Open Access

A novel variant in AIRE causing a rare, non‑classical autoimmune polyendocrine syndrome type 1

  • Authors:
    • Wen‑Bin Zheng
    • Lu‑Jiao Li
    • Di‑Chen Zhao
    • Ou Wang
    • Yan Jiang
    • Wei‑Bo Xia
    • Mei Li
  • View Affiliations

  • Published online on: June 12, 2020     https://doi.org/10.3892/mmr.2020.11227
  • Pages: 1285-1294
  • Copyright: © Zheng et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Autoimmune polyendocrine syndrome type 1 (APS‑1) is a rare inherited autoimmune disease, characterized by a classic triad, including chronic mucocutaneous candidiasis, primary adrenocortical insufficiency and hypoparathyroidism. The present study investigated phenotypes and pathogenic variants in a Chinese woman with non‑classical APS‑1. Disease‑associated variants in a patient with APS‑1 were identified via targeted next generation sequencing and the variant was confirmed via Sanger sequencing. Serum levels of calcium, phosphorus, parathyroid hormone (PTH), follicle‑stimulating hormone (FSH), luteinizing hormone (LH), estradiol and urinary levels of calcium were measured. Blood count assays and bone marrow morphology were investigated. The patient was a 32‑year‑old woman who had suffered from typical carpopedal spasms since she was 7 years old. She developed syncope, primary amenorrhea, intermittent diarrhea and general fatigue in subsequent years. Hypocalcemia, hyperphosphatemia, low levels of PTH and estradiol, elevated levels of FSH and LH, and absence of erythroblasts were observed, which indicated hypoparathyroidism, primary ovarian insufficiency and pure red cell aplasia. A novel heterozygous missense variant (NM_000383.2: c.623G>T, NP_000374.1: p.Gly208Val) in exon 5 of autoimmune regulator and a reported variant (NM_000383.2: c.371C>T, NP_000374.1: p.Pro124Leu) in exon 3 were detected, of which the c.623G>T variant may be a pathogenic variation that induces APS‑1. Under a regular follow‑up and therapeutic adjustment of calcium, calcitriol, hormone replacement therapy and methylprednisolone, the endocrine function and clinical symptoms of the patient were notably improved. The results of the present study expand the known genetic and phenotypical spectra of APS‑1.

Introduction

Autoimmune polyendocrine syndrome type 1 (APS-1; OMIM 240300), also known as autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy, is a rare disease, characterized by chronic mucocutaneous candidiasis (CMC), primary adrenocortical insufficiency (PAI) and hypoparathyroidism (HP) (1). APS-1 may present with other abnormities, such as primary ovarian insufficiency, vitiligo, alopecia, type 1 diabetes mellitus (T1D), chronic intestinal dysfunction, enamel hypoplasia, and autoimmune hepatitis (2,3). Pure red cell aplasia (PRCA), metaphyseal dysplasia, retinitis and polyarthritis have also been reported in patients with APS-1 (4,5).

APS-1 is primarily caused by variants in the autoimmune regulator gene (AIRE), which is located on chromosome 21q22.3 (6). AIRE contains 14 exons and encodes a 57-kDa transcription regulator of 545 amino acids (7,8). AIRE is primarily expressed in medullary thymic epithelial cells, which serve key roles in maintaining central immunological tolerance via promoting the negative selection of autoreactive thymocytes and building the thymic microarchitecture (6). AIRE is also distributed in the periphery of antigen-presenting cells (9). If the functions of AIRE are impaired, autoreactive T cells can escape into the periphery, resulting in a number of autoimmune abnormities such as autoimmune attack against parathyroid and adrenal glands in APS-1 (6).

APS-1 is relatively prevalent in Iranian Jews and Sardinians, with a prevalence of 1:145,000 to 1:9,000 (1012). Over 100 different variants in AIRE gene have been found in patients with APS-1, including substitutions, insertions, deletions and splice site variants [BIOBASE Human Gene Mutation Database (HGMD) professional 2019.1; hgmd.cf.ac.uk/ac/index.php]. To the best of our knowledge, however, there are very few reports of APS-1 in Chinese patients, and its pathological mechanism has not been fully elucidated.

The present study investigated the phenotype and genotype of a Chinese woman with non-classical APS-1. Characteristics of previously reported Chinese patients with APS-1 induced by AIRE gene variants were also summarized.

Materials and methods

Subjects

A 32-year-old woman of Chinese Han origin from a non-consanguineous family was recruited following admission to the Department of Endocrinology, Peking Union Medical College Hospital (PUMCH; Beijing, China) in 2018, with suspected APS-1 based on the clinical features of syncope, tetany, primary amenorrhea, intermittent diarrhea and general fatigue.

Phenotypic evaluation

The functions of multiple endocrine glands and examined levels of numerous autoantibodies were evaluated. Serum levels of calcium, phosphorus, creatinine (Cr) and alanine aminotransferase (ALT), and 24-h urinary concentrations of calcium (24hUCa) and phosphorus (24hUP) were assessed via an AU5800 Beckman Automatic Biochemical Analyzer (Beckman Coulter, Inc.). Serum levels of parathyroid hormone (PTH), follicle-stimulating hormone (FSH), luteinizing hormone (LH) and estradiol, as well as serum-free thyroxine (FT4), free triiodothyronine (FT3), thyroid-stimulating hormone (TSH) and autoantibodies to thyroid peroxidase antigen (TPOAb) and thyroglobulin (TGAb) were measured using an automated electrochemiluminescence system (Roche Diagnostics). Blood samples measured for serum cortisol and plasma adrenocorticotropic hormone (ACTH) were collected at 8 AM and measured via fluorescent immunoassay (Diagnostic Products). Serum autoantibodies to islet cells antibodies (ICA) and glutamic acid decarboxylase antibodies (GADA) were measured using radioimmunoassays kits (Cisbio; Perkin Elmer, Inc.). Serum autoantibodies to antinuclear antibody (ANA), 52 kDa Ro/SS-A molecule (RO-52), as well as anti-neutrophil cytoplasmic antibodies (ANCA) were tested using an indirect immunofluorescence kit (ANA screen 11; cat. no. EA 1590-9601-11 G; Euroimmun AG). The blood count assays were measured using a Sysmex XE-5000 hematology analyzer (Sysmex Corporation). Serum levels of erythropoietin (EPO; cat. no. DEP00) and antibodies to IFN-ω, IFN-α (cat. no. 21100-1), IL-17A (cat. no. MAB317-100), IL-17F (cat. no. MAB13353-100) and IL-22 (cat. no. MAB7822-100) were detected using ELISA kits (R&D Systems Inc.).

Morphology of thyroid, abdomen, uterus and ovary were assessed via standard ultrasound scanning (Siemens Healthineers). Calcification in the central nervous system was evaluated via CT scan.

The diagnosis of hypoparathyroidism was determined by hypocalcemia, hyperphosphatemia and decreased intact PTH level. The diagnosis of primary ovarian insufficiency was based on amenorrhea, absent pubertal development and elevated serum levels of FSH or LH. The diagnosis of PRCA was based on normal marrow in the absence of erythroblasts (<1% erythroblasts on the differential count of bone marrow cells).

Identification of pathogenic variants

DNA was harvested from peripheral leucocytes using the DNA Extraction kit (QIAamp DNA; Qiagen GmbH) according to the manufacturer's instructions. Targeted next-generation sequencing (NGS) was used to identify the pathogenic variants. A panel containing >700 genes of bone disease was created to capture all exons sequences of the candidate genes of APS-1 and hypoparathyroidism (such as AIRE, T-box transcription factor 1, tubulin-specific chaperone E and family with sequence similarity 111 member A), as previously described (13). Bioinformatic analysis was performed as previously described (13).

Fragments from the patient, her parents and 500 unrelated healthy controls covering the variant sites in AIRE identified by NGS were amplified via PCR. Primer sequences were as follows: Exon 3: Forward 5′-ACCCTACCCCTGGAGAAAAC-3′ and reverse 5′-TGGTCCAGTGTGTGGGTCTA-3′; and exon 5: forward, 5′-GCCTGCTTCTGGCATAGAGT-3′ and reverse, 5′-AACAGTCCGGTCTGCTGTG-3′.

PCR was performed using TaqMix DNA polymerase (Biomed) under the following conditions: 95°C for 3 min, followed by 35 cycles of 95°C for 30 sec, annealing at 60°C for 30 sec, and 72°C for 1 min, followed by a final extension at 72°C for 10 min. Products of PCR were purified and sequenced on the ABI377 DNA automated sequencer using Big Dye Terminators Cycle Sequencing Ready Reaction kit (Applied Biosystems; Thermo Fisher Scientific, Inc.) according to the manufacturer's instructions.

In order to predict the potential deleterious effects of the variants, PolyPhen-2 (Polymorphism Phenotyping v2; genetics.bwh.harvard.edu/pph2/), SIFT (sift.jcvi.org/) and MutationTaster (mutationtaster.org/) were utilized. The conservation of the amino acid substitution position among species was analyzed via UniProt software (uniprot.org).

A literature review of reported Chinese patients with APS-1 exhibiting AIRE variants was performed and clinical and genetic characteristics were summarized (7,1417).

Three-dimensional modeling of AIRE

In order to investigate the change in protein induced by the pathogenic variant in AIRE, the three-dimensional structure of the AIRE protein was constructed via SWISS-MODEL (swissmodel.expasy.org). Subsequently, the disease-associated p.Gly208Val substitution of AIRE protein was built using the mutagenesis module of PyMoL software (v2.3.1; pymol.org/).

The present study was approved by the Scientific Ethics Committee of PUMCH (approval no. JS-2081). A total of 500 unrelated healthy individuals were recruited as controls for genetic study. Written informed consent was provided by all subjects prior to participation. The pedigree of the family of the patient is shown in Fig. 1A.

Results

Phenotypes of the patient

The patient, a 32-year-old woman (height, 158.0 cm; weight, 52.5 kg; body mass index, 21.0 kg/m2), came from a Chinese non-consanguineous family with no history of autoimmune disease. At 7 years of age, she presented with tetany and hypocalcemia. She was supplemented with Calcichew D (500 mg elemental calcium, 200 IU vitamin D per tablet; GE Pharmaceutical Shanghai Co., Ltd.) three times daily and serum calcium levels were not monitored. At 17 years of age, she suffered from syncope and a generalized tonic-clonic seizure and was admitted to PUMCH for the first time. She was diagnosed with hypoparathyroidism based on PTH deficiency (PTH<1.0 pg/ml), hypocalcemia (serum total calcium, 1.35 mmol/l) and hyperphosphatemia (serum phosphorus, 1.91 mmol/l). CT scans revealed bilateral calcifications in the basal ganglia of the brain. One year later, she was admitted to the Department of Gynecology with primary amenorrhea and was diagnosed as having primary ovarian insufficiency, according to low estradiol (6.74 pg/ml), increased LH (33.7 mIU/ml) and FSH (82.6 mIU/ml) levels, as well as uterine hypoplasia in ultrasonography. At 31 years of age, she experienced intermittent diarrhea and general fatigue. Whole blood cell count revealed a notably decreased hemoglobin level (6.1 g/dl), with normal platelet count (PLT, 306,000/µl) and white blood cells (WBCs, 874,000/µl). The bone marrow aspirate showed normocellularity, a lack of erythroid precursors (1.0% of bone marrow nucleated cells), normal granulocyte precursors and megakaryocytes with lymphocytosis (30.5%). In addition, serum level of erythropoietin was notably elevated (>776.00 mIU/ml). The patient was diagnosed with PRCA. Gastrointestinal endoscopy and histological analysis of the biopsies revealed chronic atrophic gastritis and gastric ulcers, with normal appearance of the duodenal mucosa. Helicobacter pylori was not present in the gastric mucosa. The abdominal ultrasound was normal. Serum cortisol, plasma ACTH, serum FT4, FT3 and TSH levels were within the normal range. For autoantibodies, RO-52, GADA and antibodies to IFN-ω and IFN-α were positive, whereas ANA, ANCA, TGAb, TPOAb, ICA and antibodies to IL-17A, IL-17F and IL-22 were negative.

Combined with clinical manifestations, the patient was suspected of having APS-1. The clinical manifestations of the patient are listed in Table I.

Table I.

Clinical presentation and laboratory results of a patient with APS-1.

Table I.

Clinical presentation and laboratory results of a patient with APS-1.

Age, yearsClinical manifestationLaboratory resultsReference range
7TetanyNA/
17SyncopeSerum total calcium, 1.350 mmol/l2.13–2.70 mmol/l
Generalized tonic-clonic seizureSerum phosphorus, 1.910 mmol/l0.81–1.45 mmol/l
Serum parathyroid hormone, <1.000 pg/ml7.00–53.00 pg/ml
18AmenorrheaSerum estradiol, 6.740 pg/ml50.00–154.40 pg/ml
Serum luteinizing hormone, 33.700 mIU/ml4.40–7.10 mIU/ml
Serum follicle-stimulating hormone, 82.600 mIU/ml5.10–7.00 mIU/ml
Ultrasonography, uterine hypoplasia
31Intermittent diarrheaHemoglobin, 6.100 g/dl11.00–15.00 g/dl
General fatigueHematocrit, 19.000%35.00–50.00%
Platelets, 306,000.000/µl 100,000.00–350,000.00/µl
White blood cells, 874,000.000/µl 350,000.00–950,000.00/µl
Plasma adrenocorticotropic hormone, 27.000 pg/ml0.00–46.00 pg/ml
Serum cortisol, 19.430 µg/dl4.00–22.30 µg/dl
Serum free thyroxine, 1.100 ng/dl0.81–1.89 ng/dl
Serum free triiodothyronine, 3.370 pg/ml1.80–4.10 g/ml
Serum thyroid-stimulating hormone, 2.306 µIU/ml0.38–4.34 µIU/ml
Thyroglobulin antibodies, 12.270 IU/ml<115.00 IU/ml
Thyroperoxidase antibodies, 10.910 IU/ml<34.00 IU/ml
Erythropoietin, >776.000 mIU/ml4.5.0–31.88 mIU/ml
Bone marrow aspirate, normocelluiarity, lack of erythroid precursors
IFN-ω Abs/IFN-α Abs/RO-52/GADA, PositiveGADA, <10.00 IU/ml
IL-17A Abs/IL-17F Abs/IL-22IFN-ω Abs/IFN-α
Abs/ICA/IA-2/ANA/ANCA, Negative Abs/RO-52/IL-17A
Abs/IL-17F Abs/IL-22
Abs/ICA/IA-2/ANA/ANCA,
Negative

[i] NA, not available; IFN-ω Abs, IFN-ω antibodies; IFN-α Abs, IFN-α antibodies; GADA, glutamic acid decarboxylase antibodies; ANA, anti-nuclear antibodies; RO52, 52 kDa Ro/SS-A molecule; IL-17A Abs, IL-17A antibodies; IL-17F Abs, IL-17F antibodies; IL-22 Abs, IL-22 antibodies; ICA, islet cell antibodies; ANCA, anti-neutrophil cytoplasmic antibodies.

The parents did not display symptoms or signs of APS-1; therefore, they were reluctant to receive further biochemical and autoantibody profile examinations.

Treatment and follow-up

The patient was prescribed 3 tablets of calcium carbonate (200 mg elemental calcium per tablet) and one capsule of calcitriol (0.25 µg per capsule) three times daily to manage hypoparathyroidism. For primary ovarian insufficiency, she received a 21-day sequential therapy with Climen (2 mg estradiol valerate, days 1–21 with 1 mg cyproterone acetate, days 12–21; Bayer, AG), followed by a 7-day treatment-free interval. The patient received a transfusion of packed red blood cells, and then 75 mg cyclosporine A was given twice daily. As the serum ALT level increased to 304 U/l one month later, she switched to methylprednisolone (4 mg per tablet). The therapeutic dosage was adjusted according to serum levels of calcium, phosphate, 24-h urinary calcium excretion and level of hemoglobin.

During the treatment, the frequency of tetany was notably decreased, and the patient experienced regular menstruation. On the final examination, serum calcium level increased to 1.84 mmol/l and inorganic phosphate decreased to 1.42 mmol/l (Fig. 2A). Urinary calcium and phosphate levels were monitored (Fig. 2B). Decreased LH and FSH were also observed (Fig. 2C). The hemoglobin level increased to 10.2 g/dl (Fig. 2D).

Variants in AIRE

A heterozygous single-base-pair variation in exon 5 (NM_000383.2: c.623G>T) of AIRE was identified, causing an amino acid change of glycine to valine at position 208 (NP_000374.1: p.Gly208Val) (Fig. 1B). This variant was predicted to be pathogenic, with scores of 1.00 by PolyPhen-2 and 0.00 by SIFT. The present study also identified a sequence alteration (NM_000383.2: c.371C>T) in exon 3 of AIRE, which caused substitution of proline to leucine at position 124 (NP_000374.1: p.Pro124Leu) (Fig. 1C). This variant was predicted to be benign, with scores of 0.00 by PolyPhen-2 and 1.00 by SIFT. Moreover, the affected amino acid residue, p.Gly208, was conserved among different species, indicating the functional importance of this amino acid residue throughout evolution (Fig. 1D). The 124th amino acid of the AIRE protein varied between species, indicating that this residue may be less important for the function and structure of the protein (Fig. 1E).

Both sequence alterations were present in the patient's father but were absent in the mother and the 500 unaffected controls (Fig. 1B and C). Searching the HGMD (hgmd.cf.ac.uk/ac/index.php), Leiden Open Variation Database 3.0 (lovd.nl/3.0/home), PubMed database, the 1000 Genomes Project (browser.1000genomes.org) and Exome Aggregation Consortium database (exac.broadinstitute.org/) indicated that c.623G>T was a novel variant.

The change in the 3D structure of AIRE protein induced by the pathogenic variant present in the patient is presented in Fig. 3A. The glycine to valine substitution at position 208 changes the structure of Sp100, AIRE-1, NucP41/75, DEAF-1 (SAND).

Discussion

The diagnosis of APS-1 is primarily based on clinical manifestations such as hypoparathyroidism, primary adrenocortical insufficiency and chronic mucocutaneous candidiasis. More recent clinical studies have demonstrated that APS-1 can be classified into classical and non-classical type (18,19). Classical APS-1 is characterized by autosomal recessive inheritance and the presence of at least two of the three main features of hypoparathyroidism, primary adrenocortical insufficiency and chronic mucocutaneous candidiasis. Non-classical APS-1 follows an autosomal dominant inheritance pattern and presents with varying late-onset autoimmune phenotypes (1821). To the best of our knowledge, the present study is the first to report a first Chinese patient with non-classical APS-1 caused by a novel variant in AIRE in an autosomal dominant fashion. At present, there are 15 known variants of AIRE that follow an autosomal dominant inheritance pattern (including those identified in the present study) (18,19). The majority of variants are clustered within the first plant homeodomain (PHD1) zinc finger domain (18). While two variants (p.Gly208Val and p.Gly228Trp) were in the SAND domain, one variant (p.Cys446Gly) was in the second plant homeodomain (PHD2) zinc finger domain (18,19,20). The majority of patients with a dominant inheritance often did not match the classical diagnostic criteria of APS-1. These patients exhibited more variable phenotypes, ranging from no autoimmunity to late-onset classical APS-1, and hypoparathyroidism was the most prevalent phenotype (18,19). The present patient suffered from tetany and syncope, primary amenorrhea, intermittent diarrhea and general fatigue. Upon laboratory examination, she was diagnosed as having hypoparathyroidism, primary ovarian insufficiency and PRCA. A novel heterozygous variant of c.623G>T in exon 5 was identified and regarded as a pathogenic variant of non-classical APS-1. Another reported variant c.371C>T in exon 3 of AIRE was also identified.

The AIRE gene encodes a 57-kDa protein that promotes T-cell tolerance to self-antigens by upregulating the expression levels of tissue-specific self-antigens in medullary thymic epithelial cells (22). Impaired functional AIRE leads to the escape of self-reactive T-lymphocytes to the periphery and induces pathogenic autoimmune reactions (23). Proper subcellular location is important for the function of AIRE and its ability to maintain macromolecular interactions. Previous reports have shown that non-mutated AIRE localizes to the nucleus, whereas mutant AIRE is retained in the cytoplasm, where it is unable to regulate the transcription of a number of genes, such as major histocompatibility complex class I and class II gene products, and causes abnormal immune responses such as impaired negative selection of autoreactive thymocytes (16,2325).

AIRE contains a number of domains, including a highly conserved caspase recruitment domain/N-terminal homogenously staining region (CARD/HSR) domain, a conserved bipartite nuclear localization signal (NLS), a SAND domain, two plant PHD zinc-finger motifs separated by a proline-rich region, and four LXXLL (L, leucine; X, any amino acid) nuclear receptor-binding motifs (26). The SAND domain is important for mediating the nuclear localization of AIRE (27). The variant c.623G>T in exon 5 may change the structure of the SAND domain, resulting in aggregation of AIRE in the cytoplasm and impaired DNA binding (Fig. 3A). However, p.Pro124Leu caused by c.371C>T was between bipartite of NLS, which constituted amino acids 110–114 and 131–33 (28), which may explain why this variant was not pathogenic (Fig. 3B).

A previous study demonstrated that AIRE variants in patients with hypoparathyroidism may induce autoantibodies to NACHT leucine-rich-repeat protein 5, which is selectively expressed in chief cells of the parathyroid glands (29). An autoimmune reaction targeting the calcium-sensing receptor was also found in parathyroid cells of patients with APS-1 (30). In addition, a loss of T-lymphocyte infiltration was observed surrounding follicles in female AIRE-knockout mice (22). These studies helped to elucidate the potential pathogenesis of the AIRE variant underlying APS-1.

APS-1 is more frequent in European countries, with a prevalence of 1:25,000 in Finland, 1: 14,000 in Sardinia, 1:43,000 in Slovenia and 1:500,000 in France (4,6,24). p.R257X, located in the SAND domain, is the most common variant in Finland, Russia and Eastern Europe (4,6,24). R139X is most frequent in patients from Sardinia and results in more severe phenotypes of APS-1 (10). The present study adds the clinical and genetic characteristics of the patient to those of six previously reported Chinese patients with APS-1 (7,1417). A total of 8 variants (7 missense variants and 1 insertion variant) were detected in seven patients with APS-1 from six unrelated families (Table II). Previous research has revealed that the most common variants of the AIRE gene exist in exons 1, 2, 6, 8 and 10 (24,31), whereas the variants of the AIRE gene in Chinese patients with APS-1occurred in exons 1–6 (c.55G>A, c.206A>C, c.371C>T, c.463G>A, c.483_484insC, c.623G>T and c.769C>T), without obvious hotspot variants. These variants primarily affected the CARD, NLS and SAND domains (Fig. 3B). Variants in the CARD domain may influence the multimerization of the AIRE protein and suppress its transcriptional activation capacity (32). A number of gross deletions and splice site variants (such as c.483_484insC in patient 2) have been identified as associated with APS-1 because AIRE protein translation is affected (33,34).

Table II.

Summary of clinical and genetic features of 7 Chinese patients with APS-1 and AIRE mutations.

Table II.

Summary of clinical and genetic features of 7 Chinese patients with APS-1 and AIRE mutations.

Gender/onsetManifestations


Patient no.age, yearsMajor componentsOther componentsAIRE mutation/inheritanceProtein changeDomain(Refs.)
1F/4CMC; HP; PAINoneExon 1: c.55G>A/ARp.Ala19ThrCARD(14)
Exon 6: c.769C>T/ARp.Arg257XSAND
2F/10CMC; PAIAutoimmune thyroiditisExon 4: c.483_484insC/ARTruncated proteinCARD(16)
3M/2HP; PAIEnamel dystrophy; binocular cataract; chronic intestinal dysfunctionExon 2: c.206A>C/ARp.Gln69ProSAND(7)
4aM/8HP; PAIEpilepsy; pernicious anemia; chronic/tension headaches; keratopathy; type 1 diabetes mellitusExon 3: c.463G>A/ARp.Gly155SerNA(15)
5aF/1CMC; HP; PAIEpilepsy; pernicious anemiaExon 3: c.463G>A/ARp.Gly155SerNA(15)
6F/12CMC; HPAnemiaExon 5: c.622G>T/NAp.Gly208TrpSAND(17)
7F/7HPPrimary ovarian insufficiency;Exon 3: c.371C>T/ADp.Pro124LeuNot domainThis
Pure red cell aplasia; chronic atrophic gastritisExon 5: c.623G>T/ADp.Gly208ValSANDstudy

a siblings. F, female; M, male; CMC, chronic mucocutaneous candidiasis; HP, hypoparathyroidism; PAI, primary adrenocortical insufficiency; Ala, alanine; Thr, threonine; Arg, arginine; Gln, glutanine; Pro, proline; Gly, glycine; Ser, serine; Trp, tryptophan; Leu, leucine; Val, valine; AR, autosomal recessive; AD, autosomal dominant; CARD, caspase recruitment domain; SAND, Sp100, AIRE-1, NucP41/75, DEAF-1; NA, not available.

Regarding the clinical phenotypes of Chinese patients, the classic triad of CMC, PAI and HP were observed in two patients, with four patients exhibiting two of the three major manifestations. Generally, CMC is the most common, with an occurrence of 77–100% in European patients with APS-1 (3,3537), but it is rare in Iranian Jews (17%) (11). CMC was only detected in four Chinese patients with APS-1 and hypoparathyroidism was the most common manifestation (six cases).

In patients with APS-1, the same pathogenic gene can lead to different clinical phenotypes. A Chinese patient with APS-1 was diagnosed as having APS-1 without CMC, but his sister exhibited this symptom (15). Cetani et al (19), reported a missense (p.Gly228Trp) mutation of the AIRE gene in a family with APS-1. The p.Gly228Trp mutation was detected in the proband as well as her mother, son and sister, and acted in a dominant manner. However, only the proband and her sister exhibited the typical components of APS-1. In another study, a proband and four of her children carried the p.Cys311Tyr variant. The proband and three of her children exhibited varying components and severity of APS-1, whereas a son was diagnosed with autoantibodies against tyrosine hydroxylase without any autoimmune manifestations (18). The patient's father exhibited the same AIRE variants, but he did not present with symptoms of APS-1. The mechanism governing the difference in penetration rates is not clear in patients with APS-1.

The diagnosis of APS-1 is often delayed (2). The present patient presented with major manifestations of hypoparathyroidism and other late-onset autoimmune phenotypes, which was inconsistent with the phenotypes of classic APS-1. However, APS-1 was confirmed by the detection of AIRE variants. Thus, AIRE sequencing should be performed at an early stage when patients are suspected of having APS-1.

There is currently a lack of studies concerning the effects of immunomodulatory and gene-targeted therapy on APS-1. The primary therapeutic strategy for APS-1 is hormone replacement therapy to correct endocrine function. For hypoparathyroidism, supplementation with calcium and calcitriol is given to achieve low-normal serum calcium and normal urine calcium levels, relieve the symptoms of hypocalcemia and decrease long-term complications (38). Hormone replacement therapy is an alternative treatment used to manage primary ovarian insufficiency. Cyclosporine A, with or without concurrent corticosteroids, has been suggested to be an effective immunotherapy for pure red aplastic anemia (39). Using these approaches, the clinical symptoms of the present patient notably improved.

There were a number of limitations in in the present study. First, the genotype-phenotype association was not well established due to the small sample size. Second, the lack of functional studies made it difficult to elucidate the detailed mechanism underlying how variants in AIRE lead to APS-1. Third, certain autoantibodies, such as 21-hydroxylase and steroid-producing cells autoantibodies, were not investigated. Last, detailed information on the patient's father was not available.

In conclusion, a novel variant of c.623G>T (p.Gly208Val) of the AIRE gene is associated with an APS-1 clinical syndrome of hypoparathyroidism, primary ovarian insufficiency and PRCA. The novel missense variant of c.623G>T in AIRE may impair its binding and regulation of the macromolecular DNA binding complex, resulting in a clinical picture of APS-1. The results of the present study expand the known genetic and phenotypic spectra of APS-1.

Acknowledgements

Not applicable.

Funding

The present study was supported by the National Natural Science Foundation of China (grant nos. 81570802 and 81873668, respectively), Chinese Academy of Medical Sciences Innovative Fund for Medical Sciences (grant no. 2016-I2M-3-003) and the National Key Research and Development Program of China (grant no. 2016YFC0901501).

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

WBZ performed molecular genetic studies, participated in sequence alignment, analyzed data and wrote the manuscript. LJL and DCZ collected, analyzed and interpreted the data.. OW, YJ and WBX provided important medical decisions in the clinical diagnose and treatment of patient, contributed the analysis of clinical data and reviewed the manuscript. ML contributed to the conception and design of the research, acquisition and interpretation of data and revised the manuscript. All authors read and approved the final manuscript.

Ethics approval and consent to participate

The study was approved by the Scientific Ethics Committee of Peking Union Medical College Hospital (approval no. JS-2081). Written informed consent was obtained from all subjects prior to participation.

Patient consent for publication

The patient, her parents and the healthy controls provided informed consent for publication of their data.

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

CMC

chronic mucocutaneous candidiasis

PAI

primary adrenocortical insufficiency

HP

hypoparathyroidism

PRCA

pure red cell aplasia

Cr

creatine

ALT

alanine aminotransferase

PTH

parathyroid hormone

FSH

follicle-stimulating hormone

LH

luteinizing hormone

FT4

free thyroxine

FT3

free triiodothyronine

TSH

thyroid-stimulating hormone

TPOAb

autoantibodies to thyroid peroxidase antigen

TGAb

autoantibodies to thyroglobulin

ACTH

adrenocorticotropic hormone

ICA

islet cell antibodies

GADA

glutamic acid decarboxylase antibodies

ANA

autoantibodies to antinuclear antibody

RO-52

52 kDa Ro/SS-A molecule

ANCA

anti-neutrophil cytoplasmic antibodies

References

1 

Sepe V, Velluzzi F and Songini M: Autoimmune polyendocrine syndromes. N Engl J Med. 378:25432018.PubMed/NCBI

2 

Husebye ES, Anderson MS and Kämpe O: Autoimmune polyendocrine syndromes. N Engl J Med. 378:1132–1141. 2018. View Article : Google Scholar : PubMed/NCBI

3 

Bruserud Ø, Oftedal BE, Landegren N, Erichsen MM, Bratland E, Lima K, Jørgensen AP, Myhre AG, Svartberg J, Fougner KJ, et al: A longitudinal follow-up of autoimmune polyendocrine syndrome type 1. J Clin Endocrinol Metab. 101:2975–2983. 2016. View Article : Google Scholar : PubMed/NCBI

4 

Orlova EM, Sozaeva LS, Kareva MA, Oftedal BE, Wolff ASB, Breivik L, Zakharova EY, Ivanova ON, Kämpe O, Dedov II, et al: Expanding the phenotypic and genotypic landscape of autoimmune polyendocrine syndrome type 1. J Clin Endocrinol Metab. 102:3546–3556. 2017. View Article : Google Scholar : PubMed/NCBI

5 

Gutierrez MJ, Gilson J, Zacharias J, Ishmael F and Bingham CA: Childhood polyarthritis as early manifestation of autoimmune polyendocrinopathy with candidiasis and ectodermal dystrophy syndrome. Front Immunol. 8:3772017. View Article : Google Scholar : PubMed/NCBI

6 

Bruserud Ø, Oftedal BE, Wolff AB and Husebye ES: AIRE-mutations and autoimmune disease. Curr Opin Immunol. 43:8–15. 2016. View Article : Google Scholar : PubMed/NCBI

7 

Zhu W, Hu Z, Liao X, Chen X, Huang W, Zhong Y and Zeng Z: A new mutation site in the AIRE gene causes autoimmune polyendocrine syndrome type 1. Immunogenetics. 69:643–651. 2017. View Article : Google Scholar : PubMed/NCBI

8 

Nagamine K, Peterson P, Scott HS, Kudoh J, Minoshima S, Heino M, Krohn KJ, Lalioti MD, Mullis PE, Antonarakis SE, et al: Positional cloning of the APECED gene. Nat Genet. 17:393–398. 1997. View Article : Google Scholar : PubMed/NCBI

9 

Zhao B, Chang L, Fu H, Sun G and Yang W: The role of autoimmune regulator (AIRE) in peripheral tolerance. J Immunol Res. 2018:39307502018. View Article : Google Scholar : PubMed/NCBI

10 

Meloni A, Willcox N, Meager A, Atzeni M, Wolff AS, Husebye ES, Furcas M, Rosatelli MC, Cao A and Congia M: Autoimmune polyendocrine syndrome type 1: An extensive longitudinal study in Sardinian patients. J Clin Endocrinol Metab. 97:1114–1124. 2012. View Article : Google Scholar : PubMed/NCBI

11 

Zlotogora J and Shapiro MS: Polyglandular autoimmune syndrome type I among Iranian Jews. J Med Genet. 29:824–826. 1992. View Article : Google Scholar : PubMed/NCBI

12 

Rosatelli MC, Meloni A, Meloni A, Devoto M, Cao A, Scott HS, Peterson P, Heino M, Krohn KJ, Nagamine K, et al: A common mutation in Sardinian autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy patients. Hum Genet. 103:428–434. 1998. View Article : Google Scholar : PubMed/NCBI

13 

Liu Y, Asan, Ma D, Lv F, Xu X, Wang J, Xia W, Jiang Y, Wang O, Xing X, et al: Gene mutation spectrum and genotype-phenotype correlation in a cohort of Chinese osteogenesis imperfecta patients revealed by targeted next generation sequencing. Osteoporos Int. 28:2985–2995. 2017. View Article : Google Scholar : PubMed/NCBI

14 

Liu C, Shi Y, Yin H, Li H, Fan S, Wu S and Yuan P: Autoimmune regulator gene mutations in a Chinese family with autoimmune polyendocrinopathy syndrome type I. Zhonghua Yi Xue Yi Chuan Xue Za Zhi. 27:18–22. 2010.(In Chinese). PubMed/NCBI

15 

Zhang J, Liu H, Liu Z, Liao Y, Guo L, Wang H, He L, Zhang X and Xing Q: A functional alternative splicing mutation in AIRE gene causes autoimmune polyendocrine syndrome type 1. PLoS One. 8:e539812013. View Article : Google Scholar : PubMed/NCBI

16 

Jin P, Zhang Q, Dong CS, Zhao SL and Mo ZH: A novel mutation in autoimmune regulator gene causes autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy. J Endocrinol Invest. 37:941–948. 2014. View Article : Google Scholar : PubMed/NCBI

17 

Sun YX, He YF and Li XL: Clinical analysis and autoimmune regulator gene mutation of autoimmune polyendocrinopathy syndrome type I in a family: A report of one case. Zhongguo Dang Dai Er Ke Za Zhi. 18:147–151. 2016.(In Chinese). PubMed/NCBI

18 

Oftedal BE, Hellesen A, Erichsen MM, Bratland E, Vardi A, Perheentupa J, Kemp EH, Fiskerstrand T, Viken MK, Weetman AP, et al: Dominant mutations in the autoimmune regulator AIRE are associated with common organ-specific autoimmune diseases. Immunity. 42:1185–1196. 2015. View Article : Google Scholar : PubMed/NCBI

19 

Cetani F, Barbesino G, Borsari S, Pardi E, Cianferotti L, Pinchera A and Marcocci C: A novel mutation of the autoimmune regulator gene in an Italian kindred with autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy, acting in a dominant fashion and strongly cosegregating with hypothyroid autoimmune thyroiditis. J Clin Endocrinol Metab. 86:4747–4752. 2001. View Article : Google Scholar : PubMed/NCBI

20 

Ilmarinen T, Eskelin P, Halonen M, Ruppell T, Kilpikari R, Torres GD, Kangas H and Ulmanen I: Functional analysis of SAND mutations in AIRE supports dominant inheritance of the G228W mutation. Hum Mutat. 26:322–331. 2005. View Article : Google Scholar : PubMed/NCBI

21 

Su MA, Giang K, Zumer K, Jiang H, Oven I, Rinn JL, Devoss JJ, Johannes KP, Lu W, Gardner J, et al: Mechanisms of an autoimmunity syndrome in mice caused by a dominant mutation in Aire. J Clin Invest. 118:1712–1726. 2008. View Article : Google Scholar : PubMed/NCBI

22 

Anderson MS, Venanzi ES, Klein L, Chen Z, Berzins SP, Turley SJ, von Boehmer H, Bronson R, Dierich A, Benoist C and Mathis D: Projection of an immunological self shadow within the thymus by the aire protein. Science. 298:1395–1401. 2002. View Article : Google Scholar : PubMed/NCBI

23 

Kisand K and Peterson P: Autoimmune polyendocrinopathy candidiasis ectodermal dystrophy: Known and novel aspects of the syndrome. Ann N Y Acad Sci. 1246:77–91. 2011. View Article : Google Scholar : PubMed/NCBI

24 

Björses P, Halonen M, Palvimo JJ, Kolmer M, Aaltonen J, Ellonen P, Perheentupa J, Ulmanen I and Peltonen L: Mutations in the AIRE gene: Effects on subcellular location and transactivation function of the autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy protein. Am J Hum Genet. 66:378–392. 2000. View Article : Google Scholar : PubMed/NCBI

25 

Johnnidis JB, Venanzi ES, Taxman DJ, Ting JP, Benoist CO and Mathis DJ: Chromosomal clustering of genes controlled by the aire transcription factor. Proc Natl Acad Sci USA. 102:7233–7238. 2005. View Article : Google Scholar : PubMed/NCBI

26 

Peterson P, Org T and Rebane A: Transcriptional regulation by AIRE: Molecular mechanisms of central tolerance. Nat Rev Immunol. 8:948–957. 2008. View Article : Google Scholar : PubMed/NCBI

27 

Ramsey C, Bukrinsky A and Peltonen L: Systematic mutagenesis of the functional domains of AIRE reveals their role in intracellular targeting. Hum Mol Genet. 11:3299–3308. 2002. View Article : Google Scholar : PubMed/NCBI

28 

Ilmarinen T, Melen K, Kangas H, Julkunen I, Ulmanen I and Eskelin P: The monopartite nuclear localization signal of autoimmune regulator mediates its nuclear import and interaction with multiple importin alpha molecules. FEBS J. 273:315–324. 2006. View Article : Google Scholar : PubMed/NCBI

29 

Alimohammadi M, Björklund P, Hallgren A, Pöntynen N, Szinnai G, Shikama N, Keller MP, Ekwall O, Kinkel SA, Husebye ES, et al: Autoimmune polyendocrine syndrome type 1 and NALP5, a parathyroid autoantigen. N Engl J Med. 358:1018–1028. 2008. View Article : Google Scholar : PubMed/NCBI

30 

Habibullah M, Porter JA, Kluger N, Ranki A, Krohn KJE, Brandi ML, Brown EM, Weetman AP and Kemp EH: Calcium-sensing receptor autoantibodies in patients with autoimmune polyendocrine syndrome type 1: Epitopes, specificity, functional affinity, IgG subclass, and effects on receptor activity. J Immunol. 201:3175–3183. 2018. View Article : Google Scholar : PubMed/NCBI

31 

Meloni A, Perniola R, Faà V, Corvaglia E, Cao A and Rosatelli MC: Delineation of the molecular defects in the AIRE gene in autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy patients from Southern Italy. J Clin Endocrinol Metab. 87:841–846. 2002. View Article : Google Scholar : PubMed/NCBI

32 

Pitkänen J, Doucas V, Sternsdorf T, Nakajima T, Aratani S, Jensen K, Will H, Vähämurto P, Ollila J, Scott HS, et al: The autoimmune regulator protein has transcriptional transactivating properties and interacts with the common coactivator CREB-binding protein. J Biol Chem. 275:16802–16809. 2000. View Article : Google Scholar : PubMed/NCBI

33 

Cihakova D, Trebusak K, Heino M, Fadeyev V, Tiulpakov A, Battelino T, Tar A, Halasz Z, Blümel P, Tawfik S, et al: Novel AIRE mutations and P450 cytochrome autoantibodies in Central and Eastern European patients with APECED. Hum Mutat. 18:225–232. 2001. View Article : Google Scholar : PubMed/NCBI

34 

Bøe Wolff AS, Oftedal B, Johansson S, Bruland O, Lovas K, Meager A, Pedersen C, Husebye ES and Knappskog PM: AIRE variations in Addison's disease and autoimmune polyendocrine syndromes (APS): Partial gene deletions contribute to APS I. Genes Immun. 9:130–136. 2008. View Article : Google Scholar : PubMed/NCBI

35 

Wolff AS, Erichsen MM, Meager A, Magitta NF, Myhre AG, Bollerslev J, Fougner KJ, Lima K, Knappskog PM and Husebye ES: Autoimmune polyendocrine syndrome type 1 in Norway: Phenotypic variation, autoantibodies, and novel mutations in the autoimmune regulator gene. J Clin Endocrinol Metab. 92:595–603. 2007. View Article : Google Scholar : PubMed/NCBI

36 

Perheentupa J: Autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy. J Clin Endocrinol Metab. 91:2843–2850. 2006. View Article : Google Scholar : PubMed/NCBI

37 

Ahonen P, Myllärniemi S, Sipilä I and Perheentupa J: Clinical variation of autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy (APECED) in a series of 68 patients. N Engl J Med. 322:1829–1836. 1990. View Article : Google Scholar : PubMed/NCBI

38 

Brandi ML, Bilezikian JP, Shoback D, Bouillon R, Clarke BL, Thakker RV, Khan AA and Potts JT Jr: Management of hypoparathyroidism: Summary statement and guidelines. J Clin Endocrinol Metab. 101:2273–2283. 2016. View Article : Google Scholar : PubMed/NCBI

39 

Means RT Jr: Pure red cell aplasia. Blood. 128:2504–2509. 2016. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

August-2020
Volume 22 Issue 2

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zheng WB, Li LJ, Zhao DC, Wang O, Jiang Y, Xia WB and Li M: A novel variant in AIRE causing a rare, non‑classical autoimmune polyendocrine syndrome type 1. Mol Med Rep 22: 1285-1294, 2020
APA
Zheng, W., Li, L., Zhao, D., Wang, O., Jiang, Y., Xia, W., & Li, M. (2020). A novel variant in AIRE causing a rare, non‑classical autoimmune polyendocrine syndrome type 1. Molecular Medicine Reports, 22, 1285-1294. https://doi.org/10.3892/mmr.2020.11227
MLA
Zheng, W., Li, L., Zhao, D., Wang, O., Jiang, Y., Xia, W., Li, M."A novel variant in AIRE causing a rare, non‑classical autoimmune polyendocrine syndrome type 1". Molecular Medicine Reports 22.2 (2020): 1285-1294.
Chicago
Zheng, W., Li, L., Zhao, D., Wang, O., Jiang, Y., Xia, W., Li, M."A novel variant in AIRE causing a rare, non‑classical autoimmune polyendocrine syndrome type 1". Molecular Medicine Reports 22, no. 2 (2020): 1285-1294. https://doi.org/10.3892/mmr.2020.11227