
to-renin ratio (ARR) (24-26) in patients with sustained
have shown that PA patients have a higher CV morbidity
BP > 150/100 mmHg, resistant AHT, AHT with hypokal-
(3-5) and mortality rate (12,13) and increased incidence of
emia, AHT with adrenal incidentaloma AHT with sleep

Downloaded from https://academic.oup.com/endo/article/163/1/bqab240/6433012 by SISTEMAS BIBLIOINFORMA S.A user on 31 May 2022


stroke (3,4) and atrial fibrillation (14) than patients with
apnea, AHT and a family history of early onset hyperten-
EH (4,8,15).
sion or cerebrovascular accident at a young age (<40 years),
In 2017, Brown et al (16) proposed that a broader spec-
and all hypertensive first-degree relatives of patients with
trum of renin-independent PA should be clinically rele-
PA. However, other factors could influence the precise re-
vant, extending the suspicion of PA even to normotensive
sult of the ARR assay (eg, posture, age, intrasubject vari-
subjects (17), which turns PA into a subclinical and clinical
ation, method variability, medication, salt intake, sex, and
prevalent pathological condition (15,18-20). Around 8%
population used to establish cutoffs), affecting its correct
of randomly selected subjects (21), and 10% of essential
interpretation (15,24,56,57).
hypertensives have PA (22,23) for which an early identifica-
Aldosterone and renin, due to its low concentration, re-
tion and management of both overt PA and subclinical PA
quire a sensitive immunoassay or liquid chromatography-
are relevant to avoid CV risk (24-26).
tandem mass spectrometry technology to be detected and
quantified. Currently, plasma renin activity (PRA) has been
Effects of Aldosterone in Epithelial and replaced by direct renin concentration (DRC) in auto-
Nonepithelial Tissues mated equipment, and their normal values are different
from plasma activity renin, which change the ARR cutoffs.
In PA, the chronic and unregulated aldosterone-
Although the ARR screening has been used worldwide to
mineralocorticoid receptor (MR) activation increases
identify subjects suspected to have PA, the previous men-
the sodium transport and water reabsorption in renal
tioned factors influencing the ARR could affect the PA case
epithelia (24,27), which leads to an excess of intravas-
detection (24,58). Hence, novel biomarkers and develop-
cular volume that will increase the arterial blood pres-
ment of novel diagnostic algorithms to detect, confirm, or
sure (BP) leading to AHT. Aldosterone binds the MR,
follow up the PA or subclinical PA are required (Fig. 1).
which lead to genomic and nongenomic effects (28,29),
which upregulate the expression and abundance of
SCNN1 (epithelial sodium channel alpha-subunit), Biomarkers for Primary Aldosteronism
ATP1A1 (ATPase Na+/K+ transporting subunit alpha
Several efforts have been done in order to advance in the
1), SGK1 (serum and glucocorticoid regulated kinase 1),
identification of novel biomarkers for PA that support its
NEDD4-2, YWHAZ (14-3-3 zeta/delta protein) genes,
early detection and also other reported effects as the in-
leading to water/sodium reabsorption, K + secretion,
flammation, endothelial dysfunction, renal damage, vas-
and further regulation of BP. Nongenomic rapid effects
cular remodeling (40,41) and oxidative stress (47,48).
activate signal transducers as SGK1, Na-K-ATPase, and
Early “surrogate biomarkers,” have been recommended by
sodium-hydrogen exchanger (NHE) activation (30,31),
American Heart Association to assess the CV risk, such as
and membrane receptors as the G-protein-estrogen re-
high sensitive C-reactive protein, plasminogen inhibitor ac-
ceptor 30 (GPER or GPR30) (32).
tivator-1, matrix metallopeptidase 9, and malondialdehyde
Similar to renal epithelial tissues, the aldosterone-MR
(37,38), as well as free Cystatin-C, osteopontin, atrial
complex also has effects on nonepithelial tissues (33) as
natriuretic peptide, and neutrophil gelatinase-associated
endothelial cells, smooth muscle cells (34), cardiomyocytes
lipocalin (NGAL) (59-61). However, none of these bio-
(35), and even immune system cells (eg, dendritic cells) (36),
markers are currently available in clinical diagnoses for
which leads to vasoconstriction, increase of heart rate, vas-
AHT or PA.
cular remodeling, and even an increase of central nervous
system activity control. In PA, high circulating aldosterone
promotes inflammation (37-39), endothelial damage, vas-
cular remodeling (40,41), fibrosis (37,42-46), and oxidative Small Extracellular Vesicles and Primary
stress (47,48) affecting the heart, vessels, kidneys, immune Aldosteronism
system (36,49-54), and adipose tissue (55)—all processes EVs constitute a widespread group of nanoparticles naturally
that worsen the prognosis of PA and increase the CV risk. released or shed from different tissues and cell types. EVs are
Endocrinology, 2022, Vol. 163, No. 1 3

Downloaded from https://academic.oup.com/endo/article/163/1/bqab240/6433012 by SISTEMAS BIBLIOINFORMA S.A user on 31 May 2022


Figure 1.  Proposed flowchart for primary aldosteronism (PA) detection with extracellular vesicles (EVs) and EV cargo as novel and complementary
biomarkers of PA. PA is currently defined a broader and clinical prevalent pathological condition (16). A positive screening for PA is based in both a
suppressed plasma renin activity and a high aldosterone-to-renin ratio (16,25,26). Possible or subclinical PA can be further assayed by confirmatory
test, and/or through novel and complementary biomarkers for PA. Here, EVs and EV cargo (eg, EV proteins and EV miRNA) are being studied and
suggested as renal and extrarenal biomarker associated to PA (eg, inflammation, fibrosis, endothelial damage, among others). Potential connections
are shown in dashed line.

composed of a lipid bilayer containing transmembrane pro- (65,66). sEVs have a size of 40 to 150  nm and are bio-
teins and enclosing cytoplasmatic soluble elements and are logical signaling vehicles and important regulators of local
typically classified by their cellular biogenesis where exosomes and distant cell-cell communication (63) carrying proteins,
or small EV (sEVs; diameter 40-150 nm) are vesicles gener- lipids, RNA, and microRNAs. All cell types can generate
ated within the endosomal system and multivesicular bodies, EVs, which are released from parent cells into the extracel-
whereas microvesicles, with a diameter up to 1000 nm, ori- lular space and biofluids (as plasma, cerebrovascular fluid,
ginate by an outward budding at the plasma membrane breast milk, urine, and saliva), and reflect the activation
(62,63). sEVs and microvesicles differ in size, lipid com- status and phenotype of the parent cell from which they
position, content, and cellular origin (62). sEVs formation were derived (62,63).
is regulated via the endosomal sorting complex required
for transport (ESCRT) (multiprotein complexes that guide
intracellular cargo) and/or by non–endosomal sorting com- Plasma Extracellular Vesicles and Primary
plex required for transport–related mechanisms, including Aldosteronism
tetraspanins and membrane lipids. Microvesicle formation A recent paper by Burrello et al (67) reported an increased
is calcium-dependent and associated with loss of membrane concentration of circulating EVs in serum of PA patients
asymmetry and disruption of the cellular cytoskeleton (62). when compared with essential hypertensives and attributed
Experimental and clinical studies show that sEVs to an enhanced biological response of the endothelium to
or exosomes are potential biomarkers of disease (64), aldosterone levels, which also influenced the EV effects
including cancer, metabolic disorders, and CV diseases on the endothelial function. Supporting this observation,
4 Endocrinology, 2022, Vol. 163, No. 1

aldosterone has been shown to stimulate EV release in properties (83) and is a member of the lipocalin family.
endothelial cells in vitro (68) and to activate endothelial It acts as the carrier of basic and neutrally charged lipo-
exocytosis (69). However, how aldosterone modifies the philic compounds under normal physiological conditions
EV concentration or EV cargo is currently investigated, as well as in tissue injury, inflammation, or infection (84).
and it could be linked to a direct aldosterone action on AGP1 gene expression is modified by glucocorticoids (85-
the EV shedding or EV biogenesis or an indirect paracrine 87) and mineralocorticoids (88). Moreover, recent prote-
action of aldosterone in tissues involved in the course of omic studies showed that urinary and serum AGP1 may

Downloaded from https://academic.oup.com/endo/article/163/1/bqab240/6433012 by SISTEMAS BIBLIOINFORMA S.A user on 31 May 2022


the PA pathology (70). Beyond their role as potential vas- serve as predictors of therapeutic response and diagnostic
cular biomarkers, EVs directly could regulate endothelial and as a prognostic biomarker for inflammatory diseases
and vascular smooth muscle cell function by influencing (89), such as chronic heart failure (90,91) and some type
production of nitric oxide and reactive oxygen species, of cancer (92). Additionally, in vivo and in vitro studies
proinflammatory signaling, apoptosis, and senescence in cardiomyocytes showed that aldosterone stimulate the
(71). These processes have been implicated in various ex- AGP1 gene expression (88,93), which is inhibited by spir-
perimental models of hypertension, including angiotensin onolactone (an MR antagonist) (94), demonstrating a
II-infused and spontaneously hypertensive rats and in pa- MR-dependent-regulation of AGP1 expression.
tients with EH. (70). Similar to AGP1, NGAL or lipocalin-2 has been also
In 2021, Burrello et al (72) also assessed the potential role found associated with PA (95). NGAL is a glycoprotein
of EVs in aldosterone-related vascular damage by evaluating also belonging to the lipocalin family that was originally
a panel of 37 EV surface antigens. They showed 19 EV sur- isolated from human neutrophils (96) and expressed in
face antigens differentially expressed in patients with PA the kidney and heart (97). NGAL plasma levels are associ-
compared with hypertensives or normotensive controls, ated with mineralocorticoid effects on cardiac remodeling
including markers of activated platelets and endothelial (95). NGAL promoter is activated by the Aldo-MR com-
and immune-inflammatory cells. Further in vitro studies in plex, increasing its transcription (95,98,99). Mice deficient
human endothelial cells incubated with PA-derived EVs in- in NGAL blunts the aldosterone/MR-mediated vascular
creased the expression of AKT1, CALR, CSNK2A1, FN1, fibrosis (95). In humans, circulating NGAL conjugated
and PIK3R1 genes. These data suggest that EVs are potential with matrix metalloproteinase 9 protein (NGAL-MMP9)
biomarkers of vascular inflammation and endothelial dys- are positively correlated with plasma (95) and urine (100)
function in PA, and also that EVs are potential mediators aldosterone levels. We and others have found NGAL in
associated with accelerated organ damage (72). human urinary (101) and plasma exosomes; however, the
role of NGAL in exosomes as local and distant mediator of
inflammation and fibrosis in PA is unknown.
Urinary Extracellular Vesicles and Primary In the same way, a recent study by Ochiai-Homma et al
Aldosteronism found significant correlation between the ARR and pendrin
Urinary EVs (uEVs) have been also recognized as a source levels in uEVs from subjects with PA and a rat model of al-
of diagnostic biomarkers for different pathologies, espe- dosterone excess. These data are consistent with experimental
cially for renal diseases (73). uEVs originate mainly from studies showing the role of pendrin in aldosterone excess and
epithelial cells in the renal tubules, which carry proteins, suggest that pendrin abundance is relevant in PA and is at-
lipids, and RNA, and are protected from RNAses and tenuated by therapeutic interventions in human PA (102).
proteases proper from the urinary tract (73-75), which
have been associated to physiological and pathophysio-
logical processes in the kidney (76,77) both in vivo and MicroRNAs in Extracellular Vesicles
in vitro (78,79). Renal-derived urinary exosomes have Associated to Primary Aldosteronism
been shown to be increased in patients with PA and have Recent literature propose to microRNAs (miRNAs) in
been suggested to be markers of aldosterone induced renal EVs (103-107) as potential biomarkers and regulators
disease (70,73,80,81). We recently performed a prelim- of the cell-cell communication involved in physiological
inary proteomic study in uEVs in a cohort of PA subjects and pathophysiological processes associated to AHT
identifying alpha-1-acid glycoprotein (AGP1), also named (73,108-110), PA (73), and response to sodium intake
orosomucoid 1, as a differentially expressed protein in PA (111). miRNAs are short, noncoding RNA molecules that
(82). AGP1 is mainly synthesized by the liver but can also be are approximately 22 nucleotides in length and modu-
produced extrahepatically, including the kidney and heart late downstream gene expression by posttranscriptional
in inflammatory conditions. AGP1 is a protein associated mechanisms, specifically by binding to the 3′ untrans-
with the acute phase response with immunomodulatory lated region of a target messenger RNA, leading to target
Endocrinology, 2022, Vol. 163, No. 1 5

messenger RNA degradation or translation repression Acknowledgments


(109,112,113). In this respect, human studies and animal Financial Support: This study was supported partially by grants
models highlight some miRNAs present in EVs associated ANID-FONDECYT 1212006 ( to C.A.C.) and 3200646 (to A.T.C.);
with AHT (73,114), PA, or aldosterone metabolism. For CONICYT-FONDEQUIP EQM150023 (to C.A.C.); ANID–Millen-
example, the EV-associated miR-196b-5p has been linked nium Science Initiative Program-IMII P09/016-F, ICN09_016 (to
C.E.F.); SOCHED 2019-09 (to C.A.C.).
to the aldosterone-induced renal fibrosis in mice with
diabetes. EVs rich in miR-196b-5p mediate the crosstalk

Downloaded from https://academic.oup.com/endo/article/163/1/bqab240/6433012 by SISTEMAS BIBLIOINFORMA S.A user on 31 May 2022


between proximal tubule epithelial cells and fibroblasts Additional Information
during the development of renal fibrosis, which might
Correspondence: Cristian A.  Carvajal, PhD, Associate Professor,
be associated with STAT3/SOCO2 signaling pathway Departamento de Endocrinología, Escuela de Medicina, Pontificia
(115). Other miRNA, the uEV-hsa-miR-4516 is involved Universidad Católica de Chile, Diagonal Paraguay 362, Piso 4, San-
in signaling pathways that regulate renal sodium trans- tiago 8330077, Chile. Email: ccarvajm@uc.cl.
porters involved in PA pathophysiology, such as thiazide- Disclosure Statement: The authors have nothing to disclose.
Data Availability: Data sharing is not applicable to this article as
sensitive sodium chloride cotransporter and epithelial
no data sets were generated or analyzed during the current study.
sodium channel (110,116,117). Most interesting, Let-7i
and miR-21 have been found in urine (118) and plasma
exosomes (106) associated with the renin-angiotensin- References
aldosterone system and mediating renal inflammation, fi-
1. Mills KT, Stefanescu A, He J. The global epidemiology of hyper-
brosis (119,120), and CV diseases (119,121,122). Let-7i tension. Nat Rev Nephrol. 2020;16(4):223-237.
also would regulate downstream targets genes related 2. Pappachan JM, Buch HN. Endocrine hypertension: a practical
to inflammation and fibrosis (119,120), such as IL6, approach. Adv Exp Med Biol. 2017;956:215-237.
COL1A1, COL3A1, ELN, MMP1, VIM, FN1, ACTIN, 3. Mulatero P, Monticone S, Bertello C, et al. Long-term cardio-
TGFBR1, TIMP1, and MMP2 (120,123,124). Similarly, and cerebrovascular events in patients with primary aldoster-
miR-21 is an aldosterone-regulated miRNA mediating onism. J Clin Endocrinol Metab. 2013;98(12):4826-4833.
4. Monticone  S, D’Ascenzo  F, Moretti  C, et  al. Cardiovascular
renin-angiotensin-aldosterone system effects in the ad-
events and target organ damage in primary aldosteronism com-
renal gland (108), heart (121,122), inflammation (125),
pared with essential hypertension: a systematic review and
and fibrosis (103,126). miR-21 would affect downstream meta-analysis. Lancet Diabetes Endocrinol. 2018;6(1):41-50.
targets genes as IL-1B, TIAM1, NEDD4, MMP2, MMP9, 5. Hundemer  GL, Curhan  GC, Yozamp  N, Wang  M, Vaidya  A.
TLR4, STAT3, and CCR1, among others. Further re- Cardiometabolic outcomes and mortality in medically treated
search is needed to clarify the role of these EV-miRNA primary aldosteronism: a retrospective cohort study. Lancet
in the pathophysiology of PA and its potential as novel Diabetes Endocrinol. 2018;6(1):51-59.
6. Tomaschitz A, Pilz S, Ritz E, Obermayer-Pietsch B, Pieber TR.
or complementary biomarker of PA.
Aldosterone and arterial hypertension. Nat Rev Endocrinol.
2010;6(2):83-93.
7. Rossi GP. A comprehensive review of the clinical aspects of pri-
Conclusion
mary aldosteronism. Nat Rev Endocrinol. 2011;7(8):485-495.
PA is an endocrine disorder associated to high circulating 8. Dick SM, Queiroz M, Bernardi BL, Dall’Agnol A, Brondani LA,
aldosterone, which increase the arterial BP, inflammation, Silveiro  SP. Update in diagnosis and management of primary
and fibrosis, all effects that are mainly addressed by high aldosteronism. Clin Chem Lab Med. 2018;56(3):360-372.
aldosterone-MR activity. Recently, a broad spectrum of 9. Omata K, Satoh F, Morimoto R, et al. Cellular and genetic causes of
idiopathic hyperaldosteronism. Hypertension. 2018;72(4):874-880.
PA has been recognized, which brings new challenges as-
10. Boulkroun S, Fernandes-Rosa FL, Zennaro MC. Molecular and
sociated with early identification of this condition that
cellular mechanisms of aldosterone producing adenoma devel-
affects renal epithelia and extrarenal tissues. Recent re- opment. Front Endocrinol (Lausanne). 2015;6:95.
ports have shown the potential role of EVs and EV cargo 11. Lenders JWM, Eisenhofer G, Reincke M. Subtyping of patients
as novel and complementary biomarkers in the diagnosis with primary aldosteronism: an update. Horm Metab Res.
and prognosis of PA, supported by in vivo and in vitro 2017;49(12):922-928.
studies that have identified specific EV surface antigens, 12. Reincke  M, Fischer  E, Gerum  S, et  al; German Conn’s

EV proteins (eg, AGP1, Pendrin), and EV-miRNAs that Registry-Else Kröner-Fresenius-Hyperaldosteronism Registry.
Observational study mortality in treated primary aldosteronism:
can be useful to develop novel diagnostic algorithms to
the German Conn’s registry. Hypertension. 2012;60(3):618-624.
detect, confirm, or follow-up the PA and subclinical PA.
13. Meng Z, Dai Z, Huang K, et al. Long-term mortality for patients
Moreover, the study of EVs in the field of PA provides of primary aldosteronism compared with essential hyperten-
further insight in the pathophysiological mechanism of sion: a systematic review and meta-analysis. Front Endocrinol
the PA disease. (Lausanne). 2020;11:121.
6 Endocrinology, 2022, Vol. 163, No. 1

14. Kim  KJ, Hong  N, Yu  MH, et  al. Time-dependent risk of
32. Feldman  RD, Limbird  LE. GPER (GPR30): a nongenomic re-
atrial fibrillation in patients with primary aldosteronism ceptor (GPCR) for steroid hormones with implications for car-
after medical or surgical treatment initiation. Hypertension. diovascular disease and cancer. Annu Rev Pharmacol Toxicol.
2021;77(6):1964-1973. 2017;57:567-584.
15. Mosso L, Carvajal C, González A, et al. Primary aldosteronism 33. Gomez-Sanchez EP. Mineralocorticoid receptors in the brain and
and hypertensive disease. Hypertension. 2003;42(2):161-165. cardiovascular regulation: minority rule? Trends Endocrinol
16. Brown  JM, Robinson-Cohen  C, Luque-Fernandez  MA,
Metab. 2011;22(5):179-187.
et  al. The spectrum of subclinical primary aldosteronism 34.
Brown  NJ. Aldosterone and vascular inflammation.

Downloaded from https://academic.oup.com/endo/article/163/1/bqab240/6433012 by SISTEMAS BIBLIOINFORMA S.A user on 31 May 2022


and incident hypertension: a cohort study. Ann Intern Med. Hypertension. 2008;51(2):161-167.
2017;167(9):630-641. 35. Young  MJ, Adler  GK. Aldosterone, the mineralocorticoid re-
17. Wannachalee  T, Turcu  AF. Primary aldosteronism: a con-
ceptor and mechanisms of cardiovascular disease. Vitam Horm.
tinuum from normotension to hypertension. Curr Cardiol Rep. 2019;109:361-385.
2021;23(8):105. 36. Herrada AA, Contreras FJ, Marini NP, et al. Aldosterone pro-
18. Fardella  CE, Mosso  L, Gómez-Sánchez  C, et  al. Primary
motes autoimmune damage by enhancing Th17-mediated im-
hyperaldosteronism in essential hypertensives: prevalence, munity. J Immunol. 2010;184(1):191-202.
biochemical profile, and molecular biology. J Clin Endocrinol 37. Stehr  CB, Mellado  R, Ocaranza  MP, et  al. Increased levels of
Metab. 2000;85(5):1863-1867. oxidative stress, subclinical inflammation, and myocardial fi-
19. Mosso L, Fardella C, Montero J, et al. [High prevalence of un- brosis markers in primary aldosteronism patients. J Hypertens.
diagnosed primary hyperaldosteronism among patients with es- 2010;28(10):2120-2126.
sential hypertension]. Rev Med Chil. 1999;127(7):800-806. 38. Martinez-Aguayo  A, Carvajal  CA, Campino  C, et  al. Primary
20. Mulatero  P, Stowasser  M, Loh  KC, et  al. Increased diagnosis aldosteronism and its impact on the generation of arterial
of primary aldosteronism, including surgically correctable hypertension, endothelial injury and oxidative stress. J Pediatr
forms, in centers from five continents. J Clin Endocrinol Metab. Endocrinol Metab. 2010;23(4):323-330.
2004;89(3):1045-1050. 39. Bene NC, Alcaide P, Wortis HH, Jaffe IZ. Mineralocorticoid re-
21. Widimský  J, Bruthans  J, Wohlfahrt  P, et  al. Primary aldos-
ceptors in immune cells: emerging role in cardiovascular dis-
teronism in a general population sample. The Czech Post- ease. Steroids. 2014;91:38-45.
MONICA study. Blood Press. 2020;29(3):191-198. 40. Gilbert  KC, Brown  NJ. Aldosterone and inflammation. Curr
22. O’Shea  PM, Griffin  TP, Fitzgibbon  M. Hypertension: the role Opin Endocrinol Diabetes Obes. 2010;17(3):199-204.
of biochemistry in the diagnosis and management. Clin Chim 41. Connell JM, MacKenzie SM, Freel EM, Fraser R, Davies E. A
Acta. 2017;465:131-143. lifetime of aldosterone excess: long-term consequences of al-
23. Funder  JW. Primary aldosteronism: the next five years. Horm tered regulation of aldosterone production for cardiovascular
Metab Res. 2017;49(12):977-983. function. Endocr Rev. 2008;29(2):133-154.
24. Funder  JW, Carey  RM, Mantero  F, et  al. The management of 42. Sechi  LA, Colussi  G, Catena  C. Hyperaldosteronism and left
primary aldosteronism: case detection, diagnosis, and treat- ventricular hypertrophy. Hypertension. 2010;56(3):e26; author
ment: an Endocrine Society clinical practice guideline. J Clin reply e27.
Endocrinol Metab. 2016;101(5):1889-1916. 43. Muñoz-Durango  N, Barake  MF, Letelier  NA, Campino  C,

2 5. Rehan  M, Raizman  JE, Cavalier  E, Don-Wauchope  AC,
Fardella CE, Kalergis AM. Immune system alterations by aldos-
Holmes  DT. Laboratory challenges in primary al- terone during hypertension: from clinical observations to gen-
dosteronism screening and diagnosis. Clin Biochem. omic and non-genomic mechanisms leading to vascular damage.
2015;48(6):377-387. Curr Mol Med. 2013;13(6):1035-1046.
26. Vaidya  A, Mulatero  P, Baudrand  R, Adler  GK. The
44. Stehr  CB, Carvajal  CA, Lacourt  P, et  al. Subclinical endothe-
Expanding spectrum of primary aldosteronism: implica- lial inflammation markers in a family with type I familial
tions for diagnosis, pathogenesis, and treatment. Endocr Rev. hyperaldosteronism caused by a de novo mutation. Rev Med
2018;39(6):1057-1088. Chil. 2008;136(9):1134-1140.
27. Young MJ, Rickard AJ. Mineralocorticoid receptors in the heart: 45. Zhu  X, Manning  RD Jr, Lu  D, et  al. Aldosterone stimulates
lessons from cell-selective transgenic animals. J Endocrinol. superoxide production in macula densa cells. Am J Physiol
2015;224(1):R1-13. Renal Physiol. 2011;301(3):F529-F535.
28. Freel  EM, Connell  JM. Mechanisms of hypertension:
46. Carvajal CA, Herrada AA, Castillo CR, et al. Primary aldoster-
the expanding role of aldosterone. J Am Soc Nephrol. onism can alter peripheral levels of transforming growth factor
2004;15(8):1993-2001. beta and tumor necrosis factor alpha. J Endocrinol Invest.
29. Ferrari  P. The role of 11β-hydroxysteroid dehydrogenase
2009;32(9):759-765.
type 2 in human hypertension. Biochim Biophys Acta. 47. Milliez P, Girerd X, Plouin PF, Blacher J, Safar ME, Mourad JJ.
2010;1802(12):1178-1187. Evidence for an increased rate of cardiovascular events in
30. Hermidorff  MM, de  Assis  LV, Isoldi  MC. Genomic and rapid patients with primary aldosteronism. J Am Coll Cardiol.
effects of aldosterone: what we know and do not know thus far. 2005;45(8):1243-1248.
Heart Fail Rev. 2017;22(1):65-89. 48. Rossi GP, Bernini G, Desideri G, et al; PAPY Study Participants.
31. Zhou ZH, Bubien JK. Nongenomic regulation of ENaC by aldos- Renal damage in primary aldosteronism: results of the PAPY
terone. Am J Physiol Cell Physiol. 2001;281(4):C1118-C1130. Study. Hypertension. 2006;48(2):232-238.
Endocrinology, 2022, Vol. 163, No. 1 7

49. McCurley  A, Jaffe  IZ. Mineralocorticoid receptors in


65. Neves  KB, Rios  FJ, Jones  R, Evans  TRJ, Montezano  AC,

vascular function and disease. Mol Cell Endocrinol. Touyz  RM. Microparticles from vascular endothelial growth
2012;350(2):256-265. factor pathway inhibitor-treated cancer patients mediate endo-
50.
Martín-Fernández  B, Rubio-Navarro  A, Cortegano  I, thelial cell injury. Cardiovasc Res. 2019;115(5):978-988.
et  al. Aldosterone induces renal fibrosis and inflammatory 66. Amabile  N, Guérin  AP, Leroyer  A, et  al. Circulating endothe-
M1-macrophage subtype via mineralocorticoid receptor in rats. lial microparticles are associated with vascular dysfunction
PloS One. 2016;11(1):e0145946. in patients with end-stage renal failure. J Am Soc Nephrol.
51. Usher MG, Duan SZ, Ivaschenko CY, et al. Myeloid mineralo- 2005;16(11):3381-3388.

Downloaded from https://academic.oup.com/endo/article/163/1/bqab240/6433012 by SISTEMAS BIBLIOINFORMA S.A user on 31 May 2022


corticoid receptor controls macrophage polarization and car- 67. Burrello J, Gai C, Tetti M, et al. Characterization and gene ex-
diovascular hypertrophy and remodeling in mice. J Clin Invest. pression analysis of serum-derived extracellular vesicles in pri-
2010;120(9):3350-3364. mary aldosteronism. Hypertension. 2019;74(2):359-367.
52. Shen  ZX, Chen  XQ, Sun  XN, et  al. Mineralocorticoid re-
68. Robertson  S, Romano  A, Dababneh  E, Bursill  C. Abstract

ceptor deficiency in macrophages inhibits atherosclerosis by P002: aldosterone promotes the release of miRNA-containing
affecting foam cell formation and efferocytosis. J Biol Chem. exosomes from endothelial cells, leading to uptake by smooth
2017;292(3):925-935. muscle cells. Hypertension. 2015;66(suppl 1):AP002-AP002.
53. Shen  JZ, Morgan  J, Tesch  GH, et  al. Cardiac tissue injury
69. Jeong  Y, Chaupin  DF, Matsushita  K, et  al. Aldosterone ac-

and remodeling is dependent upon MR regulation of activa- tivates endothelial exocytosis. Proc Natl Acad Sci U S A.
tion pathways in cardiac tissue macrophages. Endocrinology. 2009;106(10):3782-3787.
2016;157(8):3213-3223. 70. Neves  KB, Touyz  RM. Extracellular vesicles as biomarkers

54. Li  C, Zhang  YY, Frieler  RA, et  al. Myeloid mineralocorticoid and biovectors in primary aldosteronism. Hypertension.
receptor deficiency inhibits aortic constriction-induced cardiac 2019;74(2):250-252.
hypertrophy in mice. PloS One. 2014;9(10):e110950. 71. Burger D, Kwart DG, Montezano AC, et al. Microparticles in-
55. Schütten MT, Houben AJ, de Leeuw PW, Stehouwer CD. The link duce cell cycle arrest through redox-sensitive processes in endo-
between adipose tissue renin-angiotensin-aldosterone system thelial cells: implications in vascular senescence. J Am Heart
signaling and obesity-associated hypertension. Physiology Assoc. 2012;1(3):e001842.
(Bethesda). 2017;32(3):197-209. 72. Burrello J, Tetti M, Forestiero V, et al. Characterization of cir-
56. Martinez-Aguayo  A, Aglony  M, Campino  C, et  al. Aldosterone, culating extracellular vesicle surface antigens in patients with
plasma Renin activity, and aldosterone/renin ratio in a normotensive primary aldosteronism. Hypertension. 2021;78(3):726-737.
healthy pediatric population. Hypertension. 2010;56(3):391-396. 73. Barros ER, Carvajal CA. Urinary exosomes and their cargo: po-
57. O’Shea PM, Griffin TP, Denieffe S, Fitzgibbon MC. The aldos- tential biomarkers for mineralocorticoid arterial hypertension?
terone to renin ratio in the diagnosis of primary aldosteronism: Front Endocrinol (Lausanne). 2017;8:230.
Promises and challenges. Int J Clin Pract. 2019;73(7):e13353. 74. Street  JM, Koritzinsky  EH, Glispie  DM, Star  RA, Yuen  PS.

58. Yozamp N, Hundemer GL, Moussa M, et al. Intraindividual vari- Urine exosomes: an emerging trove of biomarkers. Adv Clin
ability of aldosterone concentrations in primary aldosteronism: im- Chem. 2017;78:103-122.
plications for case detection. Hypertension. 2021;77(3):891-899. 75. Corbetta S, Raimondo F, Tedeschi S, et al. Urinary exosomes in
59. Goff  DC Jr, Lloyd-Jones  DM, Bennett  G, et  al; American
the diagnosis of Gitelman and Bartter syndromes. Nephrol Dial
College of Cardiology/American Heart Association Task Force Transplant. 2015;30(4):621-630.
on Practice Guidelines. 2013 ACC/AHA guideline on the assess- 76. Rigalli JP, Barros ER, Sommers V, Bindels RJM, Hoenderop JGJ.
ment of cardiovascular risk: a report of the American College of Novel aspects of extracellular vesicles in the regulation of renal
Cardiology/American Heart Association Task Force on Practice physiological and pathophysiological processes. Front Cell Dev
Guidelines. Circulation. 2014;129(25 suppl 2):S49-S73. Biol. 2020;8:244.
60. Kshirsagar AV, Coresh J, Brancati F, Colindres RE. Ankle bra- 77. Viñas JL, Burger D, Zimpelmann J, et al. Transfer of microRNA-
chial index independently predicts early kidney disease. Ren 486-5p from human endothelial colony forming cell-derived
Fail. 2004;26(4):433-443. exosomes reduces ischemic kidney injury. Kidney Int.
61. Palazzuoli A, Ruocco G, Pellegrini M, et al. Comparison of neutro- 2016;90(6):1238-1250.
phil gelatinase-associated lipocalin versus B-type natriuretic peptide 78. Street  JM, Birkhoff  W, Menzies  RI, Webb  DJ, Bailey  MA,

and cystatin c to predict early acute kidney injury and outcome in pa- Dear JW. Exosomal transmission of functional aquaporin 2 in
tients with acute heart failure. Am J Cardiol. 2015;116(1):104-111. kidney cortical collecting duct cells. J Physiol. 2011;589(Pt 24):
62. Ståhl  AL, Johansson  K, Mossberg  M, Kahn  R, Karpman  D. 6119-6127.
Exosomes and microvesicles in normal physiology, pathophysi- 79. Esteva-Font  C, Wang  X, Ars  E, et  al. Are sodium trans-

ology, and renal diseases. Pediatr Nephrol. 2019;34(1):11-30. porters in urinary exosomes reliable markers of tubular so-
63. van  Niel  G, D’Angelo  G, Raposo  G. Shedding light on the
dium reabsorption in hypertensive patients? Nephron Physiol.
cell biology of extracellular vesicles. Nat Rev Mol Cell Biol. 2010;114(3):p25-p34.
2018;19(4):213-228. 80. van der Lubbe N, Jansen PM, Salih M, et al. The phosphorylated
64. Théry C, Witwer KW, Aikawa E, et al. Minimal information for sodium chloride cotransporter in urinary exosomes is superior
studies of extracellular vesicles 2018 (MISEV2018): a position to prostasin as a marker for aldosteronism. Hypertension.
statement of the International Society for Extracellular Vesicles 2012;60(3):741-748.
and update of the MISEV2014 guidelines. J Extracell Vesicles. 81. Qi Y, Wang X, Rose KL, et al. Activation of the endogenous renin-
2018;7(1):1535750. angiotensin-aldosterone system or aldosterone administration
8 Endocrinology, 2022, Vol. 163, No. 1

increases urinary exosomal sodium channel excretion. J Am Soc 97. Leopold  JA. The central role of neutrophil gelatinase-

Nephrol. 2016;27(2):646-656. associated lipocalin in cardiovascular fibrosis. Hypertension.
82. Barros  ER, Rigalli  JP, Tapia-Castillo  A, et  al. Proteomic pro- 2015;66(1):20-22.
file of urinary extracellular vesicles identifies AGP1 as a po- 98. Kjeldsen  L, Johnsen  AH, Sengeløv  H, Borregaard  N.

tential biomarker of primary aldosteronism. Endocrinology. Isolation and primary structure of NGAL, a novel protein
2021;162(4):bqab032. associated with human neutrophil gelatinase. J Biol Chem.
83. Hochepied T, Berger FG, Baumann H, Libert C. Alpha(1)-acid 1993;268(14):10425-10432.
glycoprotein: an acute phase protein with inflammatory and 99. Xu  SY, Carlson  M, Engström  A, Garcia  R, Peterson  CG,

Downloaded from https://academic.oup.com/endo/article/163/1/bqab240/6433012 by SISTEMAS BIBLIOINFORMA S.A user on 31 May 2022


immunomodulating properties. Cytokine Growth Factor Rev. Venge  P. Purification and characterization of a human
2003;14(1):25-34. neutrophil lipocalin (HNL) from the secondary gran-
84. Lee YS, Choi JW, Hwang I, et al. Adipocytokine orosomucoid ules of human neutrophils. Scand J Clin Lab Invest.
integrates inflammatory and metabolic signals to preserve en- 1994;54(5):365-376.
ergy homeostasis by resolving immoderate inflammation. J Biol 100. Yang  K, Deng  HB, Man  AWC, et  al. Measuring non-

Chem. 2010;285(29):22174-22185. polyaminated lipocalin-2 for cardiometabolic risk assessment.
85. Maiyar  AC, Phu  PT, Huang  AJ, Firestone  GL. Repression of ESC Heart Fail. 2017;4(4):563-575.
glucocorticoid receptor transactivation and DNA binding of a 101. Gonzales  PA, Pisitkun  T, Hoffert  JD, et  al. Large-scale prote-
glucocorticoid response element within the serum/glucocorticoid- omics and phosphoproteomics of urinary exosomes. J Am Soc
inducible protein kinase (sgk) gene promoter by the p53 tumor Nephrol. 2009;20(2):363-379.
suppressor protein. Mol Endocrinol. 1997;11(3):312-329. 102. Ochiai-Homma  F, Kuribayashi-Okuma  E, Tsurutani  Y, et  al.
86. Savoldi  G, Fenaroli  A, Ferrari  F, Rigaud  G, Albertini  A,
Characterization of pendrin in urinary extracellular vesicles in
Di Lorenzo D. The glucocorticoid receptor regulates the binding a rat model of aldosterone excess and in human primary aldos-
of C/EPBbeta on the alpha-1-acid glycoprotein promoter in teronism. Hypertens Res. 2021;44:1557-1567.
vivo. DNA Cell Biol. 1997;16(12):1467-1476. 103. Chen  C, Lu  C, Qian  Y, et  al. Urinary miR-21 as a potential
87. Taguchi  K, Nishi  K, Chuang  VG, Maruyama  T, Otagiri  M.
biomarker of hypertensive kidney injury and fibrosis. Sci Rep.
Molecular aspects of human alpha-1 acid glycoprotein—struc- 2017;7(1):17737.
ture and function. In: Janciauskiene S, ed. Acute Phase Proteins. 104. Decmann A, Nyírö G, Darvasi O, et al. Circulating miRNA ex-
InTech; 2013:139-162. pression profiling in primary aldosteronism. Front Endocrinol
88. Fejes-Tóth G, Náray-Fejes-Tóth A. Early aldosterone-regulated (Lausanne). 2019;10:739.
genes in cardiomyocytes: clues to cardiac remodeling? 105. Cheng  L, Sun  X, Scicluna  BJ, Coleman  BM, Hill  AF.

Endocrinology. 2007;148(4):1502-1510. Characterization and deep sequencing analysis of exosomal
89. Xiao K, Su L, Yan P, et al. α-1-Acid glycoprotein as a biomarker and non-exosomal miRNA in human urine. Kidney Int.
for the early diagnosis and monitoring the prognosis of sepsis. J 2014;86(2):433-444.
Crit Care. 2015;30(4):744-751. 106. Huang  X, Yuan  T, Tschannen  M, et  al. Characterization of
90. Agra  RM, Varela-Román  A, González-Ferreiro  R, et  al.
human plasma-derived exosomal RNAs by deep sequencing.
Orosomucoid as prognosis factor associated with inflamma- BMC Genomics. 2013;14:319.
tion in acute or nutritional status in chronic heart failure. Int J 107. Mensà  E, Guescini  M, Giuliani  A, et  al. Small extra-

Cardiol. 2017;228:488-494. cellular vesicles deliver miR-21 and miR-217 as pro-
91. Engström  G, Stavenow  L, Hedblad  B, et  al. Inflammation-
senescence effectors to endothelial cells. J Extracell Vesicles.
sensitive plasma proteins, diabetes, and mortality and incidence 2020;9(1):1725285.
of myocardial infarction and stroke: a population-based study. 108.
Romero  DG, Plonczynski  MW, Carvajal  CA, Gomez-
Diabetes. 2003;52(2):442-447. Sanchez EP, Gomez-Sanchez CE. Microribonucleic acid-21 in-
92. Li  F, Yu  Z, Chen  P, et  al. The increased excretion of urinary creases aldosterone secretion and proliferation in H295R human
orosomucoid 1 as a useful biomarker for bladder cancer. Am J adrenocortical cells. Endocrinology. 2008;149(5):2477-2483.
Cancer Res. 2016;6(2):331-340. 109. Friso S, Carvajal CA, Fardella CE, Olivieri O. Epigenetics and
93. Messaoudi S, Gravez B, Tarjus A, et al. Aldosterone-specific ac- arterial hypertension: the challenge of emerging evidence.
tivation of cardiomyocyte mineralocorticoid receptor in vivo. Transl Res. 2015;165(1):154-165.
Hypertension. 2013;61(2):361-367. 110. Liu  ZZ, Jose  PA, Yang  J, Zeng  C. Importance of extracel-

94. Gravez  B, Tarjus  A, Jimenez-Canino  R, et  al. The diuretic
lular vesicles in hypertension. Exp Biol Med (Maywood).
torasemide does not prevent aldosterone-mediated mineralo- 2021;246(3):342-353.
corticoid receptor activation in cardiomyocytes. PloS One. 111. Gildea  JJ, Carlson  JM, Schoeffel  CD, Carey  RM, Felder  RA.
2013;8(9):e73737. Urinary exosome miRNome analysis and its applica-
95. Tarjus  A, Martínez-Martínez  E, Amador  C, et  al. Neutrophil tions to salt sensitivity of blood pressure. Clin Biochem.
gelatinase-associated lipocalin, a novel mineralocorticoid 2013;46(12):1131-1134.
biotarget, mediates vascular profibrotic effects of mineralocor- 112. Sun  Y, Koo  S, White  N, et  al. Development of a micro-array
ticoids. Hypertension. 2015;66(1):158-166. to detect human and mouse microRNAs and character-
96. Mao S, Jiang T, Shang G, Wu Z, Zhang N. Increased expres- ization of expression in human organs. Nucleic Acids Res.
sion of neutrophil gelatinase-associated lipocalin receptor by 2004;32(22):e188.
interleukin-1β in human mesangial cells via MAPK/ERK acti- 113. Tian Z, Greene AS, Pietrusz JL, Matus IR, Liang M. MicroRNA-
vation. Int J Mol Med. 2011;27(4):555-560. target pairs in the rat kidney identified by microRNA
Endocrinology, 2022, Vol. 163, No. 1 9

microarray, proteomic, and bioinformatic analysis. Genome 120. Syed  M, Ball  JP, Romero  DG. Let-7i: lethal weapon against
Res. 2008;18(3):404-411. angiotensin ii-induced cardiac injury. Hypertension.
114. Tan  PPS, Hall  D, Chilian  WM, et  al. Exosomal microRNAs 2015;66(4):739-741.
in the development of essential hypertension and its po- 121. Ball  JP, Syed  M, Marañon  RO, et  al. Role and regulation of
tential as biomarkers. Am J Physiol Heart Circ Physiol. MicroRNAs in aldosterone-mediated cardiac injury and dys-
2021;320(4):H1486-H1497. function in male rats. Endocrinology. 2017;158(6):1859-1874.
115. Hu R, Li X, Peng C, et al. miR-196b-5p-enriched extracellular 122. Syed  M, Ball  JP, Mathis  KW, et  al. MicroRNA-21 ab-

vesicles from tubular epithelial cells mediated aldosterone- lation exacerbates aldosterone-mediated cardiac injury,

Downloaded from https://academic.oup.com/endo/article/163/1/bqab240/6433012 by SISTEMAS BIBLIOINFORMA S.A user on 31 May 2022


induced renal fibrosis in mice with diabetes. BMJ Open remodeling, and dysfunction. Am J Physiol Endocrinol Metab.
Diabetes Res Care. 2020;8(1):e001101. 2018;315(6):E1154-E1167.
116. Salih M, Fenton RA, Zietse R, Hoorn EJ. Urinary extracellular 123. Zhang  K, Yang R, Chen  J, et  al. Let-7i-5p regulation of cell
vesicles as markers to assess kidney sodium transport. Curr morphology and migration through distinct signaling path-
Opin Nephrol Hypertens. 2016;25(2):67-72. ways in normal and pathogenic fibroblasts from urethra. Front
117. Gracia  T, Wang  X, Su  Y, et  al. Urinary exosomes contain
Bioeng Biotechnol. 2020;8:428.
MicroRNAs capable of paracrine modulation of tubular trans- 124. Bao  MH, Feng  X, Zhang  YW, Lou  XY, Cheng  Y, Zhou  HH.
porters in kidney. Sci Rep. 2017;7:40601. Let-7 in cardiovascular diseases, heart development and car-
118. Tapia-Castillo A, Guanzon D, Palma C, et al. Downregulation of diovascular differentiation from stem cells. Int J Mol Sci.
exosomal miR-192-5p and miR-204-5p in subjects with nonclassic 2013;14(11):23086-23102.
apparent mineralocorticoid excess. J Transl Med. 2019;17(1):392. 125. Sheedy FJ. Turning 21: Induction of miR-21 as a key switch in
119. Wang X, Wang HX, Li YL, et al. MicroRNA Let-7i negatively the inflammatory response. Front Immunol. 2015;6:19.
regulates cardiac inflammation and fibrosis. Hypertension. 126. Cheng Y, Zhang C. MicroRNA-21 in cardiovascular disease. J
2015;66(4):776-785. Cardiovasc Transl Res. 2010;3(3):251-255.

You might also like