Skip to main content
Clinical and Experimental Immunology logoLink to Clinical and Experimental Immunology
. 2023 Jan 18;212(2):117–128. doi: 10.1093/cei/uxad006

Mistuned NF-κB signaling in lymphocytes: lessons from relevant inborn errors of immunity

Gina Dabbah-Krancher 1,2,3, Andrew L Snow 4,5,
PMCID: PMC10128170  PMID: 36651621

Summary

Inborn errors of immunity (IEIs) continuously remind us that multiple checks and balances are built into the adaptive immune system to maintain homeostasis, ensuring effective pathogen defense without causing inadvertent immunopathology, autoimmunity, or lymphomagenesis. The nuclear factor of κB (NF-κB) family of transcription factors serve a vital role in the immune system, inducing scores of genes responsible for lymphocyte survival, proliferation, differentiation and effector function. In recent years, the discovery and characterization of IEIs that impact NF-κB activity have illuminated the importance of carefully tuning this pathway to ensure effective immune defense without hyperinflammation and immune dysregulation. Here we examine several illustrative cases of IEIs that arise from pathogenic mutations encoding NF-κB inducers, regulators, and NF-κB family components themselves, illuminating how these genes ensure normal adaptive immune system function by maintaining a “Goldilocks effect” state in NF-κB pathway activity.

Keywords: inborn errors of immunity, lymphocytes, NF-κB


This review discusses human inborn errors of immunity caused by genetic variants that disrupt normal NF-κB signaling, primarily in T and B lymphocytes. The discovery and investigation of these disorders underscores how mistuned NF-κB signaling can contribute to both immune deficiency and dysregulation.

Graphical Abstract

Graphical Abstract.

Graphical Abstract

Introduction

Inborn errors of immunity (IEI) represent a group of ~430 genetic disorders that result in increased susceptibility to infections, autoimmune disorders, autoinflammatory diseases, allergy, and/or malignancy [1–3]. While some IEIs simply present as pure primary immune deficiencies (PIDs) with specific and/or recurrent infections, other gene defects result in broader phenotypic spectra of immune dysregulation better classified as primary immune regulatory disorders (PIRDs) [4]. Accessible next-generation sequencing (NGS) technology has enabled rapid identification of novel genetic variants, making the diagnosis of once rare disorders more commonplace, with an aggregated prevalence between 1/1000 and 1/5000 individuals [5]. Monogenic germline mutations can result in loss of protein expression and/or loss-of-function (LOF, amorphic/hypomorphic) or gain-of-function (GOF, hypermorphic), often inherited as autosomal recessive (AR) or dominant (AD) traits, respectively. A heterozygous variant may also trigger AD disease by exhibiting dominant-negative (DN) interference or haploinsufficiency. Studies linking molecular mechanisms of pathogenesis to the broad range of clinical outcomes associated with different monogenic disorders have helped establish the foundations of human immunology, revealing non-redundant roles for specific genes in immune function, and exposing avenues for targeted therapeutics. Herein we explore the crucial role of NF-κB in lymphocyte function and immune homeostasis, informed by IEIs caused by mutations that perturb an otherwise finely tuned signaling pathway.

The NF-κB family is a complex master regulator of gene transcription in hematopoietic cells, held in latency in the cytoplasm until the engagement of a relevant receptor (e.g. pattern recognition receptor (PRR), TNF superfamily, antigen receptor (AgR)) triggers nuclear translocation, and transcriptional activity. Originally discovered as a nuclear factor binding near the κ light-chain gene in B cells [6], active NF-κB binds to -κB consensus sites found in gene promoters and enhancers. Dimeric NF-κB complexes are assembled from five monomeric subunits comprising the Rel transcription factor family: NFKB1/p105/p50, NFKB2/p100/p52, RELA/Rel A (p65), RELB/Rel B, and REL/c-Rel [7]. These proteins can homo- and hetero-dimerize to create 15 different NF-κB complexes that control the transcription of many genes responsible for cell survival, proliferation, and immune function. Each subunit contains a 300 amino acid Rel homology domain (RHD) that allows for the sequence-specific DNA binding, dimerization, and inhibitory protein binding, as well as a nuclear localization sequence (NLS) [6, 8, 9]. However, p100 and p105 do not contain transactivation (TA) domains, and therefore must dimerize with other REL proteins to drive transcriptional activity [6, 9]. The canonical NF-κB pathway utilizes p105/p50, Rel A(p65), and c-Rel, whereas the alternative pathway utilizes p100/p52 and Rel B [7] (Fig. 1A). Although numerous IEIs have been linked to gene variants affecting both pathways and summarized in detail elsewhere [6], this review primarily focuses on both PIDs and PIRDs linked to defects in canonical NF-κB signaling and their effects on adaptive immunity (Table 1).

Figure 1:

Figure 1:

(A) Schematic diagram of canonical (e.g. TCR) versus alternative (e.g. CD40) NF-κB signaling pathways in lymphocytes. (B) Illustration of ‘Goldilocks’ principle of tuned NF-κB signaling to maintain immune homeostasis. Too little or too much NF-κB signaling in leukocytes can manifest in specific gene defects which give rise to primary immune deficiency (PID) and/or primary immune regulatory disorder (PIRD) phenotypes.

Table 1:

Clinical and laboratory abnormalities found in monogenic IEIs associated with reduced or excessive NF-κB signaling in lymphocytes

Reduced NF-κB signaling
Gene Protein Effect Inheritance Major clinical presentation Other clinical symptoms Laboratory abnormalities
NFKB1 P105/P50 LOF AD (haploinsuff) CVID Respiratory/GI infections, autoimmunity, lymphoproliferation Low Ig, low CS/memory B cells, low Tregs
NFKB2 P100/P52 LOF AD CVID, adrenal insufficiency Autoimmunity Low Ig, low CS/memory B cells
RELB RELB LOF AR CID Autoimmunity, FTT Impaired T/B cell development, dysplastic thymus
NFKBIA IκB GOF AD EDA-ID Recurrent pyogenic infections Dysgammaglobulinemia, low memory T cells, poor T cell Ag responses
IKBKG NEMO LOF XL EDA-ID IP, lethal herpesvirus infections, autoinflammation Low Ig, low NK cell function, poor inflammatory cytokine production
IKBKB IKKβ LOF AR SCID Poor T/B cell Ag responses, low memory T/B cells
CARD11 CARD11
CARMA1
LOF AR SCID P. jirovecii pneumonia Low Ig, low memory T/B cells, low Tregs, poor T/B cell Ag responses
CARD11 CARD11 CARMA1 DN AD Severe atopic disease Recurrent respiratory/cutaneous viral infections, autoimmunity Low Ig, low CS/memory B cells, poor T cell Ag responses, Th2 skewing, high IgE, eosinophilia
BCL10 BCL10 LOF AR SCID Recurrent respiratory viral infections, oral candidiasis Low memory T/B cells, absent Tregs, poor T/B cell Ag responses
MALT1 MALT1 LOF AR CID + IPEX-like Eczema, enteropathy, FTT, periodontal disease Very low Tregs, poor T cell responses to Ag
Excessive NF-κB signaling
IKBKB IKKβ GOF AD Mild CID Inflammation, epithelial defects Lymphopenia, low Ig, increased/normal Tregs, increased T cell Ag response
CARD11 CARD11 CARMA1 GOF AD Polyclonal B cell lymphocytosis Splenomegaly, mild recurrent infections, Low CS/memory B cells, low T cell responses to Ag
TNFAIP3 A20 LOF AD (haploinsuff) Behcet-like autoinflammation Retinal scarring, oral ulcers, skin abscesses, fevers Elevated inflammatory cytokines (e.g. IL-1β), Th9/17 skewing

Abbreviations: AD: autosomal dominant; Ag: antigen; AR: autosomal recessive; CID: combined immunodeficiency; CS: class-switched; CVID: common variable immunodeficiency; EDA-ID: anhidrotic ectodermal dysplasia with immunodeficiency; FTT: failure to thrive; IPEX: immune dysregulation, polyendocrinopathy, enteropathy, X-linked); XL: X-linked.

CARD11 (caspase recruitment domain family member 11) encodes a scaffold protein primarily expressed in lymphocytes that bridges AgR ligation with the activation of downstream signaling pathways including NF-κB, c-Jun N-terminal kinase (JNK), and mechanistic target of rapamycin (mTOR) [7, 10–13]. Proximal AgR triggers assembly of the ‘CBM complex’ comprised of CARD11, BCL10 (B cell lymphoma 10), and MALT1 (mucosa-associated lymphoid tissue lymphoma translocation protein 1), a pivotal regulator of lymphocyte proliferation and effector function [11, 14]. The importance of the CBM complex in T/B cell function was experimentally established when lymphocytes from mice deficient in BCL10, MALT1, or CARD11 all displayed impaired NF-κB activation, survival, proliferation, and cytokine secretion. These abnormalities were later confirmed by the discovery of human patients carrying pathogenic mutations in each component, manifesting in both immunodeficiency and/or immune dysregulation phenotypes collectively referred to as ‘CBM-opathies’ [15]. In recent years, our group and others have described a rapidly expanding group of IEIs linked to impactful CARD11 variants, all of which impact canonical NF-κB signaling.

The CARD11 scaffold is built from modular domains that allow for AgR-triggered BCL10 interaction (CARD domain), oligomerization (LATCH, coiled-coil), and higher order multimerization at the plasma membrane (membrane associated guanylate kinase (MAGUK) domain). Proximal B-cell receptor (BCR) and T-cell receptor (TCR) signaling is highly symmetrical, culminating in the phosphorylation of a complex inhibitory linker domain that normally retains CARD11 in an inactive state in resting lymphocytes [16, 17]. This derepression converts CARD11 to an open conformation allowing for pre-associated BCL10-MALT1 units to bind and oligomerize to initiate CBM complex formation (Fig. 1A) [18–21]. This dynamic, filamentous signalosome then acts as a recruiting platform for downstream signaling partners including the LUBAC (linear ubiquitin chain assembly complex) and an E3 ligase, TRAF6 [13, 22–24]. The LUBAC complex facilitates the physical recruitment of the IKK complex (IKKα-IKKβ-IKKγ) through IKKγ (also known as NEMO or NF-κB essential modifier), a noncatalytic regulatory subunit of the complex, and two kinase subunits IKKα and IKKβ, to ubiquitinated MALT1 [7, 25, 26]. TRAF6 polyubiquitinates MALT1 further by adding K63-linked ubiquitin chains, which recruit the TAB2/3-TAK1 complex to phosphorylate and activate the neighboring IKK complex partners IKKα/β [22, 23, 27–29]. IKK-mediated phosphorylation triggers proteasomal degradation of IκBα, exposing the NLS sequence on Rel family proteins to release NF-κB dimers into the nucleus. p105 is processed and partially degraded to a p50 subunit that can interact with other Rel family members (e.g. p65) and translocate to the nucleus to stimulate gene transcription (Fig. 1A) [6]. Extensive studies of both murine models and IEIs (including CBM-opathies) underscore this NF-κB transcriptional program must be carefully calibrated to ensure lymphocyte homeostasis, including negative feedback loops that promptly return NF-κB to latency.

The NF-κB dependent gene tumor necrosis factor alpha-induced protein 3 (TNFAIP3) encodes A20, one such important negative regulator of NF-κB signaling that restricts excessive inflammation [30]. A20 expression may be induced in numerous cell types by proinflammatory pathways that activate NF-κB [30]. Indeed, genome-wide association studies (GWAS) linked single nucleotide polymorphisms (SNPs) in TNFAIP3 to various autoimmune inflammatory disorders, including rheumatoid arthritis, systemic lupus erythematosus, psoriasis, and inflammatory bowel disease (IBD) [31–34]. A20 was initially discovered to be a ubiquitin-editing enzyme, responsible for modulating the TNF signaling cascade through cleavage of K63-ubiquitin chains and addition of K48-ubiquitin to degrade participating proteins and terminate signaling [35, 36]. In T cells, A20 can cleave K63-linked ubiquitin chains attached to MALT1 via its deubiquitinase (DUB) activity to disrupt and downregulate NF-κB signaling (Fig. 1A) [37].

In this review, we primarily focus on the consequences of ‘mistuned’ NF-κB signaling in lymphocytes by examining illustrative IEIs that arise from mutations in NF-κB genes themselves (NFKB1/2), as well as key upstream activators (CBM complex genes, IKBKB, NFKBIA) and a crucial downstream regulator (TNFAIP3). We will review our current understanding of the genetics and pathophysiology associated with each disorder, resulting in clinical phenotypes ranging from pure immunodeficiency to complex immune dysregulation. In doing so, we reference relevant biochemical studies and murine models and highlight critical knowledge gaps that invite further mechanistic studies.

Too little NF-κB signaling

NFKB1 LOF

Monoallelic mutations in NFKB1 usually manifest as common variable immunodeficiency (CVID), one of the most commonly diagnosed IEI associated with increased susceptibility to respiratory infections, sometimes accompanied by other sequalae characteristic of a PIRD [38]. CVID is a clinically and genetically heterogeneous disorder linked to variants in at least 22 genes, although ~90% of CVID-like conditions remain genetically undefined [39]. CVID is characterized by hypogammaglobulinemia (with low IgG, IgM, and IgA), impaired antibody production (including after vaccination), and abnormal lymphocyte phenotypes including low class-switched memory B cells and regulatory T cells (Tregs), impaired natural killer (NK) cell maturation and cytotoxicity, and aberrations in T and B cell maturation and function [3, 40–45]. Milder specific antibody deficiencies restricted to IgA or IgG subclasses have been noted in relatives with or without clinical symptoms. Indeed, incomplete clinical penetrance (~70%) and age-dependent phenotypic severity has been reported, with autoimmunity also noted in a subset of patients [46]. Until recently, the mode of autosomal dominant inheritance associated with NFKB1-driven CVID was poorly understood. However, a recent report shed light on this mechanism by utilizing an NF-κB reporter assay that specifically tested p65:p50 heterodimer-dependent transcription [47]. Among 60 predicted LOF NFKB1 variants tested in this assay, 80% were confirmed LOF/hypomorphic with no evidence of DN activity, strongly suggesting haploinsufficiency as the explanation for AD NFKB1 deficiency. LOF haploinsufficiency results in partial reduction of both p105 and p50 in resting lymphocytes, with severely reduced p105 phosphorylation after stimulation. In the aforementioned study, all deleterious mutations were found in the RHD, where they were shown to disrupt dimerization, nuclear localization, DNA binding, and interaction with IκB proteins. However, mutations have been reported throughout the protein, including those localized to the C-terminus degron that disrupt p105 phosphorylation and processing to p50 [48]. These heterozygous LOF mutations have been shown to affect protein stability, subunit phosphorylation, and nuclear translocation, which likely contributes to variability in clinical phenotypes [49].

NFKB2 LOF

Although we focus here on canonical NF-κB signaling associated with AgR-signaling in lymphocytes, a growing collection of deleterious mutations identified in both NFKB2 and RELB illuminate how impaired alternative NF-κB signaling also manifests in CVID-like phenotypes in humans [50, 51]. NFKB2 deficient patients also suffer from recurrent respiratory infections in early childhood, but ~50% also develop pituitary hormone deficiencies and central adrenal insufficiency, distinguishing this IEI from NFKB1 deficiency [52]. Autoimmune manifestations affecting skin, hair, and nails are also observed in some patients; breakdown of tolerance affecting the endocrine system has been postulated but not proven. Recently discovered RELB-deficient patients share similar infectious susceptibility and humoral immune defects (despite normal serum Ig), consistent with a CID involving impaired T and B cell differentiation and reduced thymic output [53]. Reduced BAFF-R and CD40-induced alternative NF-κB signaling may account for poor B cell maturation, whereas the mechanism explaining thymic dysplasia and a skewed oligoclonal T cell compartment remains mysterious. Homozygous RELB LOF mutations result in total absence of RELB expression, whereas various LOF (−/+ DN activity) and GOF NFKB2 mutations have been described, often with incomplete penetrance [50, 54]. Indeed, two people harboring the same NFKB2 mutation can present differently, highlighting gaps in our understanding of alternative NF-κB signaling and the influence of other genetic and environmental modifiers—a common theme among many IEIs discussed here. These NFKB2 and RELB mutations have variable detrimental effects on T- and B-cell development, activation, proliferation, and survival [50, 53, 55]. Interestingly, NFKB1 and NKFB2 mutations typically manifest with similar CVID-like symptoms despite the divergent pattern of LOF mutations observed. While NFKB1 mutations affect residues throughout the p105/p50 protein, NFKB2 mutations are typically localized to the p100 degron, interrupting the ability of p100 to be degraded and converted to p52 therefore decreasing alternative NF-κB signaling [51, 56]. The inability to degrade p100 can also lead to increased binding to p65 and dominant interference of canonical NF-κB signaling as well, which may help to explain overlapping defects in lymphocyte differentiation and function [57]. Indeed, LOF mutations in MAP3K14 (NIK) cause CID due to defects in both canonical and alternative NF-κB signaling [58].

NFKBIA GOF

Among the IκB family, IκBα (encoded by NFKBIA) is the only member to date linked to a human monogenic IEI. Since 2003, 20 patients have been identified with heterozygous GOF NFKBIA mutations, resulting in autosomal dominant inheritance [59]. Patients typically present with profound CID, recurrent pyogenic and mycobacterial infections, and dysgammaglobulinemia, with most suffering from anhidrotic ectodermal dysplasia with immunodeficiency (EDA-ID). Most NFKBIA GOF mutations are either missense variants that disrupt the phosphorylation of residues Ser32 and Ser36, or N-terminal protein truncations removing these serine residues altogether. Although IκBα protein is expressed, these mutations abrogate stimulation-induced phosphorylation and Ub-mediated degradation, enhancing inhibitory function by remaining constitutively bound to NF-κB [60]. Patient T cells respond poorly to antigens, resulting in no detectable memory T cells despite significant naive lymphocytosis. Impaired TLR, TNFR, and CD40 signaling are also observed. Patients harboring one of four mutations resulting in early stop codons presented early in life with EDA-ID and similar infectious manifestations. Hematopoietic stem cell transplantation (HSCT) remains the only viable option for correcting hematopoietic deficits in these patients [59].

IKBKG LOF

Notably, X-linked EDA-ID can also arise from hemizygous hypomorphic mutations in NEMO/IKBKG, manifesting in similar infection susceptibility with milder T cell immunodeficiency relative to NFKBIA GOF patients [61]. NEMO is critical for relaying signals downstream of AgRs, TNFR, TLR3, and RIG-I-like receptors (RLRs), resulting in activation of both NF-κB and IRF3. Similar to other genes discussed herein, the phenotypic breadth of clinical symptoms varies depending on the location and potency of IKBKG mutations, ranging from PID- to PIRD-like presentations [62]. Whereas N-terminal deletions disrupt IKKα/β association to drive incontentia pigmenti, CID, and lethal herpesvirus infections, C-terminal deletions can also trigger hyperactive TLR/TNFR signaling and autoinflammatory disease due to abrogated recruitment of the negative regulator A20 (see below) [63, 64]. Intriguingly, a new PIRD dubbed NEMO deleted exon 5 autoinflammatory syndrome (NDAS) was recently linked to alternative splicing mutations that specifically remove exon 5, which contributes to differential signal dysregulation from RLRs and TNF in dermal fibroblasts versus peripheral blood mononuclear cells (PBMC) [65].

IKBKB LOF

IKBKB encodes IKKβ, a key subunit of the IKK complex responsible for driving canonical NF-κB activity. Two homozygous IKBKB LOF variants have been reported since 2013, including a missense mutation (Y395H) and a homozygous G duplication in exon 13, resulting in a frameshift starting at amino acid 432 (Gln432Profs*62). Patients harboring these variants presented early in life with bacterial, viral, fungal, and mycobacterial infections. Despite normal T and B cell development, AgR-induced responses are severely impaired, resulting in a severe combined immunodeficiency (SCID) phenotype [66, 67]. Without IKKβ, normal T/B cell activation, proliferation, and differentiation into effector and memory cells are blocked, explaining an overabundance of naïve cells. Eight patients received HSCT, but only three survived [67]. PBMCs from Y395H carriers showed degradation of IKKβ protein with normal levels of IKKα and IKKγ, resulting in reduced canonical NF-κB activation [68]. Gln432Profs*62 results in premature stop codon and results in complete loss of IKKβ protein. In these patients’ cells, IKKα and NEMO/IKKγ also showed decreased protein levels despite normal mRNA expression, implying IKKβ has some stabilizing effect on the IKK complex. Importantly, patient T- and B cells showed decreased responses to stimulation through TLRs, cytokine receptors, and mitogens as well, highlighting that both innate and adaptive immune cells are impacted in this PID [69].

CBM deficiency: SCID

Before human CBM variants were characterized, several groups demonstrated that murine knockouts of Card11, Bcl10, or Malt1 all resulted in profound T and B cell activation defects and panhypogammaglobulinemia despite normal lymphocyte development. More recently discovered CBM deficiencies in CID patients largely mirror these PID-like phenotypes. Five cases of germline homozygous LOF mutations in CARD11 have been reported in humans [70–73]. All patients were born to consanguineous parents and presented early in life with severe respiratory tract infections and pneumonia typically caused by Pneumocystis jirovecii. CARD11 mutations were localized to either the CC or MAGUK domains including Phe902_Glu946del, Gln945*, Cys150*, and Arg837* [72], often preserving the expression of a truncated but non-functional protein. Despite normal B and T cell frequencies, patients displayed increased numbers of transitional B cells, decreased (class-switched) memory B cells, predominant naïve T cells with severely reduced Tregs, and pan-hypogammaglobulinemia. As expected, patient lymphocytes exhibited diminished AgR-induced activation including decreased p65 phosphorylation and impaired IκBα degradation, decreased IL-2 production, and poor T cell proliferation. In one case, a somatic reversion (Cys150Leu) partially restored CARD11 function in a proportion of conventional T cells, manifesting in Omenn syndrome [70]. A handful of patients described with BCL10 or MALT1 deficiency (due to AR LOF variants) present with a highly similar CID phenotype, as well as immune dysregulation marked by GI and respiratory tract inflammation, likely driven by a striking death of Tregs [15]. In fact, MALT1 deficiency resembles the PIRD known as immune dysregulation, polyendocrinopathy, enteropathy, X-linked (IPEX) syndrome (e.g. eczema, enteropathy, and failure to thrive), with periodontal disease included as an unusual phenotypic twist. Curative HSCT is recommended for any CBM-deficient patient, with a relatively high success rate observed to date.

CARD11 DN: CADINS

CARD11-associated atopy with dominant interference of NF-κB Signaling (CADINS) patients have germline heterozygous LOF, DN mutations in CARD11 that manifest in severe atopic disease and CID [74–76]. To date, over 70 DN LOF CARD11 mutations have been uncovered in a rapidly expanding patient cohort. Roughly 90% present with allergic inflammation, most commonly atopic dermatitis followed by asthma, rhinitis, food allergies, or eosinophilic esophagitis, aligning with increased IgE and eosinophilia in most patients [15]. About two-thirds of patients also present with serious cutaneous viral infections (e.g. HSV-1, molluscum) and respiratory tract infections. Smaller subsets of patients also present with autoimmunity and malignancy, further classifying CADINS as a true PIRD. Upon TCR stimulation, patient T cells show decreased activation of NF-κB and mTORC1, reduced proliferation, and Th2-skewed cytokine production (elevated IL-4/IL-13, decreased IFN-γ) [74]. Most mutations are localized to the CARD and CC domains, impeding CBM complex assembly by disrupting BCL10-MALT1 binding and oligomerization post-AgR engagement. Although attenuated TCR-induced NF-κB activity helps to confirm diagnosis, effects on other downstream transcription factors (e.g. AP-1) activation remain an open question.

Before the discovery of CADINS disease, Goodnow et al. described ‘unmodulated’ mice harboring a homozygous CC Card11 mutation that developed dermatitis and high IgE levels with age [77]. More recently, a new mouse model expressing a homologous Card11 R30W mutation identified in five separate CADINS patients demonstrated similar T and B cell defects in heterozygosity [78]. Mouse CD4+ T cells showed diminished IL-2 and IFN-γ production and decreased CD25 upregulation after AgR stimulation, consistent with impaired NF-κB signaling (decreased IKKα/β phosphorylation and IκBα degradation). Milder B cell activation defects were also apparent (e.g. reduced CD86 upregulation) without an overt IκBα degradation defect, suggesting differential signaling impacts in T and B cells. Most interestingly, aged R30W mice exhibited elevated serum IgE without any signs of Th2 skewing or atopic disease, suggesting other genetic and environmental triggers in CADINS pathogenesis. Although Treg frequency and function are compromised in both mouse models [78, 79], this does not appear to translate to CADINS patients [76]. Recently, Bedsaul et al. proposed a ‘CARD11 oligomer poisoning’ model to describe how LOF CARD11 mutants can dominantly interfere with wild-type (WT) CARD11 in mixed oligomers during two distinct steps of the signaling cascade; either when CARD11 converts to an open conformation and/or interacts with cofactors (BCL10, HOIL-1) necessary for downstream signaling [10].

Too much NF-κB signaling

IKBKB GOF

V203I is the only known IKBKB GOF mutation in humans, manifesting as CID in two unrelated families. All affected patients presented with lymphopenia, recurrent respiratory infections, and hypogammaglobulinemia, with some exhibiting signs of inflammation as well. This mutation in the kinase domain results in increased IκBα and p65 phosphorylation in lymphocytes at baseline and post-AgR stimulation. Whereas both CD4 and CD8 T cells were hyperactivated, B cells also showed decreased proliferation and antibody production following stimulation. Although disease is less severe than the complete LOF of IKBKB, the fact that both LOF and GOF mutations confer immune deficiency underscores the importance of properly tuned NF-κB responses for effective adaptive immunity [80].

CARD11 GOF (BENTA)

B-cell Expansion with NF-κB and T cell Anergy (BENTA) disease is a PIRD caused by inborn heterozygous, GOF mutations in CARD11 and generally results in a mild immunodeficiency [81]. To date, 11 validated GOF mutations have been documented in human patients, largely confined to the LATCH and CC domains [15, 82–84]. BENTA patients present with selective, polyclonal B cell lymphocytosis and splenomegaly early in life, commonly combined lymphadenopathy, and recurrent sinopulmonary infections. Lab findings include increased immature/transitional (CD10+), naïve mature (IgM+IgD+) polyclonal B cells, and decreased class-switched and memory B cells [15]. Autoantibodies have been observed in some BENTA patients, which may relate to enhanced survival of self-reactive B cells [85]. Although autoimmunity is not observed in most BENTA patients, autoimmune cytopenias, atopic inflammation, and severe immune dysregulation (e.g. hemophagocytic lymphohistiocytosis) have been observed in recent case reports [83, 86, 87]. Surprisingly, patient T cells are hyporesponsive in culture with decreased proliferation and IL-2 secretion in response to TCR stimulation, appearing mildly anergic [81]. This phenomenon may help to explain more viral infections in some patients, including disseminated molluscum and moderate EBV viremia.

GOF mutations disrupt complex repressive elements in the inhibitory linker domain, forcing CARD11 into an open conformation [16]. This results in the spontaneous formation of cytoplasmic aggregates called mutant CARD11-dependent shells (mCADS) that include MALT1 and active IKKα/β that drive constitutive NF-κB signaling independent of AgR-engagement, although these aggregates are more dynamic in primary patient T/B cells [88]. Interestingly, one recent mouse study highlighted a role for Card11 in non-canonical regulation of the AKT-FOXO1 signaling axis, independent of NF-κB activation, utilizing the GOF Card11 mutants K215M and E134G [89]. Moreover, the contribution of dysregulated JNK or mTORC1 signaling to clinical pathogenesis remains largely unexplored. Collectively, these threads emphasize a need to investigate how CARD11 mutants affect other crucial non-NF-κB signaling pathways in lymphocytes, keeping in mind that T- and B-cell signaling is not necessarily symmetrical.

BENTA patient B cells have an inherent survival advantage in vitro relative to healthy controls, providing a plausible explanation for B cell accumulation in vivo [90]. This survival advantage appears to be dependent on MALT1 protease activity (unpublished data). As in B cell lymphomas harboring identical somatic CARD11 GOF mutations [91], constitutive NF-κB activation is thought to drive the expansion of the B cell compartment via enhanced survival and resistance to apoptosis. Currently, the role of negative regulators of NF-κB is unknown in BENTA patient lymphocytes. Based on our preliminary data, we hypothesize that the survival advantage in BENTA B cells is linked to a loss of A20-dependent negative regulation, resulting from constitutive cleavage of A20 by active MALT1 paracaspase (unpublished data). Indeed, genetic deletion of A20 is frequently found in B cell lymphomas with elevated NF-κB activity, and ectopic restoration of A20 expression induces apoptosis [92, 93]. CARD11 mutations highlight the importance of elucidating the impact of a novel variants on the function of the encoded proteins and thus the mechanism of pathogenicity.

TNFAIP3 LOF: A20 haploinsufficiency

Protein ubiquitination is a key post-translational modification necessary for regulating signal transduction in diverse inflammatory pathways. Ubiquitin (Ub) chains can form through any of ubiquitin’s seven lysines, allowing for polyubiquitin linkages needed for signal amplification (K63- and M1-linked ubiquitin) or proteasomal degradation of proteins for signal termination (K48-linked ubiquitin). TNFAIP3 encodes A20, a dual function enzyme built from an N-terminal OTU (ovarian tumor) domain with DUB activity, and seven C-terminal zinc finger (ZnF) domains which possess both E3 ligase activity (ZnF4) and Ub-binding capacity (ZnF7) [30]. ZnF4 has a monoubiquitin-binding domain (UBD) and E3 ligase activity which can add K48-linked ubiquitin to the direct destruction of NF-κB signaling intermediates [94]. ZnF7 was shown to inhibit LUBAC-induced NF-κB activation by binding to M1-linked (linear) ubiquitin [95, 96]. However, these experiments were conducted primarily in TNF-stimulated HEK293 cell and did not address MALT1-dependent cleavage of A20 in lymphocytes. Therefore, it is unknown if these mechanisms are involved in regulating AgR-induced NF-κB activity in lymphocytes.

A20 haploinsufficiency (HA20) is observed in patients with heterozygous LOF mutations in TNFAIP3 [97–100]. Patient PBMCs show markedly reduced A20 protein expression and increased NF-κB activation at baseline, with sustained signaling post-TNF stimulation. HA20 patients suffer early in life from retinal scarring and fibrosis, oral ulcers, and dermal lesions/abscesses; periodic fevers and other gastrointestinal and musculoskeletal autoinflammatory manifestations are also reported with remarkable variability. These diverse sequelae are largely explained by the loss of A20-dependent negative regulation of proinflammatory innate signaling pathways. Interestingly, intracellular cytokine staining of patient PBMCs stimulated with LPS or staphylococcal enterotoxin B (SEB) showed increased polarization toward CD4+ Th9 and Th17 effector cell lineages [97]. Similarly, when A20 was silenced in IBD and Vogt-Koyanagi-Harada (VKH) patients, CD4+ T cells shifted toward a Th1 and Th17 phenotype [101]. These data highlight A20 as a crucial mediator of inflammation and NF-κB signaling in both innate and adaptive immunity.

A20 is constitutively expressed in lymphocytes, with pleiotropic effects in controlling B and T cell function [30]. A20 is the most frequently inactivated gene in B-cell lymphomas, establishing A20 as a critical tumor suppressor gene [92, 93]. Compagno et al. documented biallelic inactivation of TNFAIP3 in ~30% of diffuse large B-cell lymphomas (DLBCL). Moreover, exogenous A20 expression in lymphoma-derived DLBCL lines lacking A20 resulted in suppression of cell growth, induction of apoptosis, and NF-κB downregulation. In addition, germline and somatic genetic TNFAIP3 variations were observed in 77% of patients with mucosa-associated lymphoma associated with primary Sjogren’s syndrome [102, 103]. Congruently, mice lacking A20 expression in B cells exhibited spontaneous B cell activation, expansion of germinal center B cells, and production of autoantibodies [104–106]. Loss of A20 in aged murine B cells was shown to lower their activation threshold, enhance proliferation and survival, and increase NF-κB signaling [104]. A20-deficient B cells also exhibit resistance to Fas-mediated apoptosis, likely related to enhanced expression of anti-apoptotic proteins [106]. These data collectively suggest that A20 acts to restrict B cell growth, survival, and oncogenesis by facilitating cell death, including in autoreactive B cells.

The role of A20 in regulating T cell development and activation is hazy and still under investigation. In mice, A20-deleted CD8+ T cells show heightened sensitivity to AgR stimulation, with increased NF-κB and production of IL-2 and IFN-γ [107]. Although these mice exhibit some lymphadenopathy and organ infiltration, there was no detectable pathology. Increased NF-κB activity in adoptively transferred A20-deficient CD8+ T cells confers enhanced tumor suppressive activity, with improved control of melanoma growth, enhanced production of IFN-γ and TNFα, and reduced expression of programmed cell death 1 (PD-1), making A20 a potential candidate for T cell-directed immunotherapy [108]. Moreover, A20 expression supported secondary, but not primary, T cell responses to Listeria monocytogenes infection [109]. In contrast, A20-deficient CD4+ T cells are susceptible to necroptosis and exhibit decreased autophagy and mTOR activity post-stimulation [110]. A20 also restrains intrathymic differentiation of Tregs and NKT cells [111, 112]. Whereas A20 functions to restrict B cell proliferation and survival, A20 appears to foster proper T cell development and survival, tuning of TCR signaling, and production of memory cells. Considering patients harboring LOF mutations in either CARD11 or A20 display abnormal T cell differentiation and immune dysregulation resulting in clinical disease, further work on these patients should shed new light on the importance of A20 in tuning signals downstream of the CBM complex to maintain immune homeostasis.

The precise mechanism by which A20 regulates AgR-induced NF-κB activity remains nebulous. Previous work suggested that MALT1 initially cleaves A20 in the local CBM complex environment shortly after TCR engagement, allowing signal propagation [37, 113]. NF-κB then induces A20 synthesis to dampen subsequent CBM signaling by cleaving K63-linked ubiquitin chains attached to MALT1 via its DUB activity, ostensibly breaking MALT1-IKK interactions to downregulate further NF-κB signaling [37]. Subsequent work suggests DUB activity is dispensable for A20 function in T cells [114]. In a recent study, Yin et al. demonstrated that A20 and A20-binding inhibitors of NF-κB-1 (ABIN-1) work in concert to modulate T cell activation [115]. A20 utilizes ABIN-1 as an adaptor to engage with the CBM complex and tune both initial and sustained bursts of TCR-induced NF-κB activity. This suggests that A20 and ABIN-1 are recruited to the CBM complex early on to create a threshold that TCR activation has to overcome to allow signal transduction to occur. In the final phases of sustained CBM signaling, the authors conclude that resynthesized A20 is cleaved by MALT1 to allow for signal transduction to proceed. Despite this new insight, it remains unclear which domains of A20 are essential to its localization and regulatory function, including crosstalk with other signaling proteins post-TCR engagement.

Conclusion

The discovery of several IEIs linked to monogenic variants affecting key signaling nodes in AgR-induced NF-κB activation and resolution have yielded fascinating insights into how this pathway is finely tuned to maintain immune homeostasis, effecting multiple aspects of lymphocyte biology. While too little NF-κB activity is clearly associated with impaired lymphocyte responses and PID, too much NF-κB activity often gives rise to immune dysregulation, sometimes accompanied by infectious susceptibilities noted in several PIRDs [4, 38]. Even though pathogenesis often encompasses innate immune dysregulation and non-hematopoietic phenotypes, lymphocytes remain a dominant context for understanding variations in clinical expressivity that result from mistuned NF-κB signaling. Laboratory-based screening of primary patient lymphocytes can often yield valuable diagnostic insights to decipher the impact of novel gene variants. For example, acute aberrations in mitogen or AgR-induced canonical NF-κB signaling can be captured in intracellular flow cytometric assays for differential phosphorylation and degradation of IκB, or phosphorylation/nuclear translocation of p65 [76, 116]. Comparable defects in alternative NF-κB signaling pathways (p100 processing, RELB nuclear translocation) may also be detected with appropriate stimuli (e.g. CD40L). Given the limited or oscillatory nature of NF-κB signaling enforced by negative feedback loops (e.g. IκB/A20 upregulation), more subtle defects may be difficult to ascertain without optimizing the potency and timing of AgR stimulation. Hence definitive diagnoses may require empirical testing of ectopically-expressed mutant gene constructs in relevant in vitro assays that encompass both acute signal transduction events and downstream reporters of NF-κB-dependent transcriptional activity, as we and others have employed [10, 47, 75, 81].

Effective therapeutic strategies for treating these disorders can be complicated, arguably requiring the identification of targets that restore NF-κB activity to equilibrium without overshooting in either direction (Fig. 1B). For example, MALT1 protease inhibitors offer a strategy for turning down AgR-dependent NF-κB signaling in relevant PIRDs (e.g. BENTA) without compromising activity entirely, and yet data from mice and recent human trials revealed unexpected impacts on Treg function that contribute to autoimmune manifestations [15, 117]. Hopefully the accelerating discovery of new IEIs connected to altered NF-κB activity will implicate novel regulatory mechanisms and unveil new therapeutic avenues for restoring balance in this essential pathway.

Acknowledgements

We thank Dr Brian Schaefer for helpful discussions. The opinions and assertions expressed herein are those of the authors and are not to be construed as reflecting the views of Uniformed Services University of the Health Sciences or the United States Department of Defense.

Glossary

Abbreviations

AD

autosomal dominant

AgR

antigen receptor

AR

autosomal recessive

BCL10

B cell lymphoma 10

BCR

B cell receptor

BENTA

B cell expansion with NF-κB and T cell anergy

CADINS

CARD11-associated atopy with dominant interference of NF-κB signaling;

CARD11

caspase activation and recruitment domain 11

CVID

common variable immunodeficiency

DLBCL

diffuse large B cell lymphoma

DN

dominant negative

DUB

deubiquitinase

EDA-ID

ectodermal dysplasia with immunodeficiency

GOF

gain-of-function

GWAS

genome wide association study

HSCT

hematopoietic stem cell transplantation

IBD

inflammatory bowel disease

IEI

inborn errors of immunity

IKK

IκB kinase

JNK

c-Jun N-terminal kinase

LOF

loss-of-function

LUBAC

linear ubiquitin chain assembly complex

MALT1

mucosa-associated lymphoid tissue lymphoma translocation protein 1

mTOR

mechanistic target of rapamycin

NDAS

NEMO deleted exon 5 autoinflammatory syndrome

NF-κB

nuclear factor of kappa B

NGS

next generation sequencing

NLS

nuclear localization sequence

PBMC

peripheral blood mononuclear cells

PID

primary immune deficiency

PIRD

primary immune regulatory disorder

PRR

pattern recognition receptor

RHD

Rel homology domain

RLR

RIG-I-like receptor

SCID

severe combined immunodeficiency

SNP

single nucleotide polymorphism

TA

transactivation

TCR

T cell receptor

TLR

Toll-like receptor

TNF

tumor necrosis factor

ZnF

zinc finger

Contributor Information

Gina Dabbah-Krancher, Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD, USA; Emerging Infectious Diseases Graduate Program, Uniformed Services University of the Health Sciences, Bethesda, MD, USA; Henry M. Jackson Foundation, Bethesda, MD, USA.

Andrew L Snow, Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD, USA; Emerging Infectious Diseases Graduate Program, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.

Funding

This work was supported by a Specific Defect Research Program grant from the Jeffrey Modell Foundation (A.L.S.).

Conflict of interest

The authors declare no conflicts of interest.Author Contributions

Conception: G.D. and A.L.S. Figure preparation: G.D. Writing and literature review were conducted by both authors. Critical revisions were made by A.L.S. Both authors read, edited, and approved the final version of the article.

Animal Research

ARRIVE guidelines: Not applicable.

Data Availability

Not applicable—no new data were created or analyzed.

Permission to reproduce

Government license—A.L.S. is an employee of the U.S. Government, and this article was written as part of his employment.

Clinical trial registration

Not applicable.

References

  • 1. Casanova JL, Abel L.. Lethal infectious diseases as inborn errors of immunity: toward a synthesis of the germ and genetic theories. Annu Rev Pathol 2021, 16, 23–50. doi: 10.1146/annurev-pathol-031920-101429. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2. Notarangelo LD, Bacchetta R, Casanova JL, Su HC.. Human inborn errors of immunity: an expanding universe. Sci Immunol 2020, 5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. Tangye SG, Al-Herz W, Bousfiha A, Cunningham-Rundles C, Franco JL, Holland SM, et al. Human inborn errors of immunity: 2022 update on the classification from the International Union of Immunological Societies Expert Committee. J Clin Immunol 2022, 42, 1473–1507. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4. Chan AY, Torgerson TR.. Primary immune regulatory disorders: a growing universe of immune dysregulation. Curr Opin Allergy Clin Immunol 2020, 20, 582–90. doi: 10.1097/ACI.0000000000000689. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. Zhang Q, Frange P, Blanche S, Casanova J-L. Pathogenesis of infections in HIV-infected individuals: insights from primary immunodeficiencies. Curr Opin Immunol 2017, 48, 122–33. doi: 10.1016/j.coi.2017.09.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. Zhang Q, Lenardo MJ, Baltimore D.. 30 years of NF-kappaB: a blossoming of relevance to human pathobiology. Cell 2017, 168, 37–57. doi: 10.1016/j.cell.2016.12.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7. Hayden MS, Ghosh S.. Shared principles in NF-kappaB signaling. Cell 2008, 132, 344–62. doi: 10.1016/j.cell.2008.01.020. [DOI] [PubMed] [Google Scholar]
  • 8. Hayden MS, Ghosh S.. NF-kappaB, the first quarter-century: remarkable progress and outstanding questions. Genes Dev 2012, 26, 203–34. doi: 10.1101/gad.183434.111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Smale ST. Dimer-specific regulatory mechanisms within the NF-kappaB family of transcription factors. Immunol Rev 2012, 246, 193–204. doi: 10.1111/j.1600-065X.2011.01091.x. [DOI] [PubMed] [Google Scholar]
  • 10. Bedsaul JR, Shah N, Hutcherson SM, Pomerantz JL.. Mechanistic impact of oligomer poisoning by dominant-negative CARD11 variants. iScience 2022, 25, 103810. doi: 10.1016/j.isci.2022.103810. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Lork M, Staal J, Beyaert R.. Ubiquitination and phosphorylation of the CARD11-BCL10-MALT1 signalosome in T cells. Cell Immunol 2019, 340, 103877. doi: 10.1016/j.cellimm.2018.11.001. [DOI] [PubMed] [Google Scholar]
  • 12. Gaide O, Martinon F, Micheau O, Bonnet D, Thome M, Tschopp J.. Carma1, a CARD-containing binding partner of Bcl10, induces Bcl10 phosphorylation and NF-kappaB activation. FEBS Lett 2001, 496, 121–7. doi: 10.1016/s0014-5793(01)02414-0. [DOI] [PubMed] [Google Scholar]
  • 13. Yang K, Chi H.. mTOR and metabolic pathways in T cell quiescence and functional activation. Semin Immunol 2012, 24, 421–8. doi: 10.1016/j.smim.2012.12.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Egawa T, Albrecht B, Favier B, Sunshine M-J, Mirchandani K, O'Brien W, et al. Requirement for CARMA1 in antigen receptor-induced NF-kappa B activation and lymphocyte proliferation. Curr Biol 2003, 13, 1252–8. doi: 10.1016/s0960-9822(03)00491-3. [DOI] [PubMed] [Google Scholar]
  • 15. Lu HY, Bauman BM, Arjunaraja S, Dorjbal B, Milner JD, Snow AL, Turvey SE.. The CBM-opathies—a rapidly expanding spectrum of human inborn errors of immunity caused by mutations in the CARD11-BCL10-MALT1 complex. Front Immunol 2018, 9, 2078. doi: 10.3389/fimmu.2018.02078. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. Jattani RP, Tritapoe JM, Pomerantz JL.. Intramolecular interactions and regulation of cofactor binding by the four repressive elements in the caspase recruitment domain-containing protein 11 (CARD11) inhibitory domain. J Biol Chem 2016, 291, 8338–48. doi: 10.1074/jbc.M116.717322. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17. Jattani RP, Tritapoe JM, Pomerantz JL.. Cooperative control of caspase recruitment domain-containing protein 11 (CARD11) signaling by an unusual array of redundant repressive elements. J Biol Chem 2016, 291, 8324–36. doi: 10.1074/jbc.M115.683714. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Meininger I, Krappmann D.. Lymphocyte signaling and activation by the CARMA1-BCL10-MALT1 signalosome. Biol Chem 2016, 397, 1315–33. doi: 10.1515/hsz-2016-0216. [DOI] [PubMed] [Google Scholar]
  • 19. Sommer K, Guo B, Pomerantz JL, Bandaranayake AD, Moreno-García ME, Ovechkina YL, Rawlings DJ.. Phosphorylation of the CARMA1 linker controls NF-kappaB activation. Immunity 2005, 23, 561–74. doi: 10.1016/j.immuni.2005.09.014. [DOI] [PubMed] [Google Scholar]
  • 20. Qiao Q, Yang C, Zheng C, Fontán L, David L, Yu X, et al. Structural architecture of the CARMA1/Bcl10/MALT1 signalosome: nucleation-induced filamentous assembly. Mol Cell 2013, 51, 766–79. doi: 10.1016/j.molcel.2013.08.032. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Che T, You Y, Wang D, Tanner MJ, Dixit VM, Lin X.. MALT1/paracaspase is a signaling component downstream of CARMA1 and mediates T cell receptor-induced NF-kappaB activation. J Biol Chem 2004, 279, 15870–6. doi: 10.1074/jbc.M310599200. [DOI] [PubMed] [Google Scholar]
  • 22. David L, Li Y, Ma J, Garner E, Zhang X, Wu H.. Assembly mechanism of the CARMA1-BCL10-MALT1-TRAF6 signalosome. Proc Natl Acad Sci U S A 2018, 115, 1499–504. doi: 10.1073/pnas.1721967115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Sun L, Deng L, Ea C-K, Xia Z-P, Chen ZJ.. The TRAF6 ubiquitin ligase and TAK1 kinase mediate IKK activation by BCL10 and MALT1 in T lymphocytes. Mol Cell 2004, 14, 289–301. doi: 10.1016/s1097-2765(04)00236-9. [DOI] [PubMed] [Google Scholar]
  • 24. Oeckinghaus A, Wegener E, Welteke V, Ferch U, Arslan SC, Ruland J, et al. Malt1 ubiquitination triggers NF-kappaB signaling upon T-cell activation. EMBO J 2007, 26, 4634–45. doi: 10.1038/sj.emboj.7601897. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. Yang Y, Schmitz R, Mitala J, Whiting A, Xiao W, Ceribelli M, et al. Essential role of the linear ubiquitin chain assembly complex in lymphoma revealed by rare germline polymorphisms. Cancer Discov 2014, 4, 480–93. doi: 10.1158/2159-8290.CD-13-0915. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Dubois SM, Alexia C, Wu Y, Leclair HM, Leveau C, Schol E, et al. A catalytic-independent role for the LUBAC in NF-kappaB activation upon antigen receptor engagement and in lymphoma cells. Blood 2014, 123, 2199–203. doi: 10.1182/blood-2013-05-504019. [DOI] [PubMed] [Google Scholar]
  • 27. Noels H, van Loo G, Hagens S, Broeckx V, Beyaert R, Marynen P, et al. A Novel TRAF6 binding site in MALT1 defines distinct mechanisms of NF-kappaB activation by API2middle dotMALT1 fusions. J Biol Chem 2007, 282, 10180–9. doi: 10.1074/jbc.M611038200. [DOI] [PubMed] [Google Scholar]
  • 28. Sato S, Sanjo H, Tsujimura T, Ninomiya-Tsuji J, Yamamoto M, Kawai T, et al. TAK1 is indispensable for development of T cells and prevention of colitis by the generation of regulatory T cells. Int Immunol 2006, 18, 1405–11. doi: 10.1093/intimm/dxl082. [DOI] [PubMed] [Google Scholar]
  • 29. Schuman J, Chen Y, Podd A, Yu M, Liu H-H, Wen R, et al. A critical role of TAK1 in B-cell receptor-mediated nuclear factor kappaB activation. Blood 2009, 113, 4566–74. doi: 10.1182/blood-2008-08-176057. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. Malynn BA, Ma A.. A20: A multifunctional tool for regulating immunity and preventing disease. Cell Immunol 2019, 340, 103914. doi: 10.1016/j.cellimm.2019.04.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. Wang S, Wen F, Tessneer KL, Gaffney PM.. TALEN-mediated enhancer knockout influences TNFAIP3 gene expression and mimics a molecular phenotype associated with systemic lupus erythematosus. Genes Immun 2016, 17, 165–70. doi: 10.1038/gene.2016.4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Wang S, Wen F, Wiley GB, Kinter MT, Gaffney PM.. An enhancer element harboring variants associated with systemic lupus erythematosus engages the TNFAIP3 promoter to influence A20 expression. PLoS Genet 2013, 9, e1003750. doi: 10.1371/journal.pgen.1003750. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. Adrianto I, Wen F, Templeton A, Wiley G, King JB, Lessard CJ, et al.; BIOLUPUS and GENLES Networks. Association of a functional variant downstream of TNFAIP3 with systemic lupus erythematosus. Nat Genet 2011, 43, 253–8. doi: 10.1038/ng.766. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Catrysse L, Vereecke L, Beyaert R, van Loo G.. A20 in inflammation and autoimmunity. Trends Immunol 2014, 35, 22–31. doi: 10.1016/j.it.2013.10.005. [DOI] [PubMed] [Google Scholar]
  • 35. Boone DL, Turer EE, Lee EG, Ahmad R-C, Wheeler MT, Tsui C, et al. The ubiquitin-modifying enzyme A20 is required for termination of Toll-like receptor responses. Nat Immunol 2004, 5, 1052–60. doi: 10.1038/ni1110. [DOI] [PubMed] [Google Scholar]
  • 36. 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, 694–9. doi: 10.1038/nature02794. [DOI] [PubMed] [Google Scholar]
  • 37. Duwel M, Welteke V, Oeckinghaus A, Baens M, Kloo B, Ferch U, et al. A20 negatively regulates T cell receptor signaling to NF-kappaB by cleaving Malt1 ubiquitin chains. J Immunol 2009, 182, 7718–28. doi: 10.4049/jimmunol.0803313. [DOI] [PubMed] [Google Scholar]
  • 38. Chandrakasan S, Chandra S, Davila Saldana BJ, Torgerson TR, Buchbinder D.. Primary immune regulatory disorders for the pediatric hematologist and oncologist: a case-based review. Pediatr Blood Cancer 2019, 66, e27619. doi: 10.1002/pbc.27619. [DOI] [PubMed] [Google Scholar]
  • 39. Abolhassani H, Hammarstrom L, Cunningham-Rundles C.. Current genetic landscape in common variable immune deficiency. Blood 2020, 135, 656–67. doi: 10.1182/blood.2019000929. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Lougaris V, Patrizi O, Baronio M, Tabellini G, Tampella G, Damiati E, et al. NFKB1 regulates human NK cell maturation and effector functions. Clin Immunol 2017, 175, 99–108. doi: 10.1016/j.clim.2016.11.012. [DOI] [PubMed] [Google Scholar]
  • 41. Mandola AB, Sharfe N, Nagdi Z, Dadi H, Vong L, Merico D, et al. Combined immunodeficiency caused by a novel homozygous NFKB1 mutation. J Allergy Clin Immunol 2021, 147, 727–733.e2. doi: 10.1016/j.jaci.2020.08.040. [DOI] [PubMed] [Google Scholar]
  • 42. Schroder C, Sogkas G, Fliegauf M, Dörk T, Liu D, Hanitsch LG, et al. Late-onset antibody deficiency due to monoallelic alterations in NFKB1. Front Immunol 2019, 10, 2618. doi: 10.3389/fimmu.2019.02618. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43. Tangye SG, Al-Herz W, Bousfiha A, Chatila T, Cunningham-Rundles C, Etzioni A, et al. Human inborn errors of immunity: 2019 update on the classification from the International Union of Immunological Societies Expert Committee. J Clin Immunol 2020, 40, 24–64. doi: 10.1007/s10875-019-00737-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. Bousfiha A, Jeddane L, Picard C, Al-Herz W, Ailal F, Chatila T, et al. Human inborn errors of immunity: 2019 update of the IUIS phenotypical classification. J Clin Immunol 2020, 40, 66–81. doi: 10.1007/s10875-020-00758-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. Cunningham-Rundles C. Common variable immune deficiency: dissection of the variable. Immunol Rev 2019, 287, 145–61. doi: 10.1111/imr.12728. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46. Lorenzini T, Fliegauf M, Klammer N, Frede N, Proietti M, Bulashevska A, et al.; NIHR BioResource. Characterization of the clinical and immunologic phenotype and management of 157 individuals with 56 distinct heterozygous NFKB1 mutations. J Allergy Clin Immunol 2020, 146, 901–11. doi: 10.1016/j.jaci.2019.11.051. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. Li J, Lei WT, Zhang P, Rapaport F, Seeleuthner Y, Lyu B, et al. Biochemically deleterious human NFKB1 variants underlie an autosomal dominant form of common variable immunodeficiency. J Exp Med 2021, 218. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48. Fang R, Wang J, Jiang X-Y, Wang S-H, Cheng H, Zhou Q.. Case report: a novel mutation in NFKB1 associated with Pyoderma Gangrenosum. Front Genet 2021, 12, 673453. doi: 10.3389/fgene.2021.673453. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49. Kaustio M, Haapaniemi E, Göös H, Hautala T, Park G, Syrjänen J, et al. Damaging heterozygous mutations in NFKB1 lead to diverse immunologic phenotypes. J Allergy Clin Immunol 2017, 140, 782–96. doi: 10.1016/j.jaci.2016.10.054. [DOI] [PubMed] [Google Scholar]
  • 50. Klemann C, Camacho-Ordonez N, Yang L, Eskandarian Z, Rojas-Restrepo JL, Frede N, et al. Clinical and immunological phenotype of patients with primary immunodeficiency due to damaging mutations in NFKB2. Front Immunol 2019, 10, 297. doi: 10.3389/fimmu.2019.00297. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51. Chen K, Coonrod EM, Kumánovics A, Franks ZF, Durtschi JD, Margraf RL, et al. Germline mutations in NFKB2 implicate the noncanonical NF-kappaB pathway in the pathogenesis of common variable immunodeficiency. Am J Hum Genet 2013, 93, 812–24. doi: 10.1016/j.ajhg.2013.09.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52. Bogaert DJ, Dullaers M, Lambrecht BN, Vermaelen KY, De Baere E, Haerynck F.. Genes associated with common variable immunodeficiency: one diagnosis to rule them all?. J Med Genet 2016, 53, 575–90. doi: 10.1136/jmedgenet-2015-103690. [DOI] [PubMed] [Google Scholar]
  • 53. Sharfe N, Merico D, Karanxha A, Macdonald C, Dadi H, Ngan B, et al. The effects of RelB deficiency on lymphocyte development and function. J Autoimmun 2015, 65, 90–100. doi: 10.1016/j.jaut.2015.09.001. [DOI] [PubMed] [Google Scholar]
  • 54. Kotlinowski J, Bukowska-Strakova K, Koppolu A, Kosińska J, Pydyn N, Stawinski P, et al. A novel monoallelic nonsense mutation in the NFKB2 gene does not cause a clinical manifestation. Front Genet 2019, 10, 140. doi: 10.3389/fgene.2019.00140. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55. De Leo P, Gazzurelli L, Baronio M, Montin D, Di Cesare S, Giancotta C, et al. NFKB2 regulates human Tfh and Tfr pool formation and germinal center potential. Clin Immunol 2020, 210, 108309. doi: 10.1016/j.clim.2019.108309. [DOI] [PubMed] [Google Scholar]
  • 56. Lee CE, Fulcher DA, Whittle B, Chand R, Fewings N, Field M, et al. Autosomal-dominant B-cell deficiency with alopecia due to a mutation in NFKB2 that results in nonprocessable p100. Blood 2014, 124, 2964–72. doi: 10.1182/blood-2014-06-578542. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Wirasinha RC, Davies AR, Srivastava M, Sheridan JM, Sng XY, Delmonte OM, et al. Nfkb2 variants reveal a p100-degradation threshold that defines autoimmune susceptibility. J Exp Med 2021, 218. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58. Willmann KL, Klaver S, Doğu F, Santos-Valente E, Garncarz W, Bilic I, et al. Biallelic loss-of-function mutation in NIK causes a primary immunodeficiency with multifaceted aberrant lymphoid immunity. Nat Commun 2014, 5, 5360. doi: 10.1038/ncomms6360. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59. Boisson B, Puel A, Picard C, Casanova J-L.. Human IkappaBalpha gain of function: a severe and syndromic immunodeficiency. J Clin Immunol 2017, 37, 397–412. doi: 10.1007/s10875-017-0400-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60. Courtois G, Smahi A, Reichenbach J, Döffinger R, Cancrini C, Bonnet M, et al. A hypermorphic IkappaBalpha mutation is associated with autosomal dominant anhidrotic ectodermal dysplasia and T cell immunodeficiency. J Clin Invest 2003, 112, 1108–15. doi: 10.1172/JCI18714. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61. Uzel G. The range of defects associated with nuclear factor kappaB essential modulator. Curr Opin Allergy Clin Immunol 2005, 5, 513–8. doi: 10.1097/01.all.0000191241.66373.74. [DOI] [PubMed] [Google Scholar]
  • 62. Fusco F, Pescatore A, Conte MI, Mirabelli P, Paciolla M, Esposito E, et al. EDA-ID and IP, two faces of the same coin: how the same IKBKG/NEMO mutation affecting the NF-kappaB pathway can cause immunodeficiency and/or inflammation. Int Rev Immunol 2015, 34, 445–59. doi: 10.3109/08830185.2015.1055331. [DOI] [PubMed] [Google Scholar]
  • 63. Zilberman-Rudenko J, Shawver LM, Wessel AW, Luo Y, Pelletier M, Tsai WL, et al. Recruitment of A20 by the C-terminal domain of NEMO suppresses NF-kappaB activation and autoinflammatory disease. Proc Natl Acad Sci U S A 2016, 113, 1612–7. doi: 10.1073/pnas.1518163113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64. Zonana J, Elder ME, Schneider LC, Orlow SJ, Moss C, Golabi M, et al. A novel X-linked disorder of immune deficiency and hypohidrotic ectodermal dysplasia is allelic to incontinentia pigmenti and due to mutations in IKK-gamma (NEMO). Am J Hum Genet 2000, 67, 1555–62. doi: 10.1086/316914. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65. Lee Y, Wessel AW, Xu J, Reinke JG, Lee E, Kim SM, et al. Genetically programmed alternative splicing of NEMO mediates an autoinflammatory disease phenotype. J Clin Invest 2022, 132. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66. Mousallem T, Yang J, Urban TJ, Wang H, Adeli M, Parrott RE, et al. A nonsense mutation in IKBKB causes combined immunodeficiency. Blood 2014, 124, 2046–50. doi: 10.1182/blood-2014-04-571265. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67. Cuvelier GDE, Rubin TS, Junker A, Sinha R, Rosenberg AM, Wall DA, Schroeder ML.. Clinical presentation, immunologic features, and hematopoietic stem cell transplant outcomes for IKBKB immune deficiency. Clin Immunol 2019, 205, 138–47. doi: 10.1016/j.clim.2018.10.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68. Qin T, Jia Y, Liu Y, Dai R, Zhou L, Okada S, et al. A novel homozygous mutation destabilizes IKKbeta and leads to human combined immunodeficiency. Front Immunol 2020, 11, 517544. doi: 10.3389/fimmu.2020.517544. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69. Pannicke U, Baumann B, Fuchs S, Henneke P, Rensing-Ehl A, Rizzi M, et al. Deficiency of innate and acquired immunity caused by an IKBKB mutation. N Engl J Med 2013, 369, 2504–14. doi: 10.1056/NEJMoa1309199. [DOI] [PubMed] [Google Scholar]
  • 70. Fuchs S, Rensing-Ehl A, Pannicke U, Lorenz MR, Fisch P, Jeelall Y, et al. Omenn syndrome associated with a functional reversion due to a somatic second-site mutation in CARD11 deficiency. Blood 2015, 126, 1658–69. doi: 10.1182/blood-2015-03-631374. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71. Greil J, Rausch T, Giese T, Bandapalli OR, Daniel V, Bekeredjian-Ding I, et al. Whole-exome sequencing links caspase recruitment domain 11 (CARD11) inactivation to severe combined immunodeficiency. J Allergy Clin Immunol 2013, 131, 1376–83.e3. doi: 10.1016/j.jaci.2013.02.012. [DOI] [PubMed] [Google Scholar]
  • 72. Lu HY, Sharma M, Sharma AA, Lacson A, Szpurko A, Luider J, et al. Mechanistic understanding of the combined immunodeficiency in complete human CARD11 deficiency. J Allergy Clin Immunol 2021, 148, 1559–1574.e13. doi: 10.1016/j.jaci.2021.04.006. [DOI] [PubMed] [Google Scholar]
  • 73. Stepensky P, Keller B, Buchta M, Kienzler A-K, Elpeleg O, Somech R, et al. Deficiency of caspase recruitment domain family, member 11 (CARD11), causes profound combined immunodeficiency in human subjects. J Allergy Clin Immunol 2013, 131, 477–85.e1. doi: 10.1016/j.jaci.2012.11.050. [DOI] [PubMed] [Google Scholar]
  • 74. Dadi H, Jones TA, Merico D, Sharfe N, Ovadia A, Schejter Y, et al. Combined immunodeficiency and atopy caused by a dominant negative mutation in caspase activation and recruitment domain family member 11 (CARD11). J Allergy Clin Immunol 2018, 141, 1818–1830.e2. doi: 10.1016/j.jaci.2017.06.047. [DOI] [PubMed] [Google Scholar]
  • 75. Dorjbal B, Stinson JR, Ma CA, Weinreich MA, Miraghazadeh B, Hartberger JM, et al. Hypomorphic caspase activation and recruitment domain 11 (CARD11) mutations associated with diverse immunologic phenotypes with or without atopic disease. J Allergy Clin Immunol 2019, 143, 1482–95. doi: 10.1016/j.jaci.2018.08.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76. Ma CA, Stinson JR, Zhang Y, Abbott JK, Weinreich MA, Hauk PJ, et al. Germline hypomorphic CARD11 mutations in severe atopic disease. Nat Genet 2017, 49, 1192–201. doi: 10.1038/ng.3898. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77. Jun JE, Wilson LE, Vinuesa CG, Lesage S, Blery M, Miosge LA, et al. Identifying the MAGUK protein Carma-1 as a central regulator of humoral immune responses and atopy by genome-wide mouse mutagenesis. Immunity 2003, 18, 751–62. doi: 10.1016/s1074-7613(03)00141-9. [DOI] [PubMed] [Google Scholar]
  • 78. Hutcherson SM, Bedsaul JR, Pomerantz JL.. Pathway-specific defects in T, B, and NK cells and age-dependent development of high IgE in mice heterozygous for a CADINS-associated dominant negative CARD11 allele. J Immunol 2021, 207, 1150–64. doi: 10.4049/jimmunol.2001233. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79. Altin JA, Tian L, Liston A, Bertram EM, Goodnow CC, Cook MC.. Decreased T-cell receptor signaling through CARD11 differentially compromises forkhead box protein 3-positive regulatory versus T(H)2 effector cells to cause allergy. J Allergy Clin Immunol 2011, 127, 1277–85.e5. doi: 10.1016/j.jaci.2010.12.1081. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80. Cardinez C, Miraghazadeh B, Tanita K, da Silva E, Hoshino A, Okada S, et al. Gain-of-function IKBKB mutation causes human combined immune deficiency. J Exp Med 2018, 215, 2715–24. doi: 10.1084/jem.20180639. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81. Snow AL, Xiao W, Stinson JR, Lu W, Chaigne-Delalande B, Zheng L, et al. Congenital B cell lymphocytosis explained by novel germline CARD11 mutations. J Exp Med 2012, 209, 2247–61. doi: 10.1084/jem.20120831. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82. Shields AM, Bauman BM, Hargreaves CE, Pollard AJ, Snow AL, Patel SY.. A novel, heterozygous three base-pair deletion in CARD11 results in B cell expansion with NF-kappaB and T cell anergy disease. J Clin Immunol 2020, 40, 406–11. doi: 10.1007/s10875-019-00729-x. [DOI] [PubMed] [Google Scholar]
  • 83. Urdinez L, Erra L, Palma AM, Mercogliano MF, Fernandez JB, Prieto E, et al. Expanding spectrum, intrafamilial diversity, and therapeutic challenges from 15 patients with heterozygous CARD11-associated diseases: A single center experience. Front Immunol 2022, 13, 1020927. doi: 10.3389/fimmu.2022.1020927. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84. Zhao P, Meng Q, Huang Y, Zhang L, Luo S, Zhang X, et al. Identification and characterization of a germline mutation in CARD11 from a Chinese case of B cell expansion with NF-kappaB and T cell anergy. Front Immunol 2021, 12, 676386. doi: 10.3389/fimmu.2021.676386. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85. Jeelall YS, Wang JQ, Law H-D, Domaschenz H, Fung HKH, Kallies A, et al. Human lymphoma mutations reveal CARD11 as the switch between self-antigen-induced B cell death or proliferation and autoantibody production. J Exp Med 2012, 209, 1907–17. doi: 10.1084/jem.20112744. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86. Desjardins M, Arjunaraja S, Stinson JR, Dorjbal B, Sundaresan J, Niemela J, et al. A unique heterozygous CARD11 mutation combines pathogenic features of both gain- and loss-of-function patients in a four-generation family. Front Immunol 2018, 9, 2944. doi: 10.3389/fimmu.2018.02944. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87. Gupta M, Aluri J, Desai M, Lokeshwar M, Taur P, Lenardo M, et al. Clinical, immunological, and molecular findings in four cases of B cell expansion with NF-kappaB and T cell anergy disease for the first time from India. Front Immunol 2018, 9, 1049. doi: 10.3389/fimmu.2018.01049. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88. Stinson JR, Dorjbal B, McDaniel DP, David L, Wu H, Snow AL.. Gain-of-function mutations in CARD11 promote enhanced aggregation and idiosyncratic signalosome assembly. Cell Immunol 2020, 353, 104129. doi: 10.1016/j.cellimm.2020.104129. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89. Wei Z, Zhang Y, Chen J, Hu Y, Jia P, Wang X, et al. Pathogenic CARD11 mutations affect B cell development and differentiation through a noncanonical pathway. Sci Immunol 2019, 4. [DOI] [PubMed] [Google Scholar]
  • 90. Arjunaraja S, Nosé BD, Sukumar G, Lott NM, Dalgard CL, Snow AL.. Intrinsic plasma cell differentiation defects in B cell expansion with NF-kappaB and T cell anergy patient B cells. Front Immunol 2017, 8, 913. doi: 10.3389/fimmu.2017.00913. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91. Lenz G, Davis RE, Ngo VN, Lam L, George TC, Wright GW, et al. Oncogenic CARD11 mutations in human diffuse large B cell lymphoma. Science 2008, 319, 1676–9. doi: 10.1126/science.1153629. [DOI] [PubMed] [Google Scholar]
  • 92. Compagno M, Lim WK, Grunn A, Nandula SV, Brahmachary M, Shen Q, et al. Mutations of multiple genes cause deregulation of NF-kappaB in diffuse large B-cell lymphoma. Nature 2009, 459, 717–21. doi: 10.1038/nature07968. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93. Kato M, Sanada M, Kato I, Sato Y, Takita J, Takeuchi K, et al. Frequent inactivation of A20 in B-cell lymphomas. Nature 2009, 459, 712–6. doi: 10.1038/nature07969. [DOI] [PubMed] [Google Scholar]
  • 94. Bosanac I, Wertz IE, Pan B, Yu C, Kusam S, Lam C, et al. Ubiquitin binding to A20 ZnF4 is required for modulation of NF-kappaB signaling. Mol Cell 2010, 40, 548–57. doi: 10.1016/j.molcel.2010.10.009. [DOI] [PubMed] [Google Scholar]
  • 95. Tokunaga F, Nishimasu H, Ishitani R, Goto E, Noguchi T, Mio K, et al. Specific recognition of linear polyubiquitin by A20 zinc finger 7 is involved in NF-kappaB regulation. EMBO J 2012, 31, 3856–70. doi: 10.1038/emboj.2012.241. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96. Verhelst K, Carpentier I, Kreike M, Meloni L, Verstrepen L, Kensche T, et al. A20 inhibits LUBAC-mediated NF-kappaB activation by binding linear polyubiquitin chains via its zinc finger 7. EMBO J 2012, 31, 3845–55. doi: 10.1038/emboj.2012.240. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97. 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, 67–73. doi: 10.1038/ng.3459. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98. Franco-Jarava C, Wang H, Martin-Nalda A, Alvarez de la Sierra Daniel, García-Prat M, Bodet D, et al. TNFAIP3 haploinsufficiency is the cause of autoinflammatory manifestations in a patient with a deletion of 13Mb on chromosome 6. Clin Immunol 2018, 191, 44–51. doi: 10.1016/j.clim.2018.03.009. [DOI] [PubMed] [Google Scholar]
  • 99. Berteau F, Rouviere B, Delluc A, Nau A, Le Berre R, Sarrabay G, et al. Autosomic dominant familial Behcet disease and haploinsufficiency A20: a review of the literature. Autoimmun Rev 2018, 17, 809–15. doi: 10.1016/j.autrev.2018.02.012. [DOI] [PubMed] [Google Scholar]
  • 100. 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, 728–35. doi: 10.1136/annrheumdis-2017-212403. [DOI] [PubMed] [Google Scholar]
  • 101. Hu J, Yi S, Wang C, Zhang Y, Tang J, Huang X, et al. A20 inhibits intraocular inflammation in mice by regulating the function of CD4+T cells and RPE cells. Front Immunol 2020, 11, 603939. doi: 10.3389/fimmu.2020.603939. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102. Nocturne G, Boudaoud S, Miceli-Richard C, Viengchareun S, Lazure T, Nititham J, et al. Germline and somatic genetic variations of TNFAIP3 in lymphoma complicating primary Sjogren’s syndrome. Blood 2013, 122, 4068–76. doi: 10.1182/blood-2013-05-503383. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103. Johnsen SJ, Gudlaugsson E, Skaland I, Janssen EAM, Jonsson MV, Helgeland L, et al. Low protein A20 in minor salivary glands is associated with lymphoma in primary Sjogren’s syndrome. Scand J Immunol 2016, 83, 181–7. doi: 10.1111/sji.12405. [DOI] [PubMed] [Google Scholar]
  • 104. Chu Y, Vahl JC, Kumar D, Heger K, Bertossi A, Wójtowicz E, et al. B cells lacking the tumor suppressor TNFAIP3/A20 display impaired differentiation and hyperactivation and cause inflammation and autoimmunity in aged mice. Blood 2011, 117, 2227–36. doi: 10.1182/blood-2010-09-306019. [DOI] [PubMed] [Google Scholar]
  • 105. Hovelmeyer N, Reissig S, Xuan NT, Adams-Quack P, Lukas D, Nikolaev A, et al. A20 deficiency in B cells enhances B-cell proliferation and results in the development of autoantibodies. Eur J Immunol 2011, 41, 595–601. doi: 10.1002/eji.201041313. [DOI] [PubMed] [Google Scholar]
  • 106. Tavares RM, Turer EE, Liu CL, Advincula R, Scapini P, Rhee L, et al. The ubiquitin modifying enzyme A20 restricts B cell survival and prevents autoimmunity. Immunity 2010, 33, 181–91. doi: 10.1016/j.immuni.2010.07.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107. Giordano M, Roncagalli R, Bourdely P, Chasson L, Buferne M, Yamasaki S, et al. The tumor necrosis factor alpha-induced protein 3 (TNFAIP3, A20) imposes a brake on antitumor activity of CD8 T cells. Proc Natl Acad Sci U S A 2014, 111, 11115–20. doi: 10.1073/pnas.1406259111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108. Verdeil G, Schmitt-Verhulst AM.. Unleashing antitumor T-cell activation without ensuing autoimmunity: the case for A20-deletion in adoptive CD8(+) T-cell therapy. Oncoimmunology 2014, 3, e958951e958951. doi: 10.4161/21624011.2014.958951. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109. Just S, Nishanth G, Buchbinder JH, Wang X, Naumann M, Lavrik I, Schlüter D.. A20 Curtails primary but augments secondary CD8(+) T cell responses in intracellular bacterial infection. Sci Rep 2016, 6, 39796. doi: 10.1038/srep39796. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110. Matsuzawa Y, Oshima S, Takahara M, Maeyashiki C, Nemoto Y, Kobayashi M, et al. TNFAIP3 promotes survival of CD4 T cells by restricting MTOR and promoting autophagy. Autophagy 2015, 11, 1052–62. doi: 10.1080/15548627.2015.1055439. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111. Drennan MB, Govindarajan S, Verheugen E, Coquet JM, Staal J, McGuire C, et al. NKT sublineage specification and survival requires the ubiquitin-modifying enzyme TNFAIP3/A20. J Exp Med 2016, 213, 1973–81. doi: 10.1084/jem.20151065. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112. Fischer JC, Otten V, Kober M, Drees C, Rosenbaum M, Schmickl M, et al. A20 restrains thymic regulatory T cell development. J Immunol 2017, 199, 2356–65. doi: 10.4049/jimmunol.1602102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113. Coornaert B, Baens M, Heyninck K, Bekaert T, Haegman M, Staal J, et al. T cell antigen receptor stimulation induces MALT1 paracaspase-mediated cleavage of the NF-kappaB inhibitor A20. Nat Immunol 2008, 9, 263–71. doi: 10.1038/ni1561. [DOI] [PubMed] [Google Scholar]
  • 114. De A, Dainichi T, Rathinam CV, Ghosh S.. The deubiquitinase activity of A20 is dispensable for NF-kappaB signaling. EMBO Rep 2014, 15, 775–83. doi: 10.15252/embr.201338305. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115. Yin H, Karayel O, Chao Y-Y, Seeholzer T, Hamp I, Plettenburg O, et al. A20 and ABIN-1 cooperate in balancing CBM complex-triggered NF-kappaB signaling in activated T cells. Cell Mol Life Sci 2022, 79, 112. doi: 10.1007/s00018-022-04154-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116. Lu HY, Sharma M, Biggs CM, Huang Y-H, Shopsowitz KE, Frosk P, et al. The importance of functional validation after next-generation sequencing: evaluation of a novel CARD11 variant. Pediatr Allergy Immunol 2018, 29, 663–8. doi: 10.1111/pai.12930. [DOI] [PubMed] [Google Scholar]
  • 117. Martin K, Junker U, Tritto E, Sutter E, Rubic-Schneider T, Morgan H, et al. Pharmacological inhibition of MALT1 protease leads to a progressive IPEX-like pathology. Front Immunol 2020, 11, 745. doi: 10.3389/fimmu.2020.00745. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Data Availability Statement

Not applicable—no new data were created or analyzed.


Articles from Clinical and Experimental Immunology are provided here courtesy of British Society for Immunology

RESOURCES