You are on page 1of 7

Disponible en ligne sur

ScienceDirect
www.sciencedirect.com

Annales d’Endocrinologie 79 (2018) 157–163

Klotz Communications 2018: Cortisol and all its disorders

Autoimmune Addison’s disease – An update on pathogenesis


La maladie d’Addison auto-immune – une mise à jour sur la pathogenèse
Alexander Hellesen a,b , Eirik Bratland a,b , Eystein S. Husebye a,b,c,d,∗
a Department of Clinical Science, University of Bergen, 5021 Bergen, Norway
b K.G. Jebsen Senter for Autoimmune Sykdommer, University of Bergen, 5021 Bergen, Norway
c Department of Medicine, Haukeland University Hospital, 5021 Bergen, Norway
d Department of Medicine (Solna), Karolinska Institutet, 17176 Stockholm, Sweden

Abstract
Autoimmunity against the adrenal cortex is the leading cause of Addison’s disease in industrialized countries, with prevalence estimates ranging
from 93–220 per million in Europe. The immune-mediated attack on adrenocortical cells cripples their ability to synthesize vital steroid hormones
and necessitates life-long hormone replacement therapy. The autoimmune disease etiology is multifactorial involving variants in immune genes
and environmental factors. Recently, we have come to appreciate that the adrenocortical cell itself is an active player in the autoimmune process.
Here we summarize the complex interplay between the immune system and the adrenal cortex and highlight unanswered questions and gaps in our
current understanding of the disease.
© 2018 Elsevier Masson SAS. All rights reserved.

Keywords: Addison’s disease; Autoimmunity; Pathogenesis; Adrenalitis

Résumé
L’auto-immunité contre le cortex surrénalien est la principale cause de la maladie d’Addison dans les pays industrialisés, avec des estimations
de prévalence allant de 93 à 220 cas par million d’habitants en Europe. L’attaque immunitaire des cellules corticosurrénales affaiblit leur capacité
à synthétiser des hormones stéroïdes vitales et nécessite un traitement hormonal substitutif définitif. L’étiologie de la maladie auto-immune est
multifactorielle impliquant des polymorphismes dans les gènes immunitaires et des facteurs environnementaux. Récemment, nous avons appris
que la cellule corticosurrénale elle-même joue un rôle actif dans le processus auto-immun. Nous résumons ici l’interaction complexe entre le
système immunitaire et le cortex surrénalien et soulignons les questions et les lacunes sans réponse dans notre compréhension actuelle de la
maladie d’Addison.
© 2018 Elsevier Masson SAS. Tous droits réservés.

Mots clés : Maladie d’Addison ; Glande surrénale ; Auto-immunité

1. Introduction men [1–3]. Patients may present with several diffuse symp-
toms such as fatigue, nausea, dizziness and weight loss, but can
Addison’s disease (AAD), or primary adrenocortical insuf- often be identified by specific signs like salt-craving and, espe-
ficiency, is characterized by inability of the adrenal cortex to cially, hyperpigmentation of the skin and mucosal surfaces [1,4].
produce sufficient amounts of glucocorticoids and mineralo- The latter is caused by increased stimulation of melanocortin
corticoids. The disease often manifests itself between the 2nd receptors in the skin by ACTH and pro-opiomelanocortin (the
and 4th decade of life and affects women more often than precursor for ACTH), that builds up in patients due to lack of
cortisol-mediated feedback inhibition of the HPA-axis [5]. Con-
sequently, elevated plasma ACTH as well as low serum cortisol
∗ Corresponding author. Department of Clinical Science and University of levels serves as diagnostic criteria for AAD.
Bergen, 5021 Bergen, Norway. The current recommended treatment regimen consists of
E-mail address: Eystein.Husebye@uib.no (E.S. Husebye). hormone replacement therapy in the form of hydrocortisone
https://doi.org/10.1016/j.ando.2018.03.008
0003-4266/© 2018 Elsevier Masson SAS. All rights reserved.
158 A. Hellesen et al. / Annales d’Endocrinologie 79 (2018) 157–163

(glucocorticoid replacement) and fludrocortisone (mineralocor- Table 1


ticoid replacement) [6]. If left untreated, a stressful event (e.g. Genes outside the MHC complex associated with autoimmune Addison’s
disease.
watching a high profile premier league football match [7]) may
eventually precipitate acute adrenal insufficiency (adrenal cri- Gene Function References
sis) – a potentially life-threatening condition caused by critical CTLA-4 Inhibits T cell activation [32,96]
hormone-deficiency [8]. PD-L1 Inhibits T cell activation [97]
Although tuberculosis was the predominant cause of AAD PTPN22 Regulates T cell responses to [34,98]
earlier [9], autoimmunity is now responsible for most of the weak peptide agonists; promotes
TLR-induced type I IFN
cases in developed countries [1,4]. The identification of steroid production in myeloid cells
cytochrome P450 21-hydroxylase (21OH) of the adrenal cor- NALP1 Inflammasome activation [99,100]
tex as a major autoantigen has led to routine use of 21OH STAT4 Involved in TH1 and TH17 [101]
autoantibody assays in determining the disease aetiology [6,10]. differentiation
Among Norwegian patients, 86% was found positive for such CIITA Regulator of MHC class II [102,103]
transcription
autoantibodies [1], and similar numbers have been reported in BACH2 Regulates B and T cell [23,37]
other populations [2,4,11]. Autoantibodies against the adrenal differentiation
cortex are often detectable prior to the development of overt FCRL3 Regulates signalling pathways in [104]
disease [12] and in as much as 95% of patients at the time lymphocytes
of diagnosis [13]. Less than 0.5% of the general population GPR174 Involved in suppression of IL2 [105]
production in T cells
harbor anti-21OH antibodies [14] and these “antibody biomark- GATA3 Transcription factor that acts on T [101]
ers” are therefore highly specific for autoimmune AAD. In a cell differentiation
prospective study, the cumulative risk of developing AAD in NFATC1 Involved in gene transcription in [106]
antibody-positive individuals was estimated to about 30% [12]. activated T cells and a major
Although AAD may occur in isolation, more than half of the target of cyclosporine
CLEC16A Modulates autophagy and T cell [103]
patients suffer from additional autoimmune diseases. Autoim- selection in the thymus
mune thyroid disease (AITD; hypo- or hyperthyroidism) is the CYP27B1 Enzyme involved in vitamin D [107–109]
most frequent comorbidity, followed by diseases like type 1 dia- metabolism
betes (T1D), chronic atrophic gastritis and vitiligo [2,11,15,16]. VDR Vitamin D receptor [110]
The coexistence of at least two out of the three diseases AAD,
AITD and T1D is commonly referred to as autoimmune polyen- (MICA) [29], which encodes a ligand for the activating NK and
docrine syndrome type 2 (APS-2) [17]. AAD is also a major T cell receptor NKG2D. Homozygosity for the MICA5.1 allelic
disease component of the monogenic syndrome APS-1 [18,19], variant was associated with increased frequency of progression
in which the presence of two out of the following three manifes- to AAD in 21OH autoantibody-positive individuals bearing the
tations is diagnostic: AAD, chronic mucocutaneous candidiasis high-risk genotype (DR3/DQ2-DR4/DQ8) [30].
and hypoparathyroidisim. While the two syndromes share sev- Several studies have reported SNPs in immune-related genes
eral features such as the presence of serum 21OH autoantibodies that confer increased risk for AAD. Most of these genes encode
[4] and immune infiltration of the adrenal cortex (when AAD is proteins that are active in regulating T and B cell activation
involved) [20,21], the genetic background of APS-1 differs from and differentiation (Table 1, see also [31] for a review of the
APS-2 as the former is caused by mutations in the autoimmune subject), underlining the importance of these cell types in the
regulator (AIRE) gene while APS-2 (including isolated AAD) pathobiology of AAD. A recent meta-analysis of European AAD
is a polygenic disorder. Also, APS-1 is more rare than APS-2, cohorts strengthened the link to CTLA-4 [32], hypothesizing
with an estimated prevalence of 1:90,000 in Norway [22]. that the associated genetic variants may impair CTLA-4 func-
tion and thereby lower the threshold for T cell activation as
2. Genetic susceptibility suggested in AITD [33]. Another interesting association is the
1858T-allele of PTPN22 [34] as recent publications implicate
In agreement with most other organ-specific autoim- PTPN22 in distinct processes that may be of particular impor-
mune disorders, the strongest genetic associations to AAD tance to autoimmunity; regulation of T cell responses to weak
are found within certain HLA class II alleles. Sig- self-peptide agonists [35] and promotion of type I IFN produc-
nificant risk has been confirmed in the presence of tion in myeloid cells following TLR stimulation [36]. Recently,
HLA DR3/DQ2 (DRB1*0301-DQA1*0501-DQB1*0201) and a comprehensive exome sequencing effort of patients from the
DR4/DQ8 (DRB1*0404-DQA1*0301-DQB1*0302) haplo- Swedish Addison Registry revealed BACH2 as a major risk locus
types, and especially the compound heterozygote combination for AAD [23], corroborating another recent finding that a SNP
(odds ratio = 32) [1,23,24]. Also, in a study of families with mul- in BACH2 is associated with increased risk of developing AAD
tiple affected individuals, increased risk for AAD was found to in the UK and in Norway [37]. BACH2 is a transcription factor
be associated with the HLA DR3-B8 haplotype [25]. The impor- that is essential for T and B cell development and variants in the
tance of these alleles has been further reinforced by studies on T BACH2 gene has been associated with increased risk of develop-
cell responses in AAD [25–28]. Within the HLA locus, a connec- ing a wide range of autoimmune diseases, including T1D, AITD
tion has also been made to the MHC-class I chain-related gene A and celiac disease [38–40].
A. Hellesen et al. / Annales d’Endocrinologie 79 (2018) 157–163 159

Another important aspect is that the heritability of AAD


appears to be very high [41], strongly indicating the existence
of highly penetrant AAD susceptibility alleles [31]. The genetic
risk factors described so far can only account for a small part
of the total heritability observed, which justifies large scale
genetic studies on AAD such as genome-wide association stud-
ies (GWAS) or whole genome sequencing.

3. Pathophysiology

The histopathological picture in AAD is characterized by


lymphocytic infiltration of the adrenal cortex and extensive atro-
phy of the parenchyma, while the adrenal medulla remains intact
[20,21,42]. Over the course of the disease, the integrity of the
cortex is gradually lost as the three layers of cells are progres-
sively destroyed and replaced by fibrous tissue [4,43]. Despite
continuous loss of adrenocortical cells, AAD may remain sub-
clinical for long periods before overt disease develops. In fact,
adrenocortical failure may not manifest itself until 90% of the
cells are destroyed [44]. In end-stage disease, the adrenals are
Fig. 1. T cell responses against 21OH protein in patients with AAD. Peripheral
often very small on CT scans and can be difficult to identify on
blood mononuclear cells (PBMC) from patients with AAD and healthy controls
autopsies [4]. Unfortunately, histochemical studies have been (HC) were isolated and stimulated with recombinant purified 21OH or medium
restricted to post-mortem analyses due to practical and ethical only as the negative control. A. Lymphocyte proliferation assay: 2 × 105 PBMC
reasons, and have in general been few and far between. There- were cultured in the presence or absence of 21OH for 7 days in triplicates. [3H]-
fore, a detailed analysis of the mononuclear infiltrate in AAD thymidine was added for the last 16 hours and antigen specific proliferation
was estimated based on genomic uptake of radioactive thymidine. Data are
is lacking. Rather, most work related to the immunopathogen-
reported as (mean counts per minute (cpm) in the presence of 21OH–mean cpm
esis of the disease has been performed on peripheral blood and in the presence of medium only). The positive cut-off value at 1000 cpm is
serum. displayed as a dotted horizontal line. B. IFN␥ Elispot assay: 1 × 106 PBMC
Specific immunological responses towards adrenal antigens were prestimulated with 21OH or medium only for 24 hours, before performing
in patients with AAD were first observed several decades ago. the Elispot assay. Cells were then assayed in triplicates or quadruplicates for 24
additional hours in the Elispot filter wells. Spots were counted in a dissecting
Autoantibodies against adrenal extracts was demonstrated as
microscope and are reported as (mean spot forming units (SFU) per 1 × 106
early as in 1957 [45], and was subsequently shown to be specif- PBMC in the presence of 21OH–mean SFU per 1 × 106 PBMC in the presence
ically directed against the adrenal cortex [46]. The major target of medium only). The positive cut-off value at 20 SFU per 1 × 106 PBMC is
of these adrenal cortex autoantibodies (ACAs) was later iden- displayed as a dotted horizontal line.
tified as 21OH [10], an enzyme involved in the biosynthesis of
steroid hormones in the adrenal cortex. Although 21OH autoan-
tibodies correlate with the degree of adrenal dysfunction in the
preclinical phase of AAD [47], they do not seem to inhibit 21OH
activity in vivo [48]. This is not all that surprising given that cells are detectable in the peripheral blood of AAD patients
21OH is an intracellular protein anchored to the smooth endo- (Fig. 1) [26]. Since then, epitope mapping has revealed two
plasmic reticulum membrane [49]. Evidence for a role for these specific 21OH sequences targeted by CD8+ T cells. Rottem-
antibodies in the disease pathogenesis is in fact very sparse. The bourg et al. showed IFN␥ production in response to a C-terminal
only publication of note that has suggested a functional signif- peptide of 21OH and mapped the response to an octameric
icance role of 21OH autoantibodies was Bratland et al., who sequence (21OH431-438, amino acid sequence: EPLARLEL)
observed a reduced response of patient PBMC to 21OH pro- [28]. The epitope was found to be HLA-B8 restricted, and stain-
tein in stimulation cultures with autologous plasma (containing ing of PBMCs with HLA-B8*21OH431-438 tetramers showed
21OH autoantibodies) when anti-CD32 (Fc␥RII, receptor for higher frequencies of circulating 21OH431-438-specific CD8+
IgG) blocking antibodies were added [26]. T cells in two patients with AAD compared to a HLA-B8-
Only in recent years has 21OH been established as a major T positive control. More recently, an HLA-A2-restricted epitope
cell autoantigen in AAD. Following immunization of BALB/c (21OH342-350, amino acid sequence: LLNATIAEV) was iden-
and SJL mice with recombinant 21OH or 21OH-derived pep- tified [27]. 21OH342-350-specific clones produced IFN␥ upon
tide, Husebye et al. reported proliferation of lymph node cells stimulation with the cognate peptide and were able to lyse tar-
to an immunodominant peptide (21OH342-361) [50]. A few get cells expressing 21OH through the production of granzyme
years later, T cell responses to 21OH whole protein and the pep- B (and possibly other mediators). These reports offer solid evi-
tide 21OH342-361 were demonstrated in patients with AAD, dence for the presence of 21OH-specific autoreactive T cells
providing the first evidence of 21OH as a T cell autoantigen in AAD and pave way for more functional analyses of these
in the disease, and clearly demonstrating that 21OH-specific T disease-specific cells.
160 A. Hellesen et al. / Annales d’Endocrinologie 79 (2018) 157–163

4. Intrinsic factors of the adrenal cortex

The adrenocortical cells themselves are probably active col-


laborators in the pathogenesis of AAD as they communicate
with the immune system in several ways. Adrenocortical cells
respond to inflammation by producing a number of immunolog-
ical mediators [51], including IL-1␤ [52–54], IL-6 [53], IL-8
[55], IL-18 [56], TNF␣ [57] and CXCL10 [58]. Moreover,
they regularly interact with tissue-resident or patrolling den-
dritic cells and macrophages [52,59]. Several toll-like receptors
(TLR)s, recognizing conserved molecular microbial patterns,
are expressed within the adrenal cortex and has been shown to
induce pro-inflammatory cytokines upon stimulation [55], indi-
cating an ability to promote local anti-microbial responses and
inflammation.
Fig. 2. Simplified model of the pathogenesis of autoimmune Addison’s dis-
A central question to the pathogenesis of AAD is what drives ease. Similar to most autoimmune diseases, autoimmune Addison’s disease
the infiltration of mononuclear cells into the adrenal cortex. Sim- (AAD) is believed to be caused by a combination of several unfortunate genetic
ilar to related autoimmune endocrine disorders, we and others variants and environmental factors. Although variants in several genes encod-
have shown that patients with AAD have increased serum lev- ing immunological proteins have been shown to be associated with AAD, the
els of the interferon induced chemokines CXCL9 and CXCL10 majority of the genetic component in AAD pathogenesis remains undefined.
Knowledge of possible environmental factors is sparse, although certain viruses
compared to healthy controls [58,60,61]. Furthermore, cultured are suggested candidates. The recent advent of checkpoint inhibitors in cancer
primary adrenocortical zona fasciculata cells as well as adreno- treatment has revealed that antibodies against PD-1 and maybe also CTLA4
cortical carcinoma cells have the ability to produce CXCL10 can induce AAD. AAD is characterized by a highly targeted immune response
upon stimulation with pro-inflammatory cytokines [58,60], sug- against steroidogenic enzymes in the adrenal cortex, 21OH in particular. Anti-
gesting that these cells may contribute to the elevated serum bodies against 21OH are present in the majority of patients and constitute a great
biomarker for autoimmunity as the underlying cause of adrenal insufficiency.
levels of CXCL10 in patients and potentially to the recruitment However, it is thought that 21OH-specific cytotoxic T cells are responsible for
of self-reactive lymphocytes to the adrenal cortex. the actual destruction of adrenocortical tissue. These T cells are also detectable
in the peripheral blood of the patients. The 21OH-specific T cells are proba-
5. Environmental triggering events bly recruited into the adrenal cortex by chemokines released by adrenocortical
cells themselves (intrinsic factors) as a result of inflammatory or stressful
events. Finally, AAD like many autoimmune diseases are probably permis-
Better insight into the interplay between adrenocortical cells, sive to immunotherapy aiming to inhibit the underlying immunopathological
environmental factors and the immune system should offer mechanisms. For AAD this has been demonstrated with the use of rituximab
valuable cues to the pathogenic mechanisms of AAD (Fig. 2). (anti-CD20 antibodies). Images are adapted from Servier Medical Art by Servier
Very little is currently known about the initial stages of disease (http://www.servier.com/Powerpoint-image-bank) and modified by the authors
development, that is, the establishment of inflammation and an under the following terms: CREATIVE COMMONS Attribution 3.0 Unported
(CC BY 3.0).
immune response against the adrenal cortex. It is widely appreci-
ated that in order for autoimmunity to develop, there is the need
for genetic susceptibility and a precipitating event to convert treatment for chronic hepatitis C virus (HCV) infection [78].
inherent disease susceptibility [62]. Local infections of the tar- In one instance, a woman carrying the HLA high-risk genotype
get organ and viral infections in particular, are commonly viewed for AAD developed transient AAD over a 2-year period of IFN␣
as potentially triggering factors. Under infections, parenchymal treatment [79]. Also, IFN␣ therapy has been reported to exacer-
cells are exposed to various microbial products and cytokines bate clinical or subclinical adrenocortical insufficiency, leading
that alter their status and their steady-state dialogue with the to an increased need for hormone supplement [80–82]. Collec-
immune system [63]. These changes are most likely instrumental tively, these findings could indicate a role for viruses and/or type
to the development and progression of autoimmune disease. I interferons in promoting adrenal autoimmunity. The mech-
Certain herpesviruses like herpes simplex virus 1 (HSV-1) anisms operative under such a scenario would likely involve
[64–67] and cytomegalovirus (CMV) [68–70], as well as the a number of effects on the adrenal cortex, as these cells seem
adenovirus [71], have a tropism for adrenocortical cells. In responsive to both TLR stimuli and various cytokines, including
some cases, a connection between herpes virus infection and type I IFNs [60,83–85].
adrenocortical insufficiency has indeed been suggested [72–75] The recent introduction of so-called checkpoint inhibitors
although the exact disease aetiology in these cases was uncertain. targeting CTLA-4, programmed cell death protein 1 (PD-
Recently, two studies showing seasonal variation in the onset of 1), and PD-1 ligand (PD-L1), in the treatment of malignant
AAD provide indirect evidence for a role of a seasonal microbial diseases such as malignant melanoma and lung cancer have
agent such as a virus in the pathogenesis of AAD [76,77]. brought about significant immunological side-effects [86],
A link between type I interferons (IFNs) and adrenal including endocrinopathies like autoimmune hypophysitis, thy-
autoimmunity also seems to exist. The development of 21OH roiditis and type 1 diabetes. A few cases of AAD have also
autoantibodies has been reported in patients receiving IFN␣ been observed. One patient developed thyrotoxicosis and later
A. Hellesen et al. / Annales d’Endocrinologie 79 (2018) 157–163 161

AAD with 21OH autoantibodies after treatment with pem- applicability in diagnosis and disease prediction. Endocr Rev 2002;23:
brolizumab (anti-PD-1) for metastatic melanoma [87]. A similar 327–64.
case has also been reported with nivolumab (also anti-PD-1) [5] Bancos I, et al. Diagnosis and management of adrenal insufficiency.
Lancet Diabetes Endocrinol 2015;3:216–26.
[88]. Furthermore, using immunodeficient mice reconstituted [6] Bornstein SR, et al. Diagnosis and treatment of primary adrenal insuffi-
with human stem cells it was demonstrated that treatment ciency: an endocrine society clinical practice guideline. J Clin Endocrinol
with anti-CTLA4 antibodies induced a number of presumably Metab 2016;101:364–89.
autoimmune pathologies including hepatitis, sialitis, and most [7] Choudhry A, Marchi C, Stephens W. Manchester United induced addis-
importantly, adrenalitis [89]. onian crisis. BMJ 2011;343:d7732.
[8] Hahner S, et al. High incidence of adrenal crisis in educated patients
with chronic adrenal insufficiency: a prospective study. J Clin Endocrinol
6. Preventive and regenerative treatment for Metab 2015;100:407–16.
[9] Dunlop D. Eighty-six cases of Addison’s disease. Br Med J
autoimmune adrenal insufficiency
1963;2(5362):887–91.
[10] Winqvist O, Karlsson FA, Kampe O. 21-Hydroxylase, a major
It is commonly assumed that primary adrenal insufficiency autoantigen in idiopathic Addison’s disease. Lancet 1992;339(8809):
is complete, but we and others have found that a subgroup of 1559–62.
patients retain a certain degree of cortisol production even after [11] Fichna M, et al. Screening for associated autoimmune disorders in Polish
patients with Addison’s disease. Endocrine 2010;37:349–60.
several years of disease [90–92]. Thus, there are still function-
[12] Coco G, et al. Estimated risk for developing autoimmune Addison’s dis-
ing cells that might regenerate and proliferate if the autoimmune ease in patients with adrenal cortex autoantibodies. J Clin Endocrinol
attack is contained. Pearce and co-workers have explored the Metab 2006;91:1637–45.
capacity for regeneration of adrenal function by either stim- [13] Myhre AG, et al. Autoimmune adrenocortical failure in Norway autoan-
ulation with long-acting cosyntropin or immunomodulation by tibodies and human leukocyte antigen class II associations related to
clinical features. J Clin Endocrinol Metab 2002;87:618–23.
rituximab [93,94]. In both cases, a few patients regained function
[14] Husebye ES, Lovas K. Immunology of Addison’s disease and premature
of the adrenals. Recently it was reported that steroid-producing ovarian failure. Endocrinol Metab Clin North Am 2009;38:389–405, ix.
cells could be regenerated by lentiviral transformation of stem [15] Meyer G, et al. Increasing prevalence of Addison’s disease in Ger-
cells harvested from blood and urine with different transcription man females: health insurance data 2008–2012. Eur J Endocrinol
factors [95]. Use of such cells in combination with immunomod- 2014;170:367–73.
[16] Erichsen MM, et al. Normal overall mortality rate in Addison’s dis-
ulatory therapy could constitute a future therapeutic approach in
ease, but young patients are at risk of premature death. Eur J Endocrinol
AAD. 2009;160:233–7.
Still, a deeper understanding of the pathogenesis, including [17] Eisenbarth GS, Gottlieb PA. Autoimmune polyendocrine syndromes. N
the underlying genetic background, the interplay of the adrenal Engl J Med 2004;350:2068–79.
cortex and the immune system with environmental factors, and [18] Bruserud O, et al. A longitudinal follow-up of autoimmune polyendocrine
syndrome type 1. J Clin Endocrinol Metab 2016;101:2975–83.
the immunological mechanisms destroying the steroidogenic tis-
[19] Ahonen P, et al. Clinical variation of autoimmune polyendocrinopathy-
sue, is needed to bring new immunomodulatory and regenerative candidiasis-ectodermal dystrophy (APECED) in a series of 68 patients.
therapy to the table that can restore adrenal function in the not N Engl J Med 1990;322:1829–36.
too distant future. [20] Drury MI, et al. Juvenile familial endocrinopathy. Clin Exp Immunol
1970;7:125–32.
[21] Irvine WJ, Stewart AG, Scarth L. A clinical and immunological study
Disclosure of interest of adrenocortical insufficiency (Addison’s disease). Clin Exp Immunol
1967;2:31–70.
The authors declare that they have no competing interest. [22] Wolff AS, et al. Autoimmune polyendocrine syndrome type 1 in Nor-
way: phenotypic variation, autoantibodies, and novel mutations in the
autoimmune regulator gene. J Clin Endocrinol Metab 2007;92:595–603.
Acknowledgements [23] Eriksson D, et al. Extended exome sequencing identifies BACH2
as a novel major risk locus for Addison’s disease. J Intern Med
2016;280:595–608.
The study was supported by Grants from The Regional Health [24] Yu L, et al. DRB1*04 and DQ alleles: expression of 21-hydroxylase
authorities of Western Norway, The Research Council of Nor- autoantibodies and risk of progression to Addison’s disease. J Clin
way and The Novo Nordisk Foundation. Endocrinol Metab 1999;84:328–35.
[25] Baker PR, et al. Haplotype analysis discriminates genetic risk for DR3-
associated endocrine autoimmunity and helps define extreme risk for
References Addison’s disease. J Clin Endocrinol Metab 2010;95:E263–70.
[26] Bratland E, et al. T cell responses to steroid cytochrome P450 21-
[1] Erichsen MM, et al. Clinical, immunological, and genetic features of hydroxylase in patients with autoimmune primary adrenal insufficiency.
autoimmune primary adrenal insufficiency: observations from a Norwe- J Clin Endocrinol Metab 2009;94:5117–24.
gian registry. J Clin Endocrinol Metab 2009;94:4882–90. [27] Dawoodji A, et al. High frequency of cytolytic 21-hydroxylase-specific
[2] Dalin F, et al. Clinical and immunological characteristics of autoim- CD8+ T cells in autoimmune Addison’s disease patients. J Immunol
mune Addison’s disease: a nationwide Swedish multicenter study. J Clin 2014;193:2118–26.
Endocrinol Metab 2016, jc20162522. [28] Rottembourg D, et al. 21-Hydroxylase epitopes are targeted by CD8T
[3] Olafsson AS, Sigurjonsdottir HA. Increasing prevalence of Addison dis- cells in autoimmune Addison’s disease. J Autoimmun 2010;35:309–15.
ease: results from a nationwide study. Endocr Pract 2016;22:30–5. [29] Gambelunghe G, et al. Microsatellite polymorphism of the MHC class I
[4] Betterle C, et al. Autoimmune adrenal insufficiency and autoim- chain-related (MIC-A and MIC-B) genes marks the risk for autoimmune
mune polyendocrine syndromes: autoantibodies, autoantigens, and their Addison’s disease. J Clin Endocrinol Metab 1999;84:3701–7.
162 A. Hellesen et al. / Annales d’Endocrinologie 79 (2018) 157–163

[30] Triolo TM, et al. Homozygosity of the polymorphism MICA5.1 iden- [55] Kanczkowski W, et al. Differential expression and action of Toll-
tifies extreme risk of progression to overt adrenal insufficiency among like receptors in human adrenocortical cells. Mol Cell Endocrinol
21-hydroxylase antibody-positive patients with type 1 diabetes. J Clin 2009;300:57–65.
Endocrinol Metab 2009;94:4517–23. [56] Conti B, et al. Induction of interferon-gamma inducing factor in the
[31] Mitchell AL, Pearce SH. Autoimmune Addison disease: pathophysiology adrenal cortex. J Biol Chem 1997;272:2035–7.
and genetic complexity. Nat Rev Endocrinol 2012;8:306–16. [57] Gonzalez-Hernandez JA, et al. Human adrenal cells express tumor
[32] Wolff AS, et al. CTLA-4 as a genetic determinant in autoimmune Addi- necrosis factor-alpha messenger ribonucleic acid: evidence for paracrine
son’s disease. Genes Immun 2015;16:430–6. control of adrenal function. J Clin Endocrinol Metab 1996;81:807–13.
[33] Ban Y, et al. Analysis of the CTLA-4, CD28, and inducible costim- [58] Rotondi M, et al. Elevated serum interferon-gamma-inducible
ulator (ICOS) genes in autoimmune thyroid disease. Genes Immun chemokine-10/CXC chemokine ligand-10 in autoimmune primary
2003;4:586–93. adrenal insufficiency and in vitro expression in human adrenal cells pri-
[34] Skinningsrud B, et al. Mutation screening of PTPN22: association of the mary cultures after stimulation with proinflammatory cytokines. J Clin
1858T-allele with Addison’s disease. Eur J Hum Genet 2008;16:977–82. Endocrinol Metab 2005;90:2357–63.
[35] Salmond RJ, et al. The tyrosine phosphatase PTPN22 discriminates [59] Bornstein SR, Chrousos GP. Clinical review 104: Adrenocorticotropin
weak self peptides from strong agonist TCR signals. Nat Immunol (ACTH)- and non-ACTH-mediated regulation of the adrenal cortex: neu-
2014;15:875–83. ral and immune inputs. J Clin Endocrinol Metab 1999;84:1729–36.
[36] Wang Y, et al. The autoimmunity-associated gene PTPN22 potentiates [60] Bratland E, Hellesen A, Husebye ES. Induction of CXCL10 chemokine
toll-like receptor-driven, type 1 interferon-dependent immunity. Immu- in adrenocortical cells by stimulation through toll-like receptor 3. Mol
nity 2013;39:111–22. Cell Endocrinol 2013;365:75–83.
[37] Pazderska A, et al. A variant in the BACH2 gene is associated with suscep- [61] Edvardsen K, et al. Peripheral blood cells from patients with autoimmune
tibility to autoimmune Addison’s Disease in humans. J Clin Endocrinol Addison’s disease poorly respond to interferons in vitro, despite elevated
Metab 2016;101:3865–9. serum levels of interferon-inducible chemokines. J Interferon Cytokine
[38] Cooper JD, et al. Meta-analysis of genome-wide association study Res 2015;35:759–70.
data identifies additional type 1 diabetes risk loci. Nat Genet [62] Christen U, von Herrath MG. Initiation of autoimmunity. Curr Opin
2008;40:1399–401. Immunol 2004;16:759–67.
[39] Dubois PC, et al. Multiple common variants for celiac disease influencing [63] Hill NJ, et al. The target tissue in autoimmunity–an influential niche. Eur
immune gene expression. Nat Genet 2010;42:295–302. J Immunol 2007;37:589–97.
[40] Medici M, et al. Identification of novel genetic Loci associated with [64] Potratz D, et al. Herpes simplex virus type 1 and 2 in the adrenal glands:
thyroid peroxidase antibodies and clinical thyroid disease. PLoS Genet replication and histopathology. Arch Virol 1986;90:207–22.
2014;10:e1004123. [65] Nachtigal M, Caulfield J. Early and late pathologic changes in the adrenal
[41] Skov J, et al. Heritability of Addison’s disease and prevalence of asso- glands of mice after infection with herpes simplex virus type 1. Am J
ciated autoimmunity in a cohort of 112,100 Swedish twins. Endocrine Pathol 1984;115:175.
2017;58:521–7. [66] Aita K, et al. Acute adrenal infection by HSV-1: role of apoptosis in viral
[42] al Sabri AM, Smith N, Busuttil A. Sudden death due to auto-immune replication. Arch Virol 2001;146:2009–20.
Addison’s disease in a 12-year-old girl. Int J Legal Med 1997;110:278–80. [67] Nakamura Y, et al. Herpes simplex viral infection in human neonates:
[43] Imam K, Abdullah M, Felicetta JV. Alopecia universalis as a feature of an immunohistochemical and electron microscopic study. Hum Pathol
polyglandular autoimmunity type I. West J Med 1988;149:338–41. 1985;16:1091–7.
[44] Rosenthal FD, Davies MK, Burden AC. Malignant disease presenting as [68] Trevisan M, et al. Human cytomegalovirus productively infects adreno-
Addison’s disease. Br Med J 1978;1(6127):1591–2. cortical cells and induces an early cortisol response. J Cell Physiol
[45] Anderson JR, et al. Auto-antibodies in Addison’s disease. Lancet 2009;221:629–41.
1957;272(6979):1123–4. [69] Barzon L, et al. Detection of polyomaviruses and herpesviruses in human
[46] Blizzard RM, Kyle M. Studies of the adrenal antigens and antibodies in adrenal tumors. Oncogene 2008;27:857–64.
Addison’s disease. J Clin Invest 1963;42:1653–60. [70] Pomara G, et al. Cytomegalovirus and BK-virus co-infection of a clin-
[47] Laureti S, et al. Levels of adrenocortical autoantibodies correlate with ically non-functioning adrenal adenoma: innocent bystanders or new
the degree of adrenal dysfunction in subjects with preclinical Addison’s pathogenetic agents? Eur J Histochem 2006;50:131–2.
disease. J Clin Endocrinol Metab 1998;83:3507–11. [71] Matkovic U, et al. Investigation on human adrenocortical cell
[48] Boscaro M, et al. Hormonal responses during various phases of autoim- response to adenovirus and adenoviral vector infection. J Cell Physiol
mune adrenal failure: no evidence for 21-hydroxylase enzyme activity 2009;220:45–57.
inhibition in vivo. J Clin Endocrinol Metab 1996;81:2801–4. [72] Brain RT, Pugh RC, Dudgeon JA. Adrenal necrosis in generalized herpes
[49] Hsu LC, et al. The N-terminal hydrophobic domain of P450c21 is simplex. Arch Dis Child 1957;32(162):120–6.
required for membrane insertion and enzyme stability. J Biol Chem [73] Hertel NT, et al. Adrenocortical insufficiency associated with Epstein-
1993;268:14682–6. Barr virus infection in a patient with the Wiskott-Aldrich syndrome. Eur
[50] Husebye ES, et al. The substrate-binding domain of 21-hydroxylase, the J Pediatr 1987;146:603–4.
main autoantigen in autoimmune Addison’s disease, is an immunodomi- [74] Schmitt K, et al. Autoimmune hepatitis and adrenal insufficiency in an
nant T cell epitope. Endocrinology 2006;147:2411–6. infant with human herpesvirus-6 infection. Lancet 1996;348(9032):966.
[51] Ehrhart-Bornstein M, et al. Intraadrenal interactions in the regulation of [75] Guinovart RM, et al. A black tongue in a young woman. Clin Exp Der-
adrenocortical steroidogenesis. Endocr Rev 1998;19:101–43. matol 2011;36:429–30.
[52] Engstrom L, et al. Systemic immune challenge activates an intrinsically [76] Pazderska A, et al. Impact of month of birth on the risk of devel-
regulated local inflammatory circuit in the adrenal gland. Endocrinology opment of autoimmune Addison’s disease. J Clin Endocrinol Metab
2008;149:1436–50. 2016;101:4214–8.
[53] Gonzalez-Hernandez JA, et al. IL-1 is expressed in human adrenal gland [77] Chantzichristos D, et al. Incidence, prevalence and seasonal onset
in vivo. Possible role in a local immune-adrenal axis. Clin Exp Immunol variation of Addison’s disease among persons with type 1 diabetes mel-
1995;99:137–41. litus: nationwide, matched cohort studies. Eur J Endocrinol 2018;178:
[54] Gonzalez-Hernandez JA, et al. Interleukin-6 messenger ribonucleic acid 115–22.
expression in human adrenal gland in vivo: new clue to a paracrine [78] Wesche B, et al. Induction of autoantibodies to the adrenal cortex and
or autocrine regulation of adrenal function. J Clin Endocrinol Metab pancreatic islet cells by interferon alpha therapy for chronic hepatitis C.
1994;79:1492–7. Gut 2001;48:378–83.
A. Hellesen et al. / Annales d’Endocrinologie 79 (2018) 157–163 163

[79] Krysiak R, Boldys A, Okopien B. Autoimmune polyglandular syndrome [96] Blomhoff A, et al. Polymorphisms in the cytotoxic T lymphocyte antigen-
type 2 induced by treatment with interferon alpha. Am J Med Sci 4 gene region confer susceptibility to Addison’s disease. J Clin Endocrinol
2011;341:504–7. Metab 2004;89:3474–6.
[80] Oshimoto K, et al. A case of Addison’s disease which became worse [97] Mitchell AL, et al. Programmed death ligand 1 (PD-L1) gene variants
during interferon therapy: insulin secretion under hyposmolarity. Nihon contribute to autoimmune Addison’s disease and Graves’ disease suscep-
Naibunpi Gakkai Zasshi 1994;70:511–6. tibility. J Clin Endocrinol Metab 2009;94:5139–45.
[81] Tran HA, et al. Exacerbation of hepatitis C induced subclinical hypoad- [98] Roycroft M, et al. The tryptophan 620 allele of the lymphoid tyrosine
renalism by Interferon-alpha2beta: a case report. Cases J 2008;1:157. phosphatase (PTPN22) gene predisposes to autoimmune Addison’s dis-
[82] Knost JA, et al. Increased steroid dependence after recombinant leucocyte ease. Clin Endocrinol (Oxf) 2009;70:358–62.
interferon therapy. Lancet 1981;2:1287–8. [99] Zurawek M, et al. A coding variant in NLRP1 is associated with autoim-
[83] van Koetsveld PM, et al. Potent inhibitory effects of type I interferons on mune Addison’s disease. Hum Immunol 2010;71:530–4.
human adrenocortical carcinoma cell growth. J Clin Endocrinol Metab [100] Magitta NF, et al. A coding polymorphism in NALP1 confers risk
2006;91:4537–43. for autoimmune Addison’s disease and type 1 diabetes. Genes Immun
[84] van Koetsveld PM, et al. Interferon-beta is a potent inhibitor of cell growth 2009;10:120–4.
and cortisol production in vitro and sensitizes human adrenocortical car- [101] Mitchell AL, et al. Association of autoimmune Addison’s disease
cinoma cells to mitotane. Endocr Relat Cancer 2013;20:443–54. with alleles of STAT4 and GATA3 in European cohorts. PLoS One
[85] Hellesen A, et al. The effect of types I and III interferons on adrenocortical 2014;9:e88991.
cells and its possible implications for autoimmune Addison’s disease. Clin [102] Ghaderi M, et al. MHC2TA single nucleotide polymorphism and genetic
Exp Immunol 2014;176:351–62. risk for autoimmune adrenal insufficiency. J Clin Endocrinol Metab
[86] Postow MA, Sidlow R, Hellmann MD. Immune-related adverse 2006;91:4107–11.
events associated with immune checkpoint blockade. N Engl J Med [103] Skinningsrud B, et al. Polymorphisms in CLEC16A and CIITA at 16p13
2018;378:158–68. are associated with primary adrenal insufficiency. J Clin Endocrinol
[87] Paepegaey AC, et al. Polyendocrinopathy resulting from pembrolizumab Metab 2008;93:3310–7.
in a patient with a malignant melanoma. J Endocr Soc 2017;1:646–9. [104] Owen CJ, et al. Analysis of the Fc receptor-like-3 (FCRL3) locus
[88] Trainer H, et al. Hyponatraemia secondary to nivolumab-induced primary in Caucasians with autoimmune disorders suggests a complex pat-
adrenal failure. Endocrinol Diabetes Metab Case Rep 2016:2016. tern of disease association. J Clin Endocrinol Metab 2007;92:
[89] Vudattu NK, et al. Humanized mice as a model for aberrant responses in 1106–11.
human T cell immunotherapy. J Immunol 2014;193:587–96. [105] Napier C, et al. Role of the X-linked gene GPR174 in autoimmune Addi-
[90] Methlie P, et al. Multisteroid LC-MS/MS assay for glucocorticoids and son’s disease. J Clin Endocrinol Metab 2015;100:E187–90.
androgens, and its application in Addison’s disease. Endocr Connect [106] Mitchell AL, et al. Linkage analysis in autoimmune Addison’s dis-
2013;2:125–36. ease: NFATC1 as a potential novel susceptibility locus. Plos One
[91] Smans LC, Zelissen PM. Partial recovery of adrenal function in a patient 2015;10:e0123550.
with autoimmune Addison’s disease. J Endocrinol Invest 2008;31:672–4. [107] Fichna M, et al. Association of the CYP27B1 C(-1260)A polymor-
[92] Smans LC, Zelissen PM. Does recovery of adrenal function occur in phism with autoimmune Addison’s disease. Exp Clin Endocrinol Diabetes
patients with autoimmune Addison’s disease? Clin Endocrinol (Oxf) 2010;118:544–9.
2011;74:434–7. [108] Lopez ER, et al. A promoter polymorphism of the CYP27B1 gene
[93] Pearce SH, et al. Adrenal steroidogenesis after B lymphocyte deple- is associated with Addison’s disease, Hashimoto’s thyroiditis, Graves’
tion therapy in new-onset Addison’s disease. J Clin Endocrinol Metab disease and type 1 diabetes mellitus in Germans. Eur J Endocrinol
2012;97:E1927–32. 2004;151:193–7.
[94] Gan EH, et al. Residual adrenal function in autoimmune Addison’s dis- [109] Jennings CE, et al. A haplotype of the CYP27B1 promoter is associated
ease: improvement after tetracosactide (ACTH1-24) treatment. J Clin with autoimmune Addison’s disease but not with Graves’ disease in a UK
Endocrinol Metab 2014;99:111–8. population. J Mol Endocrinol 2005;34:859–63.
[95] Ruiz-Babot G, et al. Modeling congenital adrenal hyperplasia and [110] Pani MA, et al. Vitamin D receptor genotype is associated with Addison’s
testing interventions for adrenal insufficiency using donor-specific repro- disease. Eur J Endocrinol 2002;147:635–40.
grammed cells. Cell Rep 2018;22:1236–49.

You might also like