Skip to main content
Erschienen in: Arthritis Research & Therapy 1/2018

Open Access 01.12.2018 | Letter

Juvenile onset autoinflammatory disease due to a novel mutation in TNFAIP3 (A20)

verfasst von: Shuzo Sato, Yuya Fujita, Tomonari Shigemura, Hisanori Matoba, Kazunaga Agematsu, Yuya Sumichika, Makiko Yashiro, Atsushi Ono, Yukihiko Kawasaki, Hiroko Kobayashi, Hiroshi Watanabe, Tomohiro Koga, Atsushi Kawakami, Kiyoshi Migita

Erschienen in: Arthritis Research & Therapy | Ausgabe 1/2018

Hinweise

Electronic supplementary material

The online version of this article (https://​doi.​org/​10.​1186/​s13075-018-1766-x) contains supplementary material, which is available to authorized users.
Abkürzungen
HA20
Haploinsufficiency of A20
IKK
Inhibitor of nuclear factor kappa B kinase
IL
Interleukin
IκB-α
Inhibitor of nuclear factor kappa B alpha
PBMC
Peripheral blood mononuclear cell
TNF
Tumour necrosis factor

Key message

Novel heterozygous C200A A20/TNFAIP3 gene mutation is responsible for autosomal-dominant juvenile onset autoinflammatory disorder.

Letter

A20, which is encoded by the TNFAIP3 gene, has been shown to control nuclear factor kappa B (NF-κB) signalling by deubiquitinating receptor-interacting proteins [1]. Recently, heterozygous mutations in the TNFAIP3 gene have been found to cause the haploinsufficiency of A20, which presents as an early-onset autoinflammatory disease [2, 3]. Here, we report a Japanese family containing two cases of autoinflammatory disease which exhibit an identical novel TNFAIP3 mutation.
The patient was a 17-year-old Japanese boy who was referred to our department with a 3-year history of recurrent painful oral ulcer and epigastralgia, accompanied by low-grade fever. He presented with periodic fever and oral aphtha that had occurred for the prior 3 years and was referred to our hospital. After spontaneous resolution of the symptoms, he was regularly followed in our hospital. Three years later, the patient was admitted to our hospital for impaired food intake due to epigastralgia, oral ulcer, and fever. His mother had exhibited oral and genital ulcers and erythema nodosum-like lesions on the upper extremities and had died of septicaemia 2 years prior. His younger sister exhibited similar symptoms from the age of 11 years and was suspected of Behçet’s disease. Physical examination of the boy revealed painful oral ulcers. Neither genital ulcer nor uveitis was confirmed, but multiple erosions in his stomach mucosa were observed by endoscopic examination.
Relevant laboratory data are listed in Additional file 1 (Table S1). There were no abnormalities except elevated levels of C-reactive protein (CRP). Anti-nuclear antibodies were positive with low titres; however, tests for other various autoantibodies revealed no abnormalities. The HLA-B haplotype of the patient was not B51 (B39/B62). Increased serum inflammatory cytokine levels were confirmed at the time of presentation (Additional file 1: Table S1). Although autoimmune phenomena were not prominent, this patient presented with an autoinflammatory phenotype resembling Behçet’s disease. Treatment with prednisolone relieved the clinical symptoms and normalized CRP levels.
Recently, Zhou et al. reported 11 patients from six families with a new dominantly inherited autoinflammatory disease, termed haploinsufficiency of A20 (HA20) [4]. This disease has the following characteristics: periodic fever; oral, ocular, and/or genital ulcers; and elevated levels of many cytokines [4]. Serum pro-inflammatory cytokines, particularly tumour necrosis factor (TNF)-α, interleukin (IL)-1β, IL-6, and IL-8, were elevated in our patient (Additional file 1: Table S1). Therefore, written informed consent for TNFAIP3 gene analysis was obtained from the patient and his father.
The C596_598 del A mutation, which we confirmed by Sanger sequencing (Fig. 1a), introduces a frameshift substitution of alanine for cysteine at position 200, generating a downstream stop codon (Cys200Alafsx16; C200Afs*16) in the OTU domain of A20. This heterozygous C200A fs*16 mutation was present in the patient’s younger sister, who showed similar symptoms. Sanger sequencing of the unaffected father showed no variant of TNFAIP3, suggesting that these mutations were maternally derived. This variant is absent from public databases (ExAs/dbSNP).
To address the molecular basis of this C200Afs*16 mutation, we stimulated peripheral blood mononuclear cells (PBMCs) that were isolated from the patient and from a healthy subject with TNF-α (10 ng/ml), and then performed immunoblot blot analysis with anti-A20 antibodies (Fig. 1b). Wild-type A20 protein expression was reduced in patient-derived PBMCs compared with those from the healthy subject at baseline and after TNF-α stimulation. These results suggested that the frameshift mutation identified in the present case resulted in impaired expression of A20 at baseline and after TNF-α stimulation. Additionally, cytosolic inhibitor of nuclear factor kappa B alpha (IκB-α) protein levels were reduced in patient-derived PBMCs after stimulation with TNF-α, whereas IκB-α protein levels were unchanged in control PBMCs. These findings suggest that IκB-α degradation was accelerated by TNF-α stimulation in PBMCs isolated from the patient.
A20 is a ubiquitin-editing enzyme that inhibits key proinflammatory molecules, including inhibitor of nuclear factor kappa B kinase (IKK)γ [5]. Zhou et al. reported that unrelated family cases manifested autoinflammation with heterozygous mutations in TNFAIP3; all patients exhibited oral and genital ulcers [4]. The clinical manifestations seen in our patients were consistent with those of the previously reported cases [6], whereas the disease onset (at 14 years old) was relatively late compared with those of the reported cases [6]. We identified a novel heterozygous missense mutation, which was located at the OTU domain of A20 in both affected siblings in this family.
In previously reported HA20 cases, mutant cells have shown enhanced NF-κB activity, as demonstrated by increased phosphorylation of IKKα/β and increased degradation of IκB-α [7]. In the PBMCs isolated from our patient, cellular IκB-α failed to maintain baseline levels compared with cells expressing wild-type A20 in response to extended TNF-α stimulation. IKKα/β mediated phosphorylation of IκB-α resulted in IκB-α degradation and subsequent nuclear translocation of NF-κB [8]. These findings suggest that haploinsufficiency of TNFAIP3, which leads to impaired A20 expression, could be responsible for the TNF-α-induced increased degradation of IκB-α in the present case. Although the molecular pathogenesis of our patient and previously reported cases may be similar, variations in clinical manifestations suggest additional modifying factors [6]. Further studies regarding functional and genetic analysis of A20 will elucidate the pathogenesis of A20-mediated autoimmune/autoinflammatory diseases.

Acknowledgements

Not applicable.

Funding

The study was supported by the Practical Research Project for Rare/Intractable Diseases from the Japanese Agency for Medical Research and Development (AMED).

Availability of data and materials

Please contact the corresponding author for data requests.
Ethical approval for this study (no. 2920) was provided by the Ethics Committee of Fukushima Medical University and written informed consent was obtained from each individual.
Written informed consents were obtained from all subjects for publication of this case report.

Competing interests

KM has received research grants from Chugai, Pfizer, and AbbVie. The remaining authors declare that they have no competing interests.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.
Literatur
1.
2.
Zurück zum Zitat Aeschlimann FA, Batu ED, Canna SW, Go E, Gül A, Hoffmann P, et al. A20 haploinsufficiency (HA20): clinical phenotypes and disease course of patients with a newly recognised NF-kB-mediated autoinflammatory disease. Ann Rheum Dis. 2018;77(5):728–35.CrossRefPubMed Aeschlimann FA, Batu ED, Canna SW, Go E, Gül A, Hoffmann P, et al. A20 haploinsufficiency (HA20): clinical phenotypes and disease course of patients with a newly recognised NF-kB-mediated autoinflammatory disease. Ann Rheum Dis. 2018;77(5):728–35.CrossRefPubMed
3.
Zurück zum Zitat Shigemura T, Kaneko N, Kobayashi N, Kobayashi K, Takeuchi Y, Nakano N, et al. Novel heterozygous C243Y A20/TNFAIP3 gene mutation is responsible for chronic inflammation in autosomal-dominant Behçet's disease. RMD Open. 2016;2(1):e000223.CrossRefPubMedPubMedCentral Shigemura T, Kaneko N, Kobayashi N, Kobayashi K, Takeuchi Y, Nakano N, et al. Novel heterozygous C243Y A20/TNFAIP3 gene mutation is responsible for chronic inflammation in autosomal-dominant Behçet's disease. RMD Open. 2016;2(1):e000223.CrossRefPubMedPubMedCentral
4.
Zurück zum Zitat Zhou Q, Wang H, Schwartz DM, Stoffels M, Park YH, Zhang Y, et al. Loss-of-function mutations in TNFAIP3 leading to A20 haploinsufficiency cause an early-onset autoinflammatory disease. Nat Genet. 2016;48(1):67–73.CrossRefPubMed Zhou Q, Wang H, Schwartz DM, Stoffels M, Park YH, Zhang Y, et al. Loss-of-function mutations in TNFAIP3 leading to A20 haploinsufficiency cause an early-onset autoinflammatory disease. Nat Genet. 2016;48(1):67–73.CrossRefPubMed
5.
Zurück zum Zitat Wertz IE, O'Rourke KM, Zhou H, Eby M, Aravind L, Seshagiri S, et al. De-ubiquitination and ubiquitin ligase domains of A20 downregulate NF-kappaB signalling. Nature. 2004;430(7000):694–9.CrossRefPubMed Wertz IE, O'Rourke KM, Zhou H, Eby M, Aravind L, Seshagiri S, et al. De-ubiquitination and ubiquitin ligase domains of A20 downregulate NF-kappaB signalling. Nature. 2004;430(7000):694–9.CrossRefPubMed
6.
Zurück zum Zitat Kadowaki T, Ohnishi H, Kawamoto N, Hori T, Nishimura K, Kobayashi C, et al. Haploinsufficiency of A20 causes autoinflammatory and autoimmune disorders. J Allergy Clin Immunol. 2018;141(4):1485–8.CrossRefPubMed Kadowaki T, Ohnishi H, Kawamoto N, Hori T, Nishimura K, Kobayashi C, et al. Haploinsufficiency of A20 causes autoinflammatory and autoimmune disorders. J Allergy Clin Immunol. 2018;141(4):1485–8.CrossRefPubMed
7.
Zurück zum Zitat Duncan CJA, Dinnigan E, Theobald R, Grainger A, Skelton AJ, Hussain R, et al. Early-onset autoimmune disease due to a heterozygous loss-of-function mutation in TNFAIP3 (A20). Ann Rheum Dis. 2018;77(5):783–6.CrossRefPubMed Duncan CJA, Dinnigan E, Theobald R, Grainger A, Skelton AJ, Hussain R, et al. Early-onset autoimmune disease due to a heterozygous loss-of-function mutation in TNFAIP3 (A20). Ann Rheum Dis. 2018;77(5):783–6.CrossRefPubMed
Metadaten
Titel
Juvenile onset autoinflammatory disease due to a novel mutation in TNFAIP3 (A20)
verfasst von
Shuzo Sato
Yuya Fujita
Tomonari Shigemura
Hisanori Matoba
Kazunaga Agematsu
Yuya Sumichika
Makiko Yashiro
Atsushi Ono
Yukihiko Kawasaki
Hiroko Kobayashi
Hiroshi Watanabe
Tomohiro Koga
Atsushi Kawakami
Kiyoshi Migita
Publikationsdatum
01.12.2018
Verlag
BioMed Central
Erschienen in
Arthritis Research & Therapy / Ausgabe 1/2018
Elektronische ISSN: 1478-6362
DOI
https://doi.org/10.1186/s13075-018-1766-x

Weitere Artikel der Ausgabe 1/2018

Arthritis Research & Therapy 1/2018 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.