Entry - *300248 - INHIBITOR OF NUCLEAR FACTOR KAPPA-B KINASE, REGULATORY SUBUNIT GAMMA; IKBKG - OMIM
* 300248

INHIBITOR OF NUCLEAR FACTOR KAPPA-B KINASE, REGULATORY SUBUNIT GAMMA; IKBKG


Alternative titles; symbols

INHIBITOR OF KAPPA LIGHT POLYPEPTIDE GENE ENHANCER IN B CELLS, KINASE OF, GAMMA
NF-KAPPA-B ESSENTIAL MODULATOR; NEMO
IKK-GAMMA
FIP3


HGNC Approved Gene Symbol: IKBKG

Cytogenetic location: Xq28     Genomic coordinates (GRCh38): X:154,541,238-154,565,046 (from NCBI)


Gene-Phenotype Relationships
Location Phenotype Phenotype
MIM number
Inheritance Phenotype
mapping key
Xq28 Autoinflammatory disease, systemic, X-linked 301081 XL 3
Ectodermal dysplasia and immunodeficiency 1 300291 XLR 3
Immunodeficiency 33 300636 XLR 3
Incontinentia pigmenti 308300 XLD 3

TEXT

Description

The IKBKG gene, also known as NEMO, encodes the regulatory gamma subunit of the IKB kinase (IKK) complex. Along with the other 2 subunits, IKBKA (CHUK; 600664) and IKBKB (603258), this IKK complex leads to the proteolytic degradation of the NFKB (see, e.g., 164011) inhibitor NFKBIA (164008), thus enabling NFKB to translocate to the nucleus and activate the transcription of cytokine-associated genes (summary by Heller et al., 2020).

IKBKG, or NEMO, is the founding member of an evolutionarily conserved family of NEMO-like kinases that function in numerous cell signaling pathways. NEMO-like kinases specifically phosphorylate serine or threonine residues that are followed by a proline residue (Chiu et al., 2011).


Cloning and Expression

Yamaoka et al. (1998) characterized a mutant cell line, 5R, originally isolated as a cellular flat variant of Rat-1 fibroblasts transformed by the Tax protein of human T-cell leukemia virus type 1 (HTLV-1). The 5R cell line was unresponsive to all tested NF-kappa-B (NFKB; see 164011)-activating stimuli. Using a genetic complementation approach, Yamaoka et al. (1998) cloned a component of the I-kappa-B kinase complex that they termed NEMO for 'NF-kappa-B essential modulator' from 5R cells. The 2.8-kb NEMO cDNA encodes a 412-amino acid protein that is acidic, rich in glutamic acid and glutamine residues (each 13%), and contains a leucine zipper motif at amino acids 315-342. Yamaoka et al. (1998) determined that the defective phenotype of 5R cells resulted from the absence of the NEMO protein. NEMO also complemented the 1.3E2 mutant cell line, in which NFKB is not activated in response to a large set of stimuli.

By using a monoclonal antibody against IKK-alpha, Rothwarf et al. (1998) purified the IKK complex to homogeneity from human cell lines. They determined that the IKK complex is composed of IKK-alpha, IKK-beta, and 2 other polypeptides of 50 and 52 kD which are differentially processed forms of a third subunit, IKK-gamma. Rothwarf et al. (1998) cloned IKK-gamma and identified it as the human homolog of mouse NEMO. The 419-amino acid IKK-gamma protein is composed of several potential coiled-coil motifs and a leucine zipper motif.

Using a yeast 2-hybrid assay to search for proteins that could interact with adenovirus protein E3-14.7K, known to prevent TNF-alpha (TNFA; 191160)-induced cytolysis, Li et al. (1999) cloned IKK-gamma, which they called FIP3 (14.7K-interacting protein). FIP3 contains leucine zippers and a zinc finger domain.

Using Western blot analysis and transfection studies in embryonic kidney cells, Puel et al. (2006) demonstrated that the IKBKG gene encodes a predominant 48-kD protein and an N-terminally truncated protein of 45 kD produced in smaller amounts and translated from methionine-38.


Gene Function

Yamaoka et al. (1998) found that NEMO interacted with IKK2 (IKK-beta; 603258), but not with IKK1 (IKK-alpha; 600664).

Rothwarf et al. (1998) determined that IKK-gamma forms dimers and trimers and interacts preferentially with IKK-beta. IKK-gamma was required for activation of the IKK complex. An IKK-gamma carboxy-terminal truncation mutant that binds IKK-beta blocked the activation of IKK and NF-kappa-B.

Li et al. (1999) determined that FIP3 inhibits both basal and induced transcriptional activity of NFKB and causes a late-appearing apoptosis with unique morphologic manifestations. E3-14.7K partially reversed apoptotic death induced by FIP3. FIP3 bound RIP (603453) and NIK (604655), which had been described as essential components of TNF-alpha-induced NFKB activation. FIP3 inhibited activation of NFKB induced by TNF-alpha, TNFR1 (191190), RIP, NIK, and IKK-beta, as well as basal levels of endogenous NFKB in 293 cells. FIP3 appeared both to activate a cell death pathway and to inhibit an NFKB-dependent survival mechanism.

Although in vitro studies had suggested that NEMO associates preferentially with IKK2 and not IKK1, Li et al. (1999) found that IKK1 immunoprecipitated with NEMO, suggesting that in vivo, IKK1 associates with NEMO through a component other than IKK2 in the IKK complex.

May et al. (2000) determined that an N-terminal alpha-helical region of NEMO associates with a region of IKKA and IKKB that they termed the NBD for 'NEMO-binding domain.' The NBD is a 6-amino acid C-terminal segment within the region denoted alpha-2 of IKKA and IKKB. A cell-permeable wildtype NBD peptide, but not a mutant, blocked the association of NEMO with the IKK complex and inhibited cytokine-induced NFKB activation and the expression of an NFKB-dependent gene, E-selectin (SELE; 131210), in endothelial cells in vitro. Wildtype NBD peptide also ameliorated acute inflammation, namely ear edema and peritonitis, in 2 experimental mouse models as effectively as dexamethasone. May et al. (2000) proposed that an NBD-type drug targeting the interaction of NEMO and the IKK complex might control inflammation yet maintain basal NFKB activity, thus avoiding toxic side effects.

Brummelkamp et al. (2003) designed a collection of RNA interference vectors to suppress 50 human deubiquitinating enzymes and used these vectors to identify deubiquitinating enzymes in cancer-relevant pathways. They demonstrated that inhibition of CYLD (605018) enhances activation of the transcription factor NF-kappa-B (164011). They showed that CYLD binds to the NEMO component of the IKK complex, and appears to regulate its activity through deubiquitination of TRAF2 (601895), as TRAF2 ubiquitination can be modulated by CYLD. Inhibition of CYLD increased resistance to apoptosis, suggesting a mechanism through which loss of CYLD contributes to oncogenesis. Brummelkamp et al. (2003) further demonstrated that this effect can be relieved by aspirin derivatives that inhibit NF-kappa-B activity.

Kovalenko et al. (2003) showed that CYLD interacts with NEMO and TRAF2. CYLD has a deubiquitinating activity that is directed toward the non-lys48-linked polyubiquitin chains and negatively modulates TRAF-mediated activation of IKK, strengthening the notion that ubiquitination is involved in IKK activation by TRAFs and suggesting that CYLD functions in this process.

Wu et al. (2006) demonstrated that NEMO associates with activated ATM (607585) after the induction of DNA double-strand breaks. ATM phosphorylates serine-85 of NEMO to promote its ubiquitin-dependent nuclear export. ATM is also exported in a NEMO-dependent manner to the cytoplasm, where it associates with and causes the activation of IKK in a manner dependent on another IKK regulator, a protein rich in glutamate, leucine, lysine, and serine (ELKS; 607127). Thus, Wu et al. (2006) concluded that regulated nuclear shuttling of NEMO links 2 signaling kinases, ATM and IKK, to activate NF-kappa-B by genotoxic signals.

Nenci et al. (2007) demonstrated that the transcription factor NFKB, a master regulator of proinflammatory responses, functions in gut epithelial cells to control epithelial integrity and the interaction between the mucosal immune system and gut microflora. Intestinal epithelial-specific inhibition of NFKB through conditional ablation of NEMO or both IKK1 and IKK2, IKK subunits essential for NFKB activation, spontaneously caused severe chronic intestinal inflammation in mice. NFKB deficiency led to apoptosis of colonic epithelial cells, impaired expression of antimicrobial peptides, and translocation of bacteria into the mucosa. Concurrently, this epithelial defect triggered a chronic inflammatory response in the colon, initially dominated by innate immune cells but later also involving T lymphocytes. Deficiency of the gene encoding the adaptor protein MyD88 (602170) prevented the development of intestinal inflammation, demonstrating that Toll-like receptor activation by intestinal bacteria is essential for disease pathogenesis in this mouse model. Furthermore, NEMO deficiency sensitized epithelial cells to TNF-induced apoptosis, whereas TNF receptor-1 (191190) inactivation inhibited intestinal inflammation, demonstrating that TNFR1 signaling is crucial for disease induction. Nenci et al. (2007) concluded that a primary NFKB signaling defect in intestinal epithelial cells disrupts immune homeostasis in the gastrointestinal tract, causing an inflammatory bowel disease-like phenotype. Their results further identified NFKB signaling in the gut epithelium as a critical regulator of epithelial integrity and intestinal immune homeostasis and have important implications for understanding the mechanisms controlling the pathogenesis of human inflammatory bowel disease.

Cytokine signaling is thought to require assembly of multicomponent signaling complexes at cytoplasmic segments of membrane-embedded receptors, in which receptor-proximal protein kinases are activated. Matsuzawa et al. (2008) reported that, upon ligation, CD40 (109535) formed a complex containing adaptor molecules TRAF2 and TRAF3 (601896), ubiquitin-conjugating enzyme UBC13 (UBE2N; 603679), cellular inhibitor of apoptosis protein-1 (CIAP1, or BIRC2; 601712) and -2 (CIAP2, or BIRC3; 601721), IKK-gamma, and MEKK1 (MAP3K1; 600982). TRAF2, UBC13, and IKK-gamma were required for complex assembly and activation of MEKK1 and MAP kinase cascades. However, the kinases were not activated unless the complex was translocated from the membrane to the cytosol upon CIAP1/CIAP2-induced degradation of TRAF3. Matsuzawa et al. (2008) proposed that this 2-stage signaling mechanism may apply to other innate immune receptors and may account for spatial and temporal separation of MAPK and IKK signaling.

By studying responses to the TLR4 (603030) ligand lipopolysaccharide (LPS) and to the bacterial chemoattractant fMLP in polymorphonuclear neutrophils (PMNs) from 1 patient with IRAK4 deficiency (607676) and 3 patients with NEMO deficiency causing X-linked hyper-IgM immunodeficiency with ectodermal dysplasia (300291), Singh et al. (2009) demonstrated reduced or absent superoxide production after impaired priming and activation of the oligomeric neutrophil NADPH oxidase (NOX; see 300481). The response was particularly weak or absent in IRAK4-deficient PMNs. NEMO-deficient PMNs had a phenotype intermediate between IRAK4-deficient PMNs and normal PMNs. Decreased LPS- and fMLP-induced phosphorylation of p38 (MAPK14; 600289) was observed in both deficiencies. Singh et al. (2009) proposed that decreased activation of NOX may contribute to increased risk of infection in patients with IRAK4 deficiency or NEMO deficiency.

One of the E3 ligases responsible for K63-linked NEMO polyubiquitination is TRAF6 (602355) which participates in several signaling pathways controlling immunity, osteoclastogenesis, skin development, and brain function. Gautheron et al. (2010) determined that a site at the N terminus of NEMO binds the coiled-coil domain of TRAF6 and apparently works in concert with NEMO's ubiquitin-binding domain to provide a dual mode of TRAF6 recognition. The E57K NEMO mutation, found in a mild form of incontinentia pigmenti (IP; 308300), resulted in impaired TRAF6 binding and IL1-beta (147720) signaling. In contrast, activation of NF-kappa-B (see 164011) by TNF-alpha (191160) was not affected. The authors concluded that the NEMO-TRAF6 interaction has physiologic relevance.

Gerlach et al. (2011) identified SHARPIN (611885) as a third component of the linear ubiquitin chain assembly complex (LUBAC), recruited to the CD40 and TNF receptor (see 191190) signaling complexes together with its other constituents, HOIL1 (RBCK1; 610924) and HOIP (RNF31; 612487). Mass spectrometry of TNFA signaling complexes revealed RIP1, also known as RIPK1 (603453), and NEMO to be linearly ubiquitinated. Tokunaga et al. (2011) also identified SHARPIN as a component of LUBAC, and showed that SHARPIN-containing complexes can linearly ubiquitinate NEMO and activate NK-kappa-B (see 164011). Ikeda et al. (2011) reported that SHARPIN functions as a novel component of LUBAC and that the absence of SHARPIN causes dysregulation of NF-kappa-B and apoptotic signaling pathways, explaining the severe phenotypes displayed by cpdm in Sharpin-deficient mice. Upon binding to the LUBAC subunit HOIP, SHARPIN stimulates the formation of linear ubiquitin chains in vitro and in vivo. Coexpression of SHARPIN and HOIP promotes linear ubiquitination of NEMO, an adaptor of the I-kappa-B kinases (IKKs; see 600664) and subsequent activation of NK-kappa-B signaling, whereas SHARPIN deficiency in mice causes an impaired activation of the IKK complex and NF-kappa-B in B cells, macrophages, and mouse embryonic fibroblasts. This effect is further enhanced upon concurrent downregulation of HOIL1L, another HOIP-binding component of LUBAC.

PER1 (602260) is a master regulator of circadian rhythm and functions in the nucleus to repress expression of central circadian clock genes. The periodicity of PER1 abundance, nuclear translocation, and transcriptional repression is regulated by PER1 phosphorylation, ubiquitination, and proteasomal degradation. Using Drosophila cells and various Per mutants, Chiu et al. (2011) found that Per was progressively phosphorylated by Nemo and doubletime (CSNK1E; 600863) during the circadian cycle. This progressive phosphorylation defined the length of the circadian cycle and the timing of proteasome-mediated Per degradation.


Gene Structure

The International Incontinentia Pigmenti Consortium (2000) sequenced the complete NEMO locus. NEMO is a 23-kb gene composed of 10 exons with 3 alternative noncoding first exons, 1a, 1b, and 1c (Jin and Jeang, 1999; Rothwarf et al., 1998; Li et al., 1999). The NEMO gene partially overlaps the G6PD (305900) gene, and it is transcribed in the opposite direction (Jin and Jeang, 1999).

Fusco et al. (2012) stated that the IKBKG gene has 9 coding exons (exons 2 through 10), 4 alternative noncoding first exons (exons 1A through 1D), and 2 promoters. Promoter A drives expression of IKBKG from exons 1D and 1A and is located within intron 2 of the more centromeric G6PD gene, which lies on the opposite strand. Promoter B is a bidirectional housekeeping promoter that drives expression of IKBKG from exons 1B and 1C, as well as G6PD. Fusco et al. (2012) determined that the region containing the G6PD gene and the 5-prime end of the IKBKG gene contains Alu elements.


Mapping

By sequence alignment, Jin and Jeang (1999) mapped the human NEMO gene to chromosome Xq28.

IKBKG Segmental Duplication

Aradhya et al. (2001) identified a truncated copy of NEMO (delta-NEMO), which maps 22 kb distal to NEMO and contains only exons 3 through 10. A sequence of 26 kb 3-prime of the NEMO coding sequence is also present in the same position relative to the delta-NEMO locus, bringing the total length of the duplication to 35.5 kb. The LAGE2 gene (CTAG1B; 300156) is also located within this duplicated region, and a similar but unique LAGE1 gene (CTAG2; 300396) is located just distal to the duplicated loci. Mapping and sequence information indicated that the duplicated regions are in opposite orientation. Analysis of the great apes suggested that the 35.5-kb NEMO/LAGE2 duplication occurred after divergence of the lineage leading to present day humans, chimpanzees, and gorillas, 10 to 15 million years ago. Despite this substantial evolutionary history, only 22 single-nucleotide differences exist between the 2 copies over the entire 35.5 kb, making the duplications more than 99% identical. This high sequence identity and the inverted orientations of the 2 copies, along with duplications of smaller internal sections within each copy, predispose this region to various genomic alterations. Aradhya et al. (2001) detected 4 rearrangements that involved NEMO, delta-NEMO, or LAGE1 and LAGE2. The authors hypothesized that the susceptibility of this complex genomic region to various types of pathogenic and polymorphic rearrangements may underlie the recurrent lethal deletion associated with incontinentia pigmenti (see MOLECULAR GENETICS).

Fusco et al. (2012) stated that the IKBKG gene and the more telomeric CTAG1A gene (300657) were involved in an inverted 35.7-kb segmental duplication on chromosome Xq28 that produced the CTAG1B gene and an IKBKG pseudogene, IKBKGP1, copied from IKBKG exons 3 through 10. Fusco et al. (2012) found that the duplicated regions and the regions between and near them contain a high number of short and long interspersed elements and long terminal repeats.


Molecular Genetics

X-Linked Dominant Incontinentia Pigmenti

The International Incontinentia Pigmenti Consortium (2000) demonstrated that heterozygous mutations in the IKBKG gene cause X-linked dominant incontinentia pigmenti (IP; 308300). The most common mutation in IP is a genomic rearrangement resulting in deletion of part of the NEMO gene (300248.0001). An 870-bp region of identity corresponding to an MER67B repeat exists both in intron 3 and 3-prime to exon 10; recombinations between the regions of identity delete exons 4 though 10 of NEMO. The International Incontinentia Pigmenti Consortium (2000) stated that this rearrangement, which occurs during paternal meiosis, causes 80% of new mutations. It was demonstrated that this mutation results in a lack of NF-kappa-B (see 164011) activation resulting in extreme susceptibility to apoptosis, leading to embryonic death in males and explaining the extremely skewed X inactivation seen in females with IP.

Aradhya et al. (2001) reported 2 families with IP with duplications in a 7-cytosine tract in exon 10 of the IKBKG gene (300248.0008 and 300248.0012). Aradhya et al. (2001) found that the duplication mutations of the IKBKG gene allowed males to survive and affected females to have random or slightly skewed X inactivation, whereas virtually all other mutations eliminate the production of NEMO, causing lethality in males and the typical skewing of X inactivation in females.

The International IP Consortium (2001) investigated 4 male patients with clinical hallmarks of IP. All 4 were found to carry the deletion in the NEMO gene normally associated with male lethality in utero. Survival in 1 patient was explained by a 47,XXY karyotype and skewed X inactivation. Three other patients possessed a normal 46,XY karyotype. It was demonstrated that these patients had both wildtype and deleted copies of the NEMO gene, indicating mosaicism for the common mutation. Therefore, the repeat-mediated rearrangement leading to the common deletion does not require meiotic division. Thus there are 3 mechanisms for survival of males carrying a NEMO mutation: hypomorphic alleles, a 47,XXY karyotype, and somatic mosaicism.

In an examination of families transmitting the recurrent deletion of exons 4 through 10 of the NEMO gene (300248.0001), Aradhya et al. (2001) revealed that the rearrangement occurred in the paternal germline in 70% of cases, indicating that it arises predominantly by intrachromosomal misalignment during meiosis. Expression analysis of human and mouse NEMO/Nemo showed that the gene becomes active early during embryogenesis and is expressed ubiquitously. The authors proposed a model to explain the pathophysiology of IP in terms of disruption of the NF-kappa-B signaling pathway.

Bardaro et al. (2003) presented instances in which the exon 4 to 10 delta-NEMO pseudogene deletion occurred in unaffected parents of 2 females with clinically characteristic IP, confusing the genetic analysis. They described a new PCR-based test that permitted unambiguous molecular diagnosis and proper familial genetic counseling for IP.

Fusco et al. (2004) identified NEMO mutations in 83 of 122 IP patients and measured the effects of NEMO point mutations on NFKB signaling in Nemo-deficient murine pre-B cells. A mutation in the N-terminal domain, required for IKK assembly, reduced but did not abolish NFKB activation following LPS stimulation. Mutations that disrupt the C-terminal domain (required for the recruitment of upstream factors) showed lower or no NFKB activation. There was no phenotype-genotype correlation observed; 64% of patients exhibited an extremely skewed X-inactivation pattern (80:20). Fusco et al. (2004) concluded that IP pathogenesis may depend on a combination of X inactivation and protein domain that recruits upstream factors and activates NFKB.

In a female infant who presented at birth with features of IP, Martinez-Pomar et al. (2005) identified a heterozygous mutation in the IKBKG gene (300248.0017). She also developed transient immunodeficiency that resolved spontaneously in the first years of life. The X-inactivation status of peripheral blood cells from the patient was evaluated at 24, 30, 38, and 48 months of age and was found to have progressed from random at 24 and 30 months to skewed at 38 and 48 months of age, at which point her immunodeficiency had disappeared. Martinez-Pomar et al. (2005) stated that this was the first time that selection against the mutated X chromosome in X-linked disease had been documented in vivo.

Ectodermal Dysplasia and Immunodeficiency 1

In affected males from 4 unrelated families with X-linked recessive hypohidrotic ectodermal dysplasia and immunodeficiency-1 (EDAID1; 300291), Zonana et al. (2000) identified hemizygous mutations in the IKBKG gene (300248.0007-300248.0010). All mutations occurred in exon 10 of the gene, which encodes the C-terminal zinc finger domain, and were predicted to result in a loss of NFKB activation. Since heterozygous mutations in this gene cause IP in females and are usually lethal in males, Zonana et al. (2000) hypothesized that the mutations identified in males with EDAID1 are hypomorphic; functional studies of the variants were not performed. Affected males showed dysgammaglobulinemia and, despite therapy, had significant morbidity and mortality from recurrent infections.

Doffinger et al. (2001) identified 5 novel mutations (see, e.g., 300248.0020) in IKBKG in affected males from 5 kindreds with anhidrotic ectodermal dysplasia and immune deficiency. Doffinger et al. (2001) also showed that the ectodysplasin receptor (EDAR; 604095) triggers NF-kappa-B through the NEMO protein, indicating that anhidrotic ectodermal dysplasia results from impaired NF-kappa-B signaling.

In 2 boys with EDAID1 associated with increased IgM, Jain et al. (2001) identified hemizygous missense mutations in the NEMO gene (C417R, 300248.0009 and D406V, 300248.0011). Both mutations occurred in the putative zinc finger domain, a potentially shared intracellular signaling component for EDAR and CD40L (300386). Although stimulation of patient monocytes with TNF and LPS resulted in responses similar to those of normal controls, stimulation with CD40L failed to induce phosphorylation of I-kappa-B-alpha (IKBA; 164008) and failed to upregulate expression of CD54 (ICAM1; 147840). CD40L stimulation also failed to induce class switching in patient B cells and failed to result in IL12 and TNF production. T-cell function and maturation appeared to be normal in the patients, and they had no history of opportunistic infections suggestive of T-cell dysfunction. Jain et al. (2001) concluded that NEMO has a regulatory function in NFKB activation and B-cell Ig class switching.

Jain et al. (2004) showed that 3 patients with EDAID1 due to missense mutations affecting cys417 of NEMO had markedly diminished levels of serum IgG and IgE. Activation of patient B cells with soluble CD40L and IL4 (147780) induced normal levels of I-epsilon and C-epsilon germline transcripts and expression of AID (AICDA; 605257), suggesting that expression of additional genes, possibly regulated by CD40-mediated REL (164910) activation, are necessary for Ig class switch recombination. Microarray analysis of EDAID1 B cells showed impairments in the expression of RAD50 (604040), LYL1 (151440), LIG4 (601837), and other factors relating to nonhomologous recombination but not previously linked to B-cell differentiation. Jain et al. (2004) proposed that IL4 augments some (e.g., RELA; 164014) but not all NEMO-dependent NFKB signaling.

In 3 unrelated boys with EDAID1, Orange et al. (2002) identified hemizygous mutations in the IKBKG gene. Two mutations occurred in the zinc finger domain (C417R, 300248.0009 and Q403X, 300248.0015), and 1 occurred in the first coiled-coil domain (L153R; 300248.0014). Patient cells showed defective CD40 signaling, as well as impaired natural killer (NK) cell activity, in spite of normal levels of peripheral blood NK numbers. Addition of IL2 (147680) induced NFKB activation and partially reversed the NK activity defect in vitro. Intravenous administration of IL2 to one of the patients, who had the L153R mutation and persistent cytomegalovirus infection, resulted in enhanced NK cell activity. Orange et al. (2002) proposed that NEMO is important for NFKB activation as well as NK cell cytotoxicity, and that IL2 may benefit patients with NEMO mutations.

In a family with ectodermal dysplasia and immunodeficiency previously reported by Lie et al. (1978), Orstavik et al. (2006) identified a splice site mutation in the NEMO gene (300248.0016). The family had 3 stillborn males, 3 affected males who were small for gestational age and died within 8 months, and 1 male who died at age 5 years. The 5-year-old boy had cone-shaped teeth, oligodontia, serious bacterial infections, and inflammatory bowel disease. Isolated subtle tooth anomalies were found in 3 female carriers examined, of whom 2 had random X inactivation and 1 had extreme skewing. Orstavik et al. (2006) stated that this was the first report of random X inactivation in carriers of EDAID.

Takada et al. (2010) analyzed the NEMO gene in a family in which a boy with EDAID1 died at 9 years of age from gastrointestinal bleeding, and his 6-year-old sister and mother were found to have incontinentia pigmenti and entero-Behcet disease (see 109650); all 3 carried the D406V (300248.0011) mutation. Takada et al. (2010) noted that these patients exhibited several unusual features, including the occurrence of incontinentia pigmenti and EDAID1 in a family; the presence of hypopigmented skin lesions in early infancy in the mother and daughter, since they are usually observed in the second decade to adulthood; and a lack of extremely skewed X-chromosome inactivation in the mother and daughter.

In a boy with EDAID1, Roberts et al. (2010) identified a hemizygous 2-bp deletion (c.1182_1183delTT; 300248.0027) in the IKBKG gene, predicted to result in a frameshift and premature termination. His mother, who carried the mutation in heterozygous state, showed signs of IP since childhood. Functional studies of the variant were not performed.

In a male infant with lethal EDAID1, Johnston et al. (2016) identified a hemizygous splice site mutation in the IKBKG gene (300248.0028). Western blot analysis of patient cells showed reduced size of the IKBKG protein, consistent with a frameshift and premature termination.

In a boy (patient 1) with EDAID1, Heller et al. (2020) identified a hemizygous splice site mutation (300248.0031) in the IKBKG gene, resulting in a frameshift and premature termination with loss of the zinc finger domain. Patient cells showed decreased IKBKG levels compared to controls; in vitro studies showed impaired degradation of IKBA and impaired IL6 production upon stimulation with IL1B and TNFA. His mother and sister, who were heterozygous for the mutation, had incontinentia pigmenti.

Immunodeficiency 33

Two unrelated male patients with immunodeficiency-33 (IMD33; 300636) without ectodermal dysplasia were described by Niehues et al. (2004) and Orange et al. (2004). The patient reported by Orange et al. (2004) was found to have a hemizygous splice mutation that resulted in the skipping of exon 9 of the NEMO gene, affecting the LZ domain (300248.0018) but leaving the zinc finger domain intact. In the patient reported by Niehues et al. (2004), Puel et al. (2006) identified a hemizygous 1-bp insertion in exon 2 of the NEMO gene (300248.0019). Puel et al. (2006) showed that a Kozakian methionine codon located immediately downstream from the insertion allowed the reinitiation of translation. The residual production of an NH2-truncated NEMO protein was sufficient for normal fetal development and for the subsequent normal development of skin appendages, but was insufficient for the development of protective immune responses.

In 2 unrelated boys with IMD33, Ku et al. (2005) identified hemizygous mutations in the IKBKG gene: an 18-bp deletion (300248.0025) and a missense mutation (L80P, 300248.0026). The mutations occurred in the coiled-coil domains of the protein. Western blot analysis of patient cells showed the presence of the IKBKG protein, but fibroblasts had poor IL6 production in response to TNFA and IL1B compared to controls. The findings were consistent with a hypomorphic allele. The patients had hypodontia and conical teeth, but no other features of ectodermal dysplasia. One carrier mother also had conical teeth.

In a Belgian boy with IMD33, Ku et al. (2007) identified a hemizygous splice site mutation in the IKBKG gene (300248.0023). Western blot analysis showed decreased levels of the protein compared to controls, consistent with partial IKBKG deficiency. The mother was heterozygous for this mutation.

In affected males from 3 unrelated kindreds with IMD33 manifest as susceptibility to mycobacterial infection, Filipe-Santos et al. (2006) identified 2 hemizygous missense mutations in the NEMO gene (E315A, 300248.0021 and R319Q, 300248.0022), both of which occurred in the leucine zipper (LZ) domain and were predicted to disrupt a salt bridge. Western blot and flow cytometric analyses showed normal expression of the mutant NEMO proteins. Patient mononuclear cells responded normally to most stimuli, but IFNG (147570) and IL12 (see 161561) production in response to PHA mitogen was impaired due to defective CD40 (109535) signaling in monocytes and dendritic cells. Filipe-Santos et al. (2006) concluded that mutations in NEMO that disrupt the leucine zipper domain provide a genetic etiology to X-linked recessive immunodeficiency with a particular susceptibility to mycobacterial disease.

In a 2-year-old boy with IMD33, Frans et al. (2017) identified a hemizygous missense mutation in the IKBKG gene (E57K; 300248.0029). The mutation affected the N-terminal domain of the protein. His mother, who also carried the mutation, had a history of recurrent sinorespiratory infections, but no signs of IP. The variant was present at a low frequency (0.001) in the ExAC database. Patient peripheral blood cells showed mildly decreased IL6 production after stimulation with IL1B compared to controls. However, NEMO expression in patient fibroblasts was normal, and IKBA was degraded normally upon stimulation with IL1B or TNFA, suggesting a specific effect of the mutation. In vitro functional expression studies in IKBKG-null cells transfected with the mutation showed mildly impaired IL6 production after stimulation with TNFA or IL1B compared to controls. Frans et al. (2017) noted that mutations affecting the N terminus of NEMO tend to lead to decreased production of immunoglobulins; the authors postulated a hypomorphic effect of this variant.

In 4 adult males from 3 unrelated families with IMD33 manifest as disseminated mycobacterial infections, Hsu et al. (2018) identified hemizygous splice site mutations in the 5-prime untranslated region of the IKBKG gene. Three patients from 2 families (families A and B) carried a c.1-16G-C transversion (300248.0030) at the last base of the first untranslated exon. The patient from family C carried a hemizygous c.1-16+G-T in the adjacent intron. Analysis of cells from the patient with the c.1-16G-C mutation showed a splicing abnormality, resulting in a 110-bp deletion at the 3-prime end of exon 1. This molecular defect resulted in decreased transcript and protein levels compared to controls (about 30%). Cells derived from the patients in families A and B failed to upregulate cytokines in response to certain TLR agonists, suggesting that this IKBKG mutation is hypomorphic.

In 3 boys from 2 unrelated families of European and Japanese descent with fatal IMD33, Boisson et al. (2019) identified a deep intronic mutation in the IKBKG gene (IVS4+866C-T; 300248.0024) that created a new splice donor site and resulted in a 44-nucleotide pseudoexon that produced a frameshift. The boy in the European family inherited the mutation from his mother, who had mild incontinentia pigmenti. The mutation in the Japanese boy occurred de novo. The variant was not found in the 1000 Genomes Project or gnomAD databases.

In a boy (patient 1) with IMD33, Abbott et al. (2014) identified a hemizygous missense mutation in the first coiled-coil domain of NEMO (D113N; 300248.0032). The patient had previously been reported in detail by Salt et al. (2008); his unaffected mother also carried the mutation.

Heller et al. (2020) identified hemizygosity for the D113N mutation in a boy with IMD33. His mother and grandmother, who were presumably unaffected, were heterozygous for the mutation. However, the proband's 40-year-old male cousin, who did not have recurrent infections and had normal response to polysaccharide antibodies, was hemizygous for D113N. Heller et al. (2020) noted that the allele frequency for this variant is rather high (0.009572), and that some suggest it may be a polymorphism (see Fusco et al., 2004).

X-linked Systemic Autoinflammatory Disease

In 3 unrelated boys (P1-P3) and a girl (P4) with X-linked systemic autoinflammatory disease (SAIDX; 301081), de Jesus et al. (2020) identified de novo hemizygous or heterozygous mutations in the IKBKG, all of which were predicted to disrupt splicing and result in the deletion of exon 5 (300248.0033-300248.0036). Functional studies of the variants were not performed. The patients were ascertained from a cohort of patients with autoinflammatory disease and a moderately elevated type I interferon signature.

In 3 unrelated boys with SAIDX, Lee et al. (2022) identified de novo hemizygous mutations in the IKBKG gene (300248.0033-300248.0035). The mutations, which were confirmed by Sanger sequencing, all led to overexpression of a NEMO protein lacking the domain encoded by exon 5 (NEMOdelEx5). In vitro studies showed that this splice isoform failed to associate with TANK binding kinase-1 (TBK1; 604834). Dermal fibroblasts from affected patients activated NFKB in response to TNF (191160), but showed impaired NFKB activation to TLR3 (603029) or RIGI-like receptor (RLR) stimulation with poly(I:C) positively correlated with levels of the NEMOdelEx5 splice isoform. By contrast, T cells, monocytes, and macrophages that expressed the splice site variant exhibited increased NFKB activation and IFN production. Blood cells from these patients expressed a strong IFN and NFKB transcriptional signature. Immune cells and TNF-stimulated dermal fibroblasts upregulated the inducible IKK protein (IKKi; 605048) that was stabilized by the splice variant, promoting type I IFN induction and antiviral responses. These data showed that IKBKG mutations that lead to alternative splicing of skipping exon 5 cause an autoinflammatory disorder, which the authors termed 'NEMO deleted exon 5 autoinflammatory syndrome (NDAS),' noting that it is distinct from the immunodeficiency syndrome resulting from loss-of-function IKBKG mutations.


Genotype/Phenotype Correlations

In general, males with NKBKG mutations affecting the C-terminal zinc finger domain have a more severe clinical course with immunodeficiency and ectodermal dysplasia, whereas patients with mutations affecting the leucine zipper domain or the more N-terminal coiled-coil domains have a less severe clinical course and do not show features of ectodermal dysplasia, although isolated hypotonia and/or conical teeth may be present (Orange et al., 2004, Heller et al., 2020).


Animal Model

Rudolph et al. (2000) generated NEMO/IKK-gamma-deficient mice by gene targeting. Mutant embryos died at embryonic day 12.5-13 from severe liver damage due to apoptosis. NEMO/IKK-gamma-deficient primary murine embryonic fibroblasts (MEFs) lacked detectable NF-kappa-B DNA-binding activity in response to TNF-alpha, IL1 (see 147760), bacterial lipopolysaccharide, and poly(IC) and did not show stimulus-dependent I-kappa-B kinase activity, which correlated with a lack of phosphorylation and degradation of I-kappa-B-alpha (164008). Consistent with these data, mutant MEFs showed increased sensitivity to TNF-alpha-induced apoptosis. Rudolph et al. (2000) concluded that their data provided in vivo evidence that NEMO is the first essential noncatalytic component of the IKK complex.

Makris et al. (2000) showed that female mice heterozygous for Ikbkg deficiency develop a unique dermatopathy characterized by keratinocyte hyperproliferation, skin inflammation, hyperkeratosis, and increased apoptosis. Although Ikbkg +/- females eventually recovered, IKBKG -/- males died in utero. The symptoms and inheritance pattern were very similar to those of IP. Indeed, biopsies and cells from IP patients exhibited defective IKBKG expression but normal expression of IKK catalytic subunits. The authors proposed that the IKBKG-deficient cells trigger an inflammatory reaction that eventually leads to their death.

Schmidt-Supprian et al. (2000) found that disruption of the mouse Ikbkg gene produces male embryonic lethality, completely blocks NF-kappa-B activation by proinflammatory cytokines, and interferes with the generation and/or persistence of lymphocytes. Heterozygous female mice developed patchy skin lesions with massive granulocyte infiltration and hyperproliferation and increased apoptosis of keratinocytes. Diseased animals presented severe growth retardation and early mortality. Surviving mice recovered almost completely, presumably through clearing the skin of Ikbkg-deficient keratinocytes. The authors stated that male lethality and strikingly similar skin lesions in heterozygous females are hallmarks of the human genetic disorder IP.

Using a chemical mutagenesis screen in mice, Siggs et al. (2010) identified an X-linked recessive phenotype, termed 'pan resistance-2' (panr2), marked by diminished secretion of Tnf and other Nfkb-dependent cytokines by macrophages in response to ligands for Tlr3 (603029), Tlr4, Tlr7 (300365), and Tlr9 (605474), as well as for the TLR heterodimers Tlr1 (601194)/Tlr2 (603028) and Tlr2/Tlr6 (605403). They identified the panr2 mutation as a 473T-C transition in exon 4 of the Ikbkg gene, which results in a leu153-to-pro (L153P) substitution within the first coiled-coil domain of the protein. Panr2 mutant males were viable but azoospermic, and they usually lacked inguinal lymph nodes. Other tissues were normal, although serum lacked all Ig isotypes. Panr2 mutant mice had an impairment of MAPK phosphorylation and Nfkb p65 translocation, but not of Ikba degradation. Siggs et al. (2010) concluded that IKBKG-regulated pathways are of immunologic importance beyond IKBA degradation and may account for other immunodeficiencies.


ALLELIC VARIANTS ( 36 Selected Examples):

.0001 INCONTINENTIA PIGMENTI

IKBKG, EX4-10DEL
   RCV000012200

In male and female fetuses with incontinentia pigmenti (IP; 308300), The International Incontinentia Pigmenti Consortium (2000) found a genomic rearrangement resulting in deletion of part of the NEMO gene. An 870-bp region of identity corresponding to an MER67B repeat exists in the NEMO gene both in intron 3 and 3-prime to exon 10; recombinations between the regions of identity delete exons 4 though 10 of NEMO. The International Incontinentia Pigmenti Consortium (2000) stated that this rearrangement accounts for 80% of new IP mutations. This rearrangement would result in a truncated molecule carrying the 133 N-terminal amino acids of NEMO plus at least 26 novel amino acids. It was predicted that this molecule would contain part of the first coiled-coil domain, and may still interact with IKK2 (603258), but would be unlikely to respond to upstream signals. Failure of I-kappa-B-alpha (164008) degradation and lack of NF-kappa-B (see 164011) activation were demonstrated in IP embryonic fibroblasts.

Aradhya et al. (2001) identified 277 mutations in 357 unrelated IP patients. The recurrent deletion of exons 4 to 10 accounted for 90% of the identified mutations. The remaining mutations were small duplications, substitutions, and deletions.


.0002 ECTODERMAL DYSPLASIA AND IMMUNODEFICIENCY 1, MALE-RESTRICTED

INCONTINENTIA PIGMENTI, INCLUDED
IKBKG, TER420TRP
  
RCV000012203...

In a male infant (patient IP85) with ectodermal dysplasia and immunodeficiency-1 (EDAID1; 300291), The International Incontinentia Pigmenti Consortium (2000) identified a hemizygous c.1259A-G transition in exon 10 of the IKBKG gene, resulting in a stop420-to-trp (X420W) substitution. This change results in the addition of 27 residues to the C terminus of the mature protein. The mutation was also found in heterozygous state in his mother, who had incontinentia pigmenti (IP; 308300). In addition to recurrent infections, the boy also had osteopetrosis and lymphedema. He died at age 2.5 years from a tuberculosis infection.

Doffinger et al. (2001) studied patient IP85 reported by The International Incontinentia Pigmenti Consortium (2000) as well as another boy, of French descent, with the hemizygous X420W mutation and classified both as having anhidrotic ectodermal dysplasia with immunodeficiency, osteopetrosis, and lymphedema (OLEDAID; see 300291), which is within the phenotypic spectrum of EDAID1. Infections in the French boy, who died at the age of 1.5 years, included atypical mycobacteria and pneumococcus; he also had osteopetrosis and lymphedema, indicating that these additional features are associated with this specific mutation. His mother, who carried the mutation in heterozygous state, had mild IP. Detailed in vitro functional expression studies showed that the X420W mutation resulted in a 50 to 60% reduction of NF-kappa-B activation. Cells from both male patients with this mutation showed impaired cellular responses to LPS, IL1B, and TNFA. The findings indicated that the X420W mutation impairs, but does not abolish, NFKB activation, consistent with a hypomorphic allele and postnatal survival of the boys.


.0003 INCONTINENTIA PIGMENTI

IKBKG, 10-BP INS, NT127
  
RCV000012204

In an incontinentia pigmenti (IP; 308300) proband and her affected mother, but not in unaffected sibs, The International Incontinentia Pigmenti Consortium (2000) identified a 10-bp insertion in exon 2 of the IKBKG gene, between nucleotides 127 and 128. The insertion, an exact duplication of the previous 10 nucleotides, shifts the reading frame of the protein to add 8 amino acids after residue 43 and then truncates the protein at the next in-frame stop codon.


.0004 INCONTINENTIA PIGMENTI

IKBKG, 1-BP INS, 1110C
  
RCV000012201...

In a family with incontinentia pigmenti (IP; 308300), The International Incontinentia Pigmenti Consortium (2000) identified a single C insertion between nucleotides 1110 and 1111 that segregated with the disease. This frameshift mutation would putatively append 23 new amino acids onto proline residue 370 in the translated protein.


.0005 INCONTINENTIA PIGMENTI

IKBKG, MET407VAL
  
RCV000012202...

In a female with incontinentia pigmenti (IP; 308300), The International Incontinentia Pigmenti Consortium (2000) identified an A-to-G transition in exon 10 of the IKBKG gene that changed methionine to valine at codon 407. SSCP analysis showed that this relatively conservative change segregated with the disease through a 3-generation pedigree.


.0006 INCONTINENTIA PIGMENTI

IKBKG, PRO62TER
  
RCV000012205...

In a family with incontinentia pigmenti (IP; 308300), The International Incontinentia Pigmenti Consortium (2000) identified a C-to-T transition at nucleotide 184 of the IKBKG gene resulting in a proline-to-stop mutation in exon 2.


.0007 ECTODERMAL DYSPLASIA AND IMMUNODEFICIENCY 1

IKBKG, GLU391TER
  
RCV000012206...

In 3 affected boys from a family (family 2) with hypohidrotic ectodermal dysplasia and immunodeficiency (EDAID1; 300291), Zonana et al. (2000) identified a hemizygous c.1171G-T transversion in exon 10 of the IKBKG gene, resulting in a glu391-to-ter (E391X) substitution within the C-terminal end of the protein. The truncated protein was predicted to lack the putative zinc finger domain. Western blot analysis of patient cells showed that a protein was expressed. Additional functional studies were not performed, but the authors postulated that the mutation preserves some function. Two female carriers in the family had conical teeth, some variable hyperpigmented skin lesions, and increased IgA, but no immune defects.


.0008 ECTODERMAL DYSPLASIA AND IMMUNODEFICIENCY 1

INCONTINENTIA PIGMENTI, INCLUDED
IKBKG, 1-BP DUP, 1167C
  
RCV000012209...

In 2 brothers (family 4) with lethal hypohidrotic ectodermal dysplasia with immune deficiency-1 (EDAID1; 300291), Zonana et al. (2000) identified a hemizygous 1-bp duplication (c.1167_1168dupC) in exon 10 of the IKBKG gene, predicted to result in a frameshift, the addition of novel amino acids at codons 390-393, and premature termination at codon 394. The duplication resulted from an insertion of a cytosine within a wildtype run of 7 cytosines in exon 10. The mutation was predicted to delete the putative zinc finger domain. The patients also had osteopetrosis and died of mycobacterial infection.

Kosaki et al. (2001) described an unusual family in which a girl with this heterozygous mutation had skin abnormalities with streaky hyperpigmented macules, features of ectodermal dysplasia, including decreased sweating, thin hair, and hypodontia, and onset of recurrent infections around 4 years of age. She later developed pulmonary and renovascular hypertension, and died at age 11 years after cardiac catheterization. IgD and IgE were increased. X-inactivation studies of peripheral blood leukocytes showed a random pattern. The mother had hyperpigmented macules in a streaky configuration on the trunk and limbs at several weeks of age, but never developed immunodeficiency; DNA from the mother was not studied. The family had previously been reported by Akiyama et al. (1994) as having 'linear and whorled nevoid hypermelanosis (see 614323). Zonana and Ferguson (2001) noted that the findings of random X inactivation in the girl reported by Kosaki et al. (2001) was unusual, and postulated that other molecular mechanisms may have been at play, including the possibility of skewed X inactivation in different leukocyte subsets. These authors emphasized that this phenotype in females is a very rare occurrence, and that generally, only males are affected.

Aradhya et al. (2001) identified the same frameshift mutation, referred to as a 1-bp duplication in the 7-cytosine tract (1161_1162dupC) of the IKBKG gene, in a male patient (family XL344) with EDAID1. Female members of the family, who were heterozygous for the mutation, had typical signs of incontinentia pigmenti (IP; 308300). The affected male exhibited skin pigmentation, dental problems, and immune dysfunction. He suffered multiple episodes of infection, including meningitis and pneumonia, due to poor lymphocyte function and remarkably low levels of circulating IgG. He also exhibited heat intolerance with hyperthermia, anhidrosis, eczema, and fine sparse hair, which led to a diagnosis of ectodermal dysplasia. At the age of 3 years he was receiving routine supplements of IgG to prevent recurrent infections. He showed hepatosplenomegaly and had contracted Mycobacterium avium intracellulare, an infection common among patients with AIDS. Aradhya et al. (2001) identified a different mutation involving the same C(7) tract in another family; see 300248.0012.


.0009 ECTODERMAL DYSPLASIA AND IMMUNODEFICIENCY 1

IKBKG, CYS417ARG
  
RCV000012207

In 2 brothers (family 1) with hypohidrotic ectodermal dysplasia and immunodeficiency-1 (EDAID1; 300291), Zonana et al. (2000) identified a hemizygous c.1249T-C transition in exon 10 of the IKBKG gene, resulting in a cys417-to-arg (C417R) substitution in the putative zinc finger domain. The mother, who carried the mutation in heterozygous state, was unaffected.

Orange et al. (2002) identified the C417R mutation in a 16-year-old boy (patient 3) with EDAID1 with recurrent sinopulmonary infections and bacteremia. Tetanus vaccination failed to confer detectable serologic or delayed hypersensitivity responses. The patient also had reduced natural killer cell function.

In a male patient with EDAID1, Jain et al. (2001) identified hemizygosity for the C417R mutation, which occurred in the putative zinc finger domain.


.0010 ECTODERMAL DYSPLASIA AND IMMUNODEFICIENCY 1

IKBKG, CYS417PHE
  
RCV000012211...

In a boy (family 3) with hypohidrotic ectodermal dysplasia and immunodeficiency-1 (EDAID1; 300291), Zonana et al. (2000) identified a hemizygous c.1250G-T transversion in exon 10 of the IKBKG gene, resulting in a cys417-to-phe (C417F) substitution in the zinc finger domain. The patient's mother, who carried the mutation, had mild tooth anomalies and a 'large patch of skin with neither hair nor sweating response.'


.0011 ECTODERMAL DYSPLASIA AND IMMUNODEFICIENCY 1, MALE-RESTRICTED

INCONTINENTIA PIGMENTI, INCLUDED
IKBKG, ASP406VAL
  
RCV000024285...

In a male patient with ectodermal dysplasia and immunodeficiency-1 (EDAID1; 300291), Jain et al. (2001) identified a hemizygous c.1217A-T transversion in exon 10 of the IKBKG gene, resulting in an asp406-to-val (D406V) substitution. The mutation occurred in the putative zinc finger domain.

In a family in which a boy with ectodermal dysplasia with immune deficiency-1 died at 9 years of age from gastrointestinal bleeding and his sister and mother were found to have incontinentia pigmenti (IP; 308300) and to fulfill the criteria for entero-Behcet disease (see 109650), Takada et al. (2010) identified the D406V mutation in the NEMO gene. The 6-year-old sister developed oral and perianal ulcers at 5.5 years of age and had hypopigmented lesions without atrophy in the abdominal area and extremities that had been present since early infancy. Endoscopy showed multiple ulcerative lesions in the colon, which had no reactive changes at the margins and showed chronic active inflammation on histology. The authors stated that the patient's findings fulfilled diagnostic criteria for the enteric form of Behcet disease. The 42-year-old mother developed oral and perianal ulcers at 8 years of age and was diagnosed with Behcet disease at age 12. She also had hypopigmented skin lesions in the abdominal area and extremities that had been observed since early infancy. Skewed X-chromosome inactivation was not seen in the peripheral blood mononuclear cells or in oral or intestinal mucosa of these patients.


.0012 INCONTINENTIA PIGMENTI

IKBKG, 13-BP DUP, NT1166
  
RCV000012213

In affected women from a family (family XL320) with incontinentia pigmenti (IP; 308300), Aradhya et al. (2001) identified a heterozygous 13-bp duplication (c.1166_1178dup) at the end of a cytosine tract (a C(7) tract) in exon 10 of the IKBKG gene. This duplication was predicted to cause a frameshift after amino acid P393 and protein truncation after addition of 4 novel amino acids, resulting in absence of the zinc finger domain. The family had previously been studied by Roberts et al. (1998), who reported that an affected male carrying the hemizygous mutation was carried to term, but died from severe hemorrhage 24 hours after birth. In a subsequent pregnancy, the mother experienced severe bleeding during an elective abortion, suggesting that the mutation in this family disrupted hemostasis.


.0013 ECTODERMAL DYSPLASIA AND IMMUNE DEFICIENCY 1

IKBKG, 4.4-KB DUP
   RCV000012214

In a 2-year-old boy with hypohidrotic ectodermal dysplasia and immune deficiency-1 (EDAID1; 300291), Nishikomori et al. (2004) identified a hemizygous 4.4-kb duplication of genomic sequence of a segment extending from intron 3 to exon 6 in the IKBKG gene, resulting in reduced expression of the protein. The patient had atypical features of very few naive T cells and defective mitogen-induced proliferation of peripheral blood mononuclear cells. Specific cell lineages (monocytes and neutrophils) expressed reduced levels of IKBKG, but 2 populations of T, B, and NK cells were detected, 1 with normal and 1 with reduced expression of IKBKG. This was thought most likely to be due to postzygotic reversion. The patient had hypohidrosis, coarse hair, delayed eruption of teeth, and transient lower extremity lymphedema, but did not develop the skin lesions typical of incontinentia pigmenti. At age 3 months, he was admitted to hospital because of interstitial pneumonitis. Thereafter he had frequent bacterial infections and febrile episodes due to skin abscess, pneumonia, pneumococcal sepsis, otitis media, and sepsis. He also suffered from intractable diarrhea that resulted in failure to thrive. One oral live polio vaccination resulted in no detectable anti-poliovirus titers.


.0014 ECTODERMAL DYSPLASIA AND IMMUNODEFICIENCY 1

IKBKG, LEU153ARG
  
RCV000012215

In a patient (patient 1) under 24 months of age who had ectodermal dysplasia with immunodeficiency-1 (EDAID1; 300291), Orange et al. (2002) identified a hemizygous c.458T-G transversion in exon 4 of the IKBKG gene, resulting in a leu153-to-arg (L153R) substitution within the first coiled-coil domain of the protein. In vitro functional expression studies showed impaired CD40-mediated activation of NFKB compared to controls. The patient had bacterial and viral sepsis, as well as Streptococcal bovis meningitis and cytomegalovirus colitis. He was noted to have absent sweat glands on skin biopsy in the 'Methods' section, but additional features of ectodermal dysplasia were not noted. Orange et al. (2004) also reported this patient and noted that he had 'characteristics' of ectodermal dysplasia; his mother carried the mutation and had oligodontia, some alopecia, and skin hyperpigmentation, consistent with mild features of incontinentia pigmenti.


.0015 ECTODERMAL DYSPLASIA AND IMMUNODEFICIENCY 1

IKBKG, GLN403TER
  
RCV000012216

In a 17-year-old patient (patient 2) with hypohidrotic ectodermal dysplasia with immunodeficiency-1 (EDAID1; 300291), Orange et al. (2002) identified a hemizygous c.1207C-T transition in exon 10 of the IKBKG gene, resulting in a gln403-to-ter (Q403X) truncation within the zinc finger domain of the protein. The patient had a history of cutaneous granulomas and the presence of Mycobacterium avium-intracellulare in blood, bone marrow, and skin, as well as deficient specific antibody and lymphoproliferative responses and reduced natural killer cell function.


.0016 ECTODERMAL DYSPLASIA AND IMMUNODEFICIENCY 1

IKBKG, IVS6DS, G-A, +5
  
RCV000012217

In affected members and obligate carriers of a family with ectodermal dysplasia and immunodeficiency-1 (EDAID1; 300291), originally reported by Lie et al. (1978), Orstavik et al. (2006) identified a +5G-A transition in the splice donor site of exon 6 of the IKBKG gene. The mutation was not found in unaffected family members or 140 control chromosomes. RT-PCR analysis of fibroblast RNA from an aborted affected male fetus demonstrated skipping of exons 4, 5, and 6, which resulted in a truncated protein of about 35 kD. IKBA degradation was strongly impaired in the fetal fibroblasts, suggesting impaired NFKB signaling. One healthy carrier female had a completely skewed X-inactivation pattern with the normal X active, whereas the 2 other female carriers had a random X-inactivation pattern.


.0017 INCONTINENTIA PIGMENTI

IKBKG, 1-BP DUP, 1409A
  
RCV000012218

In a female infant with incontinentia pigmenti (IP; 308300) and transient immunodeficiency (see 300291), Martinez-Pomar et al. (2005) identified a 1-bp duplication (c.1049dupA) in exon 7 of the IKBKG gene, within a run of 3 adenosines. The mutation resulted in a frameshift and a premature stop codon at position 284. The X-inactivation status of peripheral blood cells from the patient was evaluated at 24, 30, 38, and 48 months of age and was found to have progressed from random at 24 and 30 months to skewed at 38 and 48 months of age, at which point her immunodeficiency had disappeared. Martinez-Pomar et al. (2005) stated that this was the first time that selection against the mutated X chromosome in X-linked disease had been documented in vivo.


.0018 IMMUNODEFICIENCY 33

IKBKG, IVS8AS, G-A, -1
  
RCV003151721

In a 15-year-old boy with immunodeficiency-33 (IMD33; 300636), Orange et al. (2004) identified a de novo hemizygous G-to-A transition in intron 8 of the IKBKG gene (c.1056-1G-A), resulting in the skipping of exon 9 and the deletion of 19 residues (353_373). Analysis of patient peripheral blood and buccal epithelial cells showed presence of both mutant and wildtype transcripts. Western blot analysis using an antibody against the LZ domain showed barely detectable IKBKG protein levels in patient cells, whereas an antibody against the zinc finger domain showed normal protein levels. Presence of some normal transcripts may explain immunodeficiency without signs of ectodermal dysplasia in this patient. In vitro functional expression studies showed impaired, but not absent, nuclear translocation of NFKB in stimulated patient B cells compared to controls, as well as variable response to TNFA. Orange et al. (2004) postulated that exon 9 may be dispensable for ectodermal development.


.0019 IMMUNODEFICIENCY 33

IKBKG, 1-BP INS, 110C
  
RCV000012220

In a patient with immunodeficiency-33 (IMD33; 300636), originally reported by Niehues et al. (2004), Puel et al. (2006) identified a hemizygous 1-bp insertion in exon 2 of the IKBKG gene (c.110_111insC) that resulted in the most-upstream premature translation termination codon of any NEMO mutation described to that time (Met38fsTer48). An almost canonically Kozakian methionine AUG start codon (Kozak, 2002) is located at nucleotide positions 112-114, corresponding to methionine-38 in the NEMO protein. The likelihood of this AUG codon acting as a translation initiation site was estimated at 0.27 and increased to 0.51 in the sequence context found in the patient studied by Puel et al. (2006), versus 0.63 for the first AUG codon. The reinitiation of translation would result in the synthesis of a protein approximately 45 kD in size. This protein was the only isoform detected in the patient's cells.


.0020 ECTODERMAL DYSPLASIA AND IMMUNODEFICIENCY 1

IKBKG, ALA288GLY
  
RCV000012221

In a 2-year-old boy (patient 10) with anhidrotic ectodermal dysplasia and immunodeficiency-1 (EDAID1; 300291), Doffinger et al. (2001) identified a hemizygous c.863C-G transversion in the IKBKG gene, resulting in an ala288-to-gly (A288G) substitution in the second coiled-coil domain. The mutation was not found in 200 control chromosomes, and the patient's mother, who carried the mutation, showed skewed X inactivation in her blood cells. Functional studies of the variant were not performed.

Using temperature-induced unfolding, Vinolo et al. (2006) demonstrated that the A281G murine mutation in NEMO, corresponding to the human A288G mutation, causes an important loss in oligomer stability. Fluorescence studies showed that the tyrosine located in the adjacent zinc finger domain exhibits an alteration in its spectral properties. Functional complementation assays using NEMO-deficient pre-B and T lymphocytes showed that the pathogenic mutation reduces TNF-alpha (191160) and lipopolysaccharide-induced NFKB (see 164011) activation by altering the assembly of the IKK complex.


.0021 IMMUNODEFICIENCY 33

IKBKG, GLU315ALA
  
RCV000012222

In 4 affected members of an American family with immunodeficiency-33 (IMD33; 300636), Filipe-Santos et al. (2006) identified a hemizygous c.944A-C transversion in the IKBKG gene, resulting in a glu315-to-ala (E315A) substitution in the leucine zipper domain of the protein. The proband was a non-BCG-vaccinated adolescent with severe, persistent M. avium infection who did not respond well to antibiotic treatment.


.0022 IMMUNODEFICIENCY 33

IKBKG, ARG319GLN
  
RCV000012223

In affected males from 2 unrelated French and German families with immunodeficiency-33 (IMD33; 300636), Filipe-Santos et al. (2006) identified a hemizygous c.956G-A transition in the IKBKG gene, resulting in an arg319-to-gln (R319Q) substitution in the leucine zipper domain of the protein. The proband of the French family was a child with disseminated BCG who was well at age 8 years after treatment with antituberculosis antibiotics. The proband of the German family was a young non-BCG-vaccinated boy diagnosed with mycobacterial disease.


.0023 IMMUNODEFICIENCY 33

IKBKG, ARG173GLY
  
RCV000012224...

In a 4.5-year-old boy, born to unrelated Belgian parents, with immunodeficiency-33 (IMD33; 300636), Ku et al. (2007) identified a hemizygous c.518C-G transversion at the end of exon 4 of the NEMO gene, predicted to result in an arg173-to-gly (R173G) substitution at a conserved residue in the first coiled-coil domain. The mother was heterozygous for this mutation. RT-PCR analysis of patient cells showed the presence of 2 abnormal splicing products corresponding to the skipping of exons 4-6 and exons 5-6. Since the mutation occurred 2 nucleotides away from the end of the exon, it had abnormal splicing effects. Western blot analysis of patient cells showed decreased levels of NEMO compared to controls, suggesting partial NEMO deficiency. Patient fibroblasts showed impaired responses to IL1B and TNFA, suggesting impaired activation of NF-kappa-B. There was also an impaired cellular response to interleukin-1 receptor (IL1R; 147810), Toll-like receptor (TLR; see 601194), and tumor necrosis factor receptor (TNFR; see 191190) stimulation.


.0024 IMMUNODEFICIENCY 33, MALE-RESTRICTED

INCONTINENTIA PIGMENTI, INCLUDED
IKBKG, IVS4DS, C-T, +866
  
RCV000757923...

In 3 boys from 2 unrelated families of European and Japanese descent with fatal immunodeficiency-33 (IMD33; 300636), Boisson et al. (2019) identified a deep intronic mutation (IVS4+866C-T) in the IKBKG gene (chrX.153,787,731C-T, GRCh37). The boy in the European family inherited the mutation from his mother, who had mild features of incontinentia pigmenti (IP; 308300). The mutation in the Japanese boy occurred de novo. The mutation was shown to create a new splicing donor site that resulted in a 44-nucleotide pseudoexon that produced a frameshift. Patient fibroblasts showed decreased levels of mutant transcript due to nonsense-mediated mRNA decay; there were also decreased levels of the mutant protein, suggesting a quantitative defect. The amount of aberrant splicing varied between cell types in the affected boys. In leukocytes, the abnormal splice variant predominated at approximately 97%, while in fibroblasts and iPSC-derived neuronal precursor cells, the abnormal RNA was measured at about 65%. The splicing factor SRSF6 (601944) was shown to bind to the pseudoexon, resulting in its inclusion in the mature mRNA. By knocking down SFSF6 or CLK (see 601951), a protein that phosphorylates SR proteins, the authors were able to restore wildtype expression in mutant cells. The variant segregated with the disorder in both families and was not found in the 1000 Genomes Project or gnomAD databases.


.0025 IMMUNODEFICIENCY 33

IKBKG, 18-BP DEL, NT811
  
RCV001172476

In a 6.5-year-old French boy (patient 1) with immunodeficiency-33 (IMD33; 300636), Ku et al. (2005) identified a hemizygous 18-bp in-frame deletion (c.811_828del) in exon 7 of the IKBKG gene, resulting in the deletion of residues 271 to 276 in the coiled-coil 2 domain. The patient's mother, who had conical teeth, was heterozygous for the mutation; blood analysis showed random X inactivation. Western blot analysis of patient cells showed presence of the IKBKG protein, but fibroblasts responded weakly to stimulation with TNFA and IL1B compared to controls. The findings were consistent with a hypomorphic allele. The patient had hypodontia and conical teeth, but no other features of ectodermal dysplasia.


.0026 IMMUNODEFICIENCY 33

IKBKG, LEU80PRO
  
RCV001172477

In an 11-year-old German boy (patient 2) with ectodermal dysplasia and immunodeficiency-33 (IMD33; 300636), Ku et al. (2005) identified a hemizygous c.239T-C transition in exon 3 of the IKBKG gene, resulting in a leu80-to-pro (L80P) substitution in the first coiled-coil domain. The patient's unaffected mother was heterozygous for the mutation; blood analysis showed skewed X-inactivation. Western blot analysis of patient cells showed presence of the IKBKG protein, but fibroblasts responded weakly to stimulation with TNFA and IL1B compared to controls. The findings were consistent with a hypomorphic allele. The patient had hypodontia and conical teeth, but no other features of ectodermal dysplasia.


.0027 ECTODERMAL DYSPLASIA AND IMMUNODEFICIENCY 1, MALE-RESTRICTED

INCONTINENTIA PIGMENTI, INCLUDED
IKBKG, 2-BP DEL, 1182TT
   RCV001172478...

In a 6-year-old boy with ectodermal dysplasia and immunodeficiency-1 (EDAID1; 300291), Roberts et al. (2010) identified a hemizygous 2-bp deletion (c.1182_1183delTT) in the IKBKG gene, predicted to result in a frameshift and premature termination affecting the zinc finger domain. The mutation was inherited from the patient's mother, who had features of incontinentia pigmenti (IP; 308300). Functional studies of the variant and studies of patient cells were not performed. The boy also had the additional features of osteopetrosis and edema, as well as some skin features of IP. Roberts et al. (2010) described the patient as having OLEDAID.


.0028 ECTODERMAL DYSPLASIA AND IMMUNODEFICIENCY 1, MALE-RESTRICTED

INCONTINENTIA PIGMENTI, INCLUDED
IKBKG, IVS8DS, G-C, +5
  
RCV000523987...

In a male infant with lethal ectodermal dysplasia and immunodeficiency-1 (EDAID1; 300291), Johnston et al. (2016) identified a hemizygous G-to-C transversion (c.1117+5G-C) in intron 8 of the IKBKG gene, predicted to result in a splice site alteration, the skipping of exon 9, and a frameshift with premature termination of the C-terminal region (Arg352SerfsTer22). The mutation was inherited from his mother who had features of incontinentia pigmenti (IP; 308300); X-chromosome inactivation studies showed nonrandom maternal X inactivation with expression only of the wildtype allele. Western blot analysis of the proband's cells showed reduced-sized mutant IKBKG, consistent with the predicted truncated protein.


.0029 IMMUNODEFICIENCY 33

IKBKG, GLU57LYS (rs148695964)
  
RCV000059069...

In a 2-year-old boy with immunodeficiency-33 (IMD33; 300636), Frans et al. (2017) identified a hemizygous c.169G-A transition (c.169G-A, NM_003639) in the IKBKG gene, resulting in a glu57-to-lys (E57K) substitution in the N-terminal domain. His mother, who also carried the mutation, had a history of recurrent sinorespiratory infections but no signs of IP. The variant was present at a frequency of 0.001 in the European population in the ExAC database (August, 2016). Patient peripheral blood cells showed mildly decreased IL6 production after stimulation with IL1B compared to controls. However, NEMO expression in patient fibroblasts was normal, and IKBA was degraded normally upon stimulation with IL1B or TNFA, suggesting a specific effect of the mutation. In vitro functional expression studies in IKBKG-null cells transfected with the mutation showed impaired IL6 production after stimulation with TNFA or IL1B compared to controls. Frans et al. (2017) noted that mutations affecting the N terminus of NEMO tend to lead to decreased production of immunoglobulins; the authors postulated a hypomorphic effect of this variant.


.0030 IMMUNODEFICIENCY 33

IKBKG, c.1-16G-C
  
RCV001172483

In 3 adult males from 2 unrelated families (families A and B) with immunodeficiency-33 (IMD33; 300636) manifest as disseminated mycobacterial infections, Hsu et al. (2018) identified a hemizygous c.1-16G-C transversion at the last base of the first untranslated exon. Analysis of patient cells showed a splicing abnormality, resulting in a 110-bp deletion at the 3-prime end of exon 1. This molecular defect resulted in decreased transcript and protein levels compared to controls (about 30%). Cells derived from the patients in families A and B failed to upregulate cytokines in response to certain TLR agonists, suggesting that this IKBKG mutation is hypomorphic.


.0031 ECTODERMAL DYSPLASIA AND IMMUNODEFICIENCY 1, MALE-RESTRICTED

INCONTINENTIA PIGMENTI, INCLUDED
IKBKG, IVS9DS, G-A, +1
  
RCV001172484...

In a 2-year-old boy (P1) with ectodermal dysplasia and immunodeficiency-1 (EDAID1; 300291), Heller et al. (2020) identified a hemizygous G-to-A transition in intron 9 of the IKBKG gene. The mutation caused the skipping of exon 9, a frameshift, and premature termination (Arg352Serfs373Ter), with loss of the zinc finger domain. Patient cells showed decreased IKBKG levels compared to controls; in vitro studies showed impaired degradation of IKBA and impaired IL6 production upon stimulation with IL1B and TNFA. The patient had a severe phenotype with functional T- and B-cell deficiency. He underwent hematopoietic stem cell transplant. His mother and sister, who were heterozygous for the mutation, had incontinentia pigmenti (IP; 308300).


.0032 IMMUNODEFICIENCY 33

IKBKG, ASP113ASN
  
RCV000059070...

In a boy (patient 1) with immunodeficiency-33 (IMD33; 300636), Abbott et al. (2014) identified a hemizygous c.337G-A transition in the IKBKG gene, resulting in an asp113-to-asn (D113N) substitution in the first coiled-coil domain. The patient had impaired NK cell function and impaired T-cell receptor signaling with decreased NFKB activity, but Toll-like receptor signaling appeared to be intact. He underwent successful bone marrow transplant. The patient had previously been reported in detail by Salt et al. (2008); his unaffected mother also carried the mutation.

Heller et al. (2020) identified a hemizygous D113N mutation in a boy with IMD33. His mother and grandmother, who were presumably unaffected, carried the heterozygous mutation. However, the proband's 40-year-old male cousin, who did not have recurrent infections and had normal response to polysaccharide antibodies, was hemizygous for D113N. Heller et al. (2020) noted that the allele frequency for this variant is rather high (0.009572), and that some suggest it may be a polymorphism (see Fusco et al., 2004).


.0033 AUTOINFLAMMATORY DISEASE, SYSTEMIC, X-LINKED

IKBKG, 597G-A
  
RCV001762996...

In a boy (P1) with X-linked systemic autoinflammatory disease (SAIDX; 301081), de Jesus et al. (2020) identified a de novo hemizygous c.597G-A transition in exon 5 of the IKBKG gene, predicted to result in a silent substitution (V199V), but also predicted to disrupt an exonic splicing enhancer and an exon-identity element in exon 5, resulting in alternative splicing. The patient had lymphohistiocytic panniculitis, chorioretinitis, progressive B cell lymphopenia, hypogammaglobulinemia, and conical teeth, prompting targeted sequencing of the IKBKG gene. Functional studies of the variant were not performed.

In a 9-year-old boy (P1) with SAIDX, Lee et al. (2022) identified a de novo heterozygous c.597G-A transition in the IKBKG gene. Analysis of patient cells detected a shorter NEMO mRNA splice variant lacking the 153 nucleotides of exon 5. Patient cells showed a higher ratio of mutant to full-length cDNA compared to healthy controls. The mutation, which was found by targeted sequencing of NEMO and further confirmed by Sanger sequencing of genomic DNA, was not present in 1,150 healthy individuals. In vitro functional expression studies in patient peripheral blood cells showed that stimulation with poly(I:C) or viral infection resulted in enhanced type I IFN production likely due to a stabilized complex of mutant IKBKG and IKKi (605048). The patient had panniculitis, optic neuritis, chorioretinitis, hypogammaglobulinemia, conical teeth, and CNS hemorrhage.


.0034 AUTOINFLAMMATORY DISEASE, SYSTEMIC, X-LINKED

IKBKG, IVS5DS, T-G, +2
  
RCV002250446

In a boy (P3) with X-linked systemic autoinflammatory disease (SAIDX; 301081), de Jesus et al. (2020) identified a de novo hemizygous T-to-G transversion in intron 5 of the IKBKG gene (c.671+2T-G), predicted to result in the deletion of exon 5. Functional studies of the variant were not performed. The patient had panniculitis, progressive B cell lymphopenia, thrombocytopenia, and hypogammaglobulinemia without major infections.

In a 5-year-old boy (P2) with SAIDX, Lee et al. (2022) identified a de novo hemizygous T-to-G transversion in intron 5 of the IKBKG gene (chrX:153,788,776T-G), disrupting a splice site and resulting in the production of a mutant transcript lacking exon 5. Analysis of patient cells detected a shorter NEMO mRNA splice variant lacking the 153 nucleotides of exon 5. Patient cells showed a higher ratio of mutant to full-length cDNA compared to healthy controls. The mutation, which was found by targeted sequencing of NEMO and further confirmed by Sanger sequencing of genomic DNA, was not present in 1,150 healthy individuals. In vitro functional expression studies in patient peripheral blood cells showed that stimulation with poly(I:C) or viral infection resulted in enhanced type I IFN production likely due to a stabilized complex of mutant IKBKG and IKKi (605048). The patient had lymphocytic panniculitis, hypogammaglobulinemia, and a CNS cortical bleed.


.0035 AUTOINFLAMMATORY DISEASE, SYSTEMIC, X-LINKED

IKBKG, IVS5DS, G-A, +5
  
RCV002250447

In a boy (P2) with X-linked systemic autoinflammatory disease (SAIDX; 301081), de Jesus et al. (2020) identified a de novo hemizygous G-to-A transition in intron 5 of the IKBKG gene (c.671+5G-A), predicted to result in the deletion of exon 5. Functional studies of the variant were not performed. The patient had panniculitis, uveitis, progressive B cell lymphopenia, and hypogammaglobulinemia without major infections.

In a 3-year-old boy (P3) with SAIDX, Lee et al. (2022) identified a de novo heterozygous G-to-A transition (chrX:153,788,779G-A) in the IKBKG gene, resulting in a splice site alteration and the deletion of exon 5. Analysis of patient cells detected a shorter NEMO mRNA splice variant lacking the 153 nucleotides of exon 5. Patient cells showed a higher ratio of mutant to full-length cDNA compared to healthy controls. The mutation, which was found by targeted sequencing of NEMO and further confirmed by Sanger sequencing of genomic DNA, was not present in 1,150 healthy individuals. In vitro functional expression studies in patient peripheral blood cells showed that stimulation with poly(I:C) or viral infection resulted in enhanced type I IFN production likely due to a stabilized complex of mutant IKBKG and IKKi (605048). The patient had panniculitis, uveitis, lymphopenia, hypogammaglobulinemia, and a CNS hemorrhage.


.0036 AUTOINFLAMMATORY DISEASE, SYSTEMIC, X-LINKED

IKBKG, IVS5AS, A-G, -2
  
RCV002250444

In a girl (P4) with X-linked systemic autoinflammatory disease (SAIDX; 301081), de Jesus et al. (2020) identified a de novo heterozygous A-to-G transition in intron 5 of the IKBKG gene (c.519-2A-G), predicted to result in the deletion of exon 5. Functional studies of the variant were not performed. The patient had panniculitis, lipodystrophy, anemia, myositis, and progressive B cell lymphopenia without hypogammaglobulinemia.


REFERENCES

  1. Abbott, J. K., Quinones, R. R., de la Morena, M. T., Gelfand, E. W. Successful hematopoietic cell transplantation in patients with unique NF-kappa-B essential modulator (NEMO) mutations. (Letter) Bone Marrow Transplant. 49: 1446-1447, 2014. [PubMed: 25068423, related citations] [Full Text]

  2. Akiyama, M., Aranami, A., Sasaki, Y., Ebihara, T., Sugiura, M. Familial linear and whorled nevoid hypermelanosis. J. Am. Acad. Derm. 30: 831-833, 1994. [PubMed: 8169255, related citations] [Full Text]

  3. Aradhya, S., Bardaro, T., Galgoczy, P., Yamagata, T., Esposito, T., Patlan, H., Ciccodicola, A., Munnich, A., Kenwrick, S., Platzer, M., D'Urso, M., Nelson, D. L. Multiple pathogenic and benign genomic rearrangements occur at a 35 kb duplication involving the NEMO and LAGE2 genes. Hum. Molec. Genet. 10: 2557-2567, 2001. [PubMed: 11709543, related citations] [Full Text]

  4. Aradhya, S., Courtois, G., Rajkovic, A., Lewis, R. A., Levy, M., Israel, A., Nelson, D. L. Atypical forms of incontinentia pigmenti in male individuals result from mutations of a cytosine tract in exon 10 of NEMO (IKK-gamma). Am. J. Hum. Genet. 68: 765-771, 2001. [PubMed: 11179023, images, related citations] [Full Text]

  5. Aradhya, S., Woffendin, H., Jakins, T., Bardaro, T., Esposito, T., Smahi, A., Shaw, C., Levy, M., Munnich, A., D'Urso, M., Lewis, R. A., Kenwrick, S., Nelson, D. L. A recurrent deletion in the ubiquitously expressed NEMO (IKK-gamma) gene accounts for the vast majority of incontinentia pigmenti mutations. Hum. Molec. Genet. 10: 2171-2179, 2001. [PubMed: 11590134, related citations] [Full Text]

  6. Bardaro, T., Falco, G., Sparago, A., Mercadante, V., Molins, E. G., Tarantino, E., Ursini, M. V., D'Urso, M. Two cases of misinterpretation of molecular results in incontinentia pigmenti, and a PCR-based method to discriminate NEMO/IKK-gamma gene deletion. Hum. Mutat. 21: 8-11, 2003. [PubMed: 12497627, related citations] [Full Text]

  7. Boisson, B., Honda, Y., Ajiro, M., Bustamante, J., Bendavid, M., Gennery, A. R., Kawasaki, Y., Ichishima, J., Osawa, M., Nihira, H., Shiba, T., Tanaka, T., and 16 others. Rescue of recurrent deep intronic mutation underlying cell type-dependent quantitative NEMO deficiency. J. Clin. Invest. 129: 583-597, 2019. [PubMed: 30422821, images, related citations] [Full Text]

  8. Brummelkamp, T. R., Nijman, S. M. B., Dirac, A. M. G., Bernards, R. Loss of the cylindromatosis tumour suppressor inhibits apoptosis by activating NF-kappa-B. Nature 424: 797-801, 2003. [PubMed: 12917690, related citations] [Full Text]

  9. Chiu, J. C., Ko, H. W., Edery, I. NEMO/NLK phosphorylates PERIOD to initiate a time-delay phosphorylation circuit that sets circadian clock speed. Cell 145: 357-370, 2011. Note: Erratum: Cell 145: 635 only, 2011. [PubMed: 21514639, images, related citations] [Full Text]

  10. de Jesus, A. A., Hou, Y., Brooks, S., Malle, L., Biancotto, A., Huang, Y., Calvo, K. R., Marrero, B., Moir, S., Oler, A. J., Deng, Z., Montealegre Sanchez, G. A., and 75 others. Distinct interferon signatures and cytokine patterns define additional systemic autoinflammatory diseases. J. Clin. Invest. 130: 1669-1682, 2020. [PubMed: 31874111, images, related citations] [Full Text]

  11. Doffinger, R., Smahi, A., Bessia, C., Geissmann, F., Feinberg, J., Durandy, A., Bodemer, C., Kenwrick, S., Dupuis-Girod, S., Blanche, S., Wood, P., Rabia, S. H., and 16 others. X-linked anhidrotic ectodermal dysplasia with immunodeficiency is caused by impaired NF-kappa-B signaling. Nature Genet. 27: 277-285, 2001. [PubMed: 11242109, related citations] [Full Text]

  12. Filipe-Santos, O., Bustamante, J., Haverkamp, M. H., Vinolo, E., Ku, C.-L., Puel, A., Frucht, D. M., Christel, K., von Bernuth, H., Jouanguy, E., Feinberg, J., Durandy, A., and 25 others. X-linked susceptibility to mycobacteria is caused by mutations in NEMO impairing CD40-dependent IL-12 production. J. Exp. Med. 203: 1745-1759, 2006. [PubMed: 16818673, images, related citations] [Full Text]

  13. Frans, G., van der Werff Ten Bosch, J., Moens, L., Gijsbers, R., Changi-Ashtiani, M., Rokni-Zadeh, H., Shahrooei, M., Wuyts, G., Meyts, I., Bossuyt, X. Functional evaluation of an IKBK variant suspected to cause immunodeficiency without ectodermal dysplasia. J. Clin. Immun. 37: 801-810, 2017. [PubMed: 28993958, related citations] [Full Text]

  14. Fusco, F., Bardaro, T., Fimiani, G., Mercadante, V., Miano, M. G., Falco, G., Israel, A., Courtois, G., D'Urso, M., Ursini, M. V. Molecular analysis of the genetic defect in a large cohort of IP patients and identification of novel NEMO mutations interfering with NF-kappa-B activation. Hum. Molec. Genet. 13: 1763-1773, 2004. [PubMed: 15229184, related citations] [Full Text]

  15. Fusco, F., Paciolla, M., Napolitano, F., Pescatore, A., D'Addario, I., Bal, E., Lioi, M. B., Smahl, A., Miano, M. G., Ursini, M. V. Genomic architecture at the incontinentia pigmenti locus favours de novo pathological alleles through different mechanisms. Hum. Molec. Genet. 21: 1260-1271, 2012. [PubMed: 22121116, related citations] [Full Text]

  16. Gautheron, J., Pescatore, A., Fusco, F., Esposito, E., Yamaoka, S., Agou, F., Ursini, M. V., Courtois, G. Identification of a new NEMO/TRAF6 interface affected in incontinentia pigmenti pathology. Hum. Molec. Genet. 19: 3138-3149, 2010. [PubMed: 20529958, related citations] [Full Text]

  17. Gerlach, B., Cordier, S. M., Schmukle, A. C., Emmerich, C. H., Rieser, E., Haas, T. L., Webb, A. I., Rickard, J. A., Anderton, H., Wong, W. W.-L., Nachbur, U., Gangoda, L., Warnken, U., Purcell, A. W., Silke, J., Walczak, H. Linear ubiquitination prevents inflammation and regulates immune signalling. Nature 471: 591-596, 2011. [PubMed: 21455173, related citations] [Full Text]

  18. Heller, S., Kolsch, U., Magg, T., Kruger, R., Scheuern, A., Schneider, H., Eichinger, A., Wahn, V., Unterwalder, N., Lorenz, M., Schwarz, K., Meisel, C., Schulz, A., Hauck, F., von Bernuth, H. T cell impairment is predictive for a severe clinical course in NEMO deficiency. J. Clin. Immun. 40: 421-434, 2020. [PubMed: 31965418, related citations] [Full Text]

  19. Hsu, A. P., Zerbe, C. S., Foruraghi, L., Iovine, N. L., Leiding, J. W., Mushatt, D. M., Wild, L., Kuhns, D. B., Holland, S. M. IKBKG (NEMO) 5-prime untranslated splice mutations lead to severe, chronic disseminated mycobacterial infections. Clin. Infect. Dis. 67: 456-459, 2018. [PubMed: 29534156, related citations] [Full Text]

  20. Ikeda, F., Deribe, Y. L., Skanland, S. S., Stieglitz, B., Grabbe, C., Franz-Wachtel, M., van Wijk, S. J. L., Goswami, P., Nagy, V., Terzic, J., Tokunaga, F., Androulidaki, A., Nakagawa, T., Pasparakis, M., Iwai, K., Sundberg, J. P., Schaefer, L., Rittinger, K., Macek, B., Dikic, I. SHARPIN forms a linear ubiquitin ligase complex regulating NF-kappa-B activity and apoptosis. Nature 471: 637-641, 2011. [PubMed: 21455181, images, related citations] [Full Text]

  21. Jain, A., Ma, C. A., Liu, S., Brown, M., Cohen, J., Strober, W. Specific missense mutations in NEMO result in hyper-IgM syndrome with hypohydrotic (sic) ectodermal dysplasia. Nature Immun. 2: 223-228, 2001. [PubMed: 11224521, related citations] [Full Text]

  22. Jain, A., Ma, C. A., Lopez-Granados, E., Means, G., Brady, W., Orange, J. S., Liu, S., Holland, S., Derry, J. M. J. Specific NEMO mutations impair CD40-mediated c-Rel activation and B cell terminal differentiation. J. Clin. Invest. 114: 1593-1602, 2004. [PubMed: 15578091, images, related citations] [Full Text]

  23. Jin, D. Y., Jeang, K. T. Isolation of full-length cDNA and chromosomal localization of human NF-kappaB modulator NEMO to Xq28. J. Biomed. Sci. 6: 115-120, 1999. [PubMed: 10087442, related citations] [Full Text]

  24. Johnston, A. M., Niemela, J., Rosenzweig, S. D., Fried, A. J., Delmonte, O. M., Fleisher, T. A., Kuehn, H. A novel mutation in IKBKG/NEMO leads to ectodermal dysplasia with severe immunodeficiency (EDA-ID). (Letter) J. Clin. Immun 36: 541-543, 2016. [PubMed: 27368913, images, related citations] [Full Text]

  25. Kosaki, K., Shimasaki, N., Fukushima, H., Hara, M., Ogata, T., Matsuo, N. Female patient showing hypohidrotic ectodermal dysplasia and immunodeficiency (HED-ID). (Letter) Am. J. Hum. Genet. 69: 664-665, 2001. [PubMed: 11484156, related citations] [Full Text]

  26. Kovalenko, A., Chable-Bessia, C., Cantarella, G., Israel, A., Wallach, D., Courtois, G. The tumour suppressor CYLD negatively regulates NF-kappa-B signalling by deubiquitination. Nature 424: 801-805, 2003. [PubMed: 12917691, related citations] [Full Text]

  27. Kozak, M. Pushing the limits of the scanning mechanism for initiation of translation. Gene 299: 1-34, 2002. [PubMed: 12459250, images, related citations] [Full Text]

  28. Ku, C.-L., Dupuis-Girod, S., Dittrich, A.-M., Bustamante, J., Santos, O. F., Schulze, I., Bertrand, Y., Couly, G., Bodemer, C., Bossuyt, X., Picard, C., Casanova, J.-L. NEMO mutations in 2 unrelated boys with severe infections and conical teeth. Pediatrics 115: e615-e619, 2005. Note: Electronic Article. [PubMed: 15833888, related citations] [Full Text]

  29. Ku, C.-L., Picard, C., Erdos, M., Jeurissen, A., Bustamante, J., Puel, A., von Bernuth, H., Filipe-Santos, O., Chang, H.-H., Lawrence, T., Raes, M., Marodi, L., Bossuyt, X., Casanova, J.-L. IRAK4 and NEMO mutations in otherwise healthy children with recurrent invasive pneumococcal disease. J. Med. Genet. 44: 16-23, 2007. [PubMed: 16950813, images, related citations] [Full Text]

  30. Lee, Y., Wessel, A. W., Xu, J., Reinke, J. G., Lee, E., Kim, S. M., Hsu, A. P., Zilberman-Rudenko, J., Cao, S., Enos, C., Brooks, S. R., Deng, Z., and 11 others. Genetically programmed alternative splicing of NEMO mediates an autoinflammatory disease phenotype. J. Clin. Invest. 132: e128808, 2022. [PubMed: 35289316, images, related citations] [Full Text]

  31. Li, Q., Van Antwerp, D., Mercurio, F., Lee, K.-F., Verma, I. M. Severe liver degeneration in mice lacking the I-kappa-B kinase 2 gene. Science 284: 321-325, 1999. [PubMed: 10195897, related citations] [Full Text]

  32. Li, Y., Kang, J., Friedman, J., Tarassishin, L., Ye, J., Kovalenko, A., Wallach, D., Horwitz, M. S. Identification of a cell protein (FIP-3) as a modulator of NF-kappa-B activity and as a target of an adenovirus inhibitor of tumor necrosis factor alpha-induced apoptosis. Proc. Nat. Acad. Sci. 96: 1042-1047, 1999. [PubMed: 9927690, images, related citations] [Full Text]

  33. Lie, S. O., Froland, S., Brandtzaeg, P., Vandvik, B., Steen-Johnsen, J. Transient B cell immaturity with intractable diarrhoea: a possible new immunodeficiency syndrome. J. Inherit. Metab. Dis. 1: 137-143, 1978. [PubMed: 117248, related citations] [Full Text]

  34. Makris, C., Godfrey, V. L., Krahn-Senftleben, G., Takahashi, T., Roberts, J. L., Schwarz, T., Feng, L., Johnson, R. S., Karin, M. Female mice heterozygous for IKK-gamma/NEMO deficiencies develop a dermatopathy similar to the human X-linked disorder incontinentia pigmenti. Molec. Cell 5: 969-979, 2000. [PubMed: 10911991, related citations] [Full Text]

  35. Martinez-Pomar, N., Munoz-Saa, I., Heine-Suner, D., Martin, A., Smahi, A., Matamoros, N. A new mutation in exon 7 of NEMO gene: late skewed X-chromosome inactivation in an incontinentia pigmenti female patient with immunodeficiency. Hum. Genet. 118: 458-465, 2005. [PubMed: 16228229, related citations] [Full Text]

  36. Matsuzawa, A., Tseng, P.-H., Vallabhapurapu, S., Luo, J.-L., Zhang, W., Wang, H., Vignali, D. A. A., Gallagher, E., Karin, M. Essential cytoplasmic translocation of a cytokine receptor-assembled signaling complex. Science 321: 663-668, 2008. Note: Erratum: Science 322: 375 only, 2008. [PubMed: 18635759, images, related citations] [Full Text]

  37. May, M. J., D'Acquisto, F., Madge, L. A., Glockner, J., Pober, J. S., Ghosh, S. Selective inhibition of NF-kappa-B activation by a peptide that blocks the interaction of NEMO with the I-kappa-B kinase complex. Science 289: 1550-1554, 2000. [PubMed: 10968790, related citations] [Full Text]

  38. Nenci, A., Becker, C., Wullaert, A., Gareus, R., van Loo, G., Danese, S., Huth, M., Nikolaev, A., Neufert, C., Madison, B., Gumucio, D., Neurath, M. F., Pasparakis, M. Epithelial NEMO links innate immunity to chronic intestinal inflammation. Nature 446: 557-561, 2007. [PubMed: 17361131, related citations] [Full Text]

  39. Niehues, T., Reichenbach, J., Neubert, J., Gudowius, S., Puel, A., Horneff, G., Lainka, E., Dirksen, U., Schroten, H., Doffinger, R., Casanova, J. L., Wahn, V. Nuclear factor kappa-B essential modulator-deficient child with immunodeficiency yet without anhidrotic ectodermal dysplasia. J. Allergy Clin. Immun. 114: 1456-1462, 2004. [PubMed: 15577852, related citations] [Full Text]

  40. Nishikomori, R., Akutagawa, H., Maruyama, K., Nakata-Hizume, M., Ohmori, K., Mizuno, K., Yachie, A., Yasumi, T., Kusunoki, T., Heike, T., Nakahata, T. X-linked ectodermal dysplasia and immunodeficiency caused by reversion mosaicism of NEMO reveals a critical role for NEMO in human T-cell development and/or survival. Blood 103: 4565-4572, 2004. [PubMed: 14726382, related citations] [Full Text]

  41. Orange, J. S., Brodeur, S. R., Jain, A., Bonilla, F. A., Schneider, L. C., Kretschmer, R., Nurko, S., Rasmussen, W. L., Kohler, J. R., Gellis, S. E., Ferguson, B. M., Strominger, J. L., Zonana, J., Ramesh, N., Ballas, Z. K., Geha, R. S. Deficient natural killer cell cytotoxicity in patients with IKK-gamma/NEMO mutations. J. Clin. Invest. 109: 1501-1509, 2002. [PubMed: 12045264, images, related citations] [Full Text]

  42. Orange, J. S., Jain, A., Ballas, Z. K., Schneider, L. C., Geha, R. S., Bonilla, F. A. The presentation and natural history of immunodeficiency caused by nuclear factor kappa-B essential modulator mutation. J. Allergy Clin. Immun. 113: 725-733, 2004. [PubMed: 15100680, related citations] [Full Text]

  43. Orange, J. S., Levy, O., Brodeur, S. R., Krzewski, K., Roy, R. M., Niemela, J. E., Fleisher, T. A., Bonilla, F. A., Geha, R. S. Human nuclear factor kappa-B essential modulator mutation can result in immunodeficiency without ectodermal dysplasia. J. Allergy Clin. Immun. 114: 650-656, 2004. [PubMed: 15356572, related citations] [Full Text]

  44. Orstavik, K. H., Kristiansen, M., Knudsen, G. P., Storhaug, K., Vege, A., Eiklid, K., Abrahamsen, T. G., Smahi, A., Steen-Johnsen, J. Novel splicing mutation in the NEMO (IKK-gamma) gene with severe immunodeficiency and heterogeneity of X-chromosome inactivation. Am. J. Med. Genet. 140A: 31-39, 2006. [PubMed: 16333836, related citations] [Full Text]

  45. Puel, A., Reichenbach, J., Bustamante, J., Ku, C.-L., Feinberg, J., Doffinger, R., Bonnet, M., Filipe-Santos, O., de Beaucoudrey, L., Durandy, A., Horneff, G., Novelli, F., Wahn, V., Smahi, A., Israel, A., Niehues, T., Casanova, J.-L. The NEMO mutation creating the most-upstream premature stop codon is hypomorphic because of a reinitiation of translation. Am. J. Hum. Genet. 78: 691-701, 2006. [PubMed: 16532398, images, related citations] [Full Text]

  46. Roberts, C. M. L., Angus, J. E., Leach, I. H., McDermott, E. M., Walker, D. A., Ravenscroft, J. C. A novel NEMO gene mutation causing osteopetrosis, lymphoedema, hypohidrotic ectodermal dysplasia and immunodeficiency (OL-HED-ID). Europ. J. Pediat. 169: 1403-1407, 2010. [PubMed: 20499091, related citations] [Full Text]

  47. Roberts, J. L., Morrow, B., Vega-Rich, C., Salafia, C. M., Nitowsky, H. M. Incontinentia pigmenti in a newborn male infant with DNA confirmation. Am. J. Med. Genet. 75: 159-163, 1998. [PubMed: 9450877, related citations] [Full Text]

  48. Rothwarf, D. M., Zandi, E., Natoli, G., Karin, M. IKK-gamma is an essential regulatory subunit of the I-kappa-B kinase complex. Nature 395: 297-300, 1998. [PubMed: 9751060, related citations] [Full Text]

  49. Rudolph, D., Yeh, W.-C., Wakeham, A., Rudolph, B., Nallainathan, D., Potter, J., Elia, A. J., Mak, T. W. Severe liver degeneration and lack of NF-kappa-B activation in NEMO/IKK-gamma-deficient mice. Genes Dev. 14: 854-862, 2000. [PubMed: 10766741, images, related citations]

  50. Salt, B. H., Niemela, J. E., Pandey, R., Hanson, E. P., Deering, R. P., Quinones, R., Jain, A., Orange, J. S., Gelfand, E. W. IKBKG (nuclear factor-kappa-B essential modulator) mutation can be associated with opportunistic infection without impairing Toll-like receptor function. J. Allergy Clin. Immun. 121: 976-982, 2008. [PubMed: 18179816, images, related citations] [Full Text]

  51. Schmidt-Supprian, M., Bloch, W., Courtois, G., Addicks, K., Israel, A., Rajewsky, K., Pasparakis, M. NEMO/IKK-gamma-deficient mice model incontinentia pigmenti. Molec. Cell 5: 981-992, 2000. [PubMed: 10911992, related citations] [Full Text]

  52. Siggs, O. M., Berger, M., Krebs, P., Arnold, C. N., Eidenschenk, C., Huber, C., Pirie, E., Smart, N. G., Khovananth, K., Xia, Y., McInerney, G., Hedestam, G. B. K., Nemazee, D., Beutler, B. A mutation of Ikbkg causes immune deficiency without impairing degradation of I-kappa-B-alpha. Proc. Nat. Acad. Sci. 107: 3046-3051, 2010. [PubMed: 20133626, images, related citations] [Full Text]

  53. Singh, A., Zarember, K. A., Kuhns, D. B., Gallin, J. I. Impaired priming and activation of the neutrophil NADPH oxidase in patients with IRAK4 or NEMO deficiency. J. Immun. 182: 6410-6417, 2009. [PubMed: 19414794, images, related citations] [Full Text]

  54. Takada, H., Nomura, A., Ishimura, M., Ichiyama, M., Ohga, S., Hara, T. NEMO mutation as a cause of familial occurrence of Behcet's disease in female patients. Clin. Genet. 78: 575-579, 2010. [PubMed: 20412081, related citations] [Full Text]

  55. The International Incontinentia Pigmenti Consortium. Genomic rearrangement in NEMO impairs NF-kappa-B activation and is a cause of incontinentia pigmenti. Nature 405: 466-472, 2000. [PubMed: 10839543, related citations] [Full Text]

  56. The International IP Consortium. Survival of male patients with incontinentia pigmenti carrying a lethal mutation can be explained by somatic mosaicism or Klinefelter syndrome. Am. J. Hum. Genet. 69: 1210-1217, 2001. [PubMed: 11673821, images, related citations] [Full Text]

  57. Tokunaga, F., Nakagawa, T., Nakahara, M., Saeki, Y., Taniguchi, M., Sakata, S., Tanaka, K., Nakano, H., Iwai, K. SHARPIN is a component of the NF-kappa-B-activating linear ubiquitin chain assembly complex. Nature 471: 633-636, 2011. [PubMed: 21455180, related citations] [Full Text]

  58. Vinolo, E., Sebban, H., Chaffotte, A., Israel, A., Courtois, G., Veron, M., Agou, F. A point mutation in NEMO associated with anhidrotic ectodermal dysplasia with immunodeficiency pathology results in destabilization of the oligomer and reduces lipopolysaccharide- and tumor necrosis factor-mediated NF-kappa-B activation. J. Biol. Chem. 281: 6334-6348, 2006. [PubMed: 16379012, related citations] [Full Text]

  59. Wu, Z.-H., Shi, Y., Tibbetts, R. S., Miyamoto, S. Molecular linkage between the kinase ATM and NF-kappaB signaling in response to genotoxic stimuli. Science 311: 1141-1146, 2006. [PubMed: 16497931, related citations] [Full Text]

  60. Yamaoka, S., Courtois, G., Bessia, C., Whiteside, S. T., Weil, R., Agou, F., Kirk, H. E., Kay, R. J., Israel, A. Complementation cloning of NEMO, a component of the I-kappa-B kinase complex essential for NF-kappa-B activation. Cell 93: 1231-1240, 1998. [PubMed: 9657155, related citations] [Full Text]

  61. Zonana, J., Elder, M. E., Schneider, L. C., Orlow, S. J., Moss, C., Golabi, M., Shapira, S. K., Farndon, P. A., Wara, D. W., Emmal, S. A., Ferguson, B. M. 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. 67: 1555-1562, 2000. [PubMed: 11047757, images, related citations] [Full Text]

  62. Zonana, J., Ferguson, B. Reply to Kosaki et al. Am. J. Hum. Genet. 69: 665-666, 2001.


Cassandra L. Kniffin - updated : 05/24/2022
Cassandra L. Kniffin - updated : 06/03/2020
Alan F. Scott - updated : 02/19/2019
Matthew B. Gross - updated : 9/8/2014
George E. Tiller - updated : 8/28/2013
Patricia A. Hartz - updated : 7/11/2013
Marla J. F. O'Neill - updated : 5/18/2012
Patricia A. Hartz - updated : 4/6/2012
Paul J. Converse - updated : 3/15/2012
Ada Hamosh - updated : 7/19/2011
Paul J. Converse - updated : 2/28/2011
Paul J. Converse - updated : 8/28/2008
Ada Hamosh - updated : 4/12/2007
Victor A. McKusick - updated : 2/21/2007
Paul J. Converse - updated : 2/5/2007
George E. Tiller - updated : 1/16/2007
Marla J. F. O'Neill - updated : 6/23/2006
Ada Hamosh - updated : 4/19/2006
Victor A. McKusick - updated : 3/15/2006
Marla J. F. O'Neill - updated : 2/15/2006
Marla J. F. O'Neill - updated : 1/25/2006
Paul J. Converse - updated : 9/22/2005
Paul J. Converse - updated : 2/3/2005
Victor A. McKusick - updated : 8/23/2004
Ada Hamosh - updated : 8/27/2003
Ada Hamosh - updated : 8/26/2003
Victor A. McKusick - updated : 1/14/2003
George E. Tiller - updated : 5/14/2002
George E. Tiller - updated : 2/13/2002
Victor A. McKusick - updated : 12/20/2001
Victor A. McKusick - updated : 9/27/2001
Victor A. McKusick - updated : 3/20/2001
Paul J. Converse - updated : 3/6/2001
Carol A. Bocchini - updated : 12/20/2000
Victor A. McKusick - updated : 12/13/2000
Paul J. Converse - updated : 8/31/2000
Stylianos E. Antonarakis - updated : 8/3/2000
Ada Hamosh - updated : 5/24/2000
Creation Date:
Ada Hamosh : 5/24/2000
carol : 12/14/2023
carol : 06/02/2022
carol : 06/01/2022
alopez : 05/27/2022
ckniffin : 05/24/2022
carol : 06/23/2020
mgross : 06/22/2020
carol : 06/10/2020
carol : 06/09/2020
ckniffin : 06/03/2020
carol : 02/20/2019
carol : 02/19/2019
carol : 11/15/2018
carol : 11/09/2018
carol : 09/13/2017
carol : 05/19/2015
mgross : 9/8/2014
carol : 9/16/2013
tpirozzi : 8/30/2013
tpirozzi : 8/29/2013
tpirozzi : 8/28/2013
mgross : 7/16/2013
mgross : 7/11/2013
terry : 3/15/2013
carol : 5/22/2012
terry : 5/18/2012
mgross : 5/15/2012
terry : 5/1/2012
terry : 4/6/2012
mgross : 3/21/2012
terry : 3/15/2012
terry : 7/25/2011
alopez : 7/19/2011
joanna : 6/6/2011
mgross : 3/1/2011
terry : 2/28/2011
alopez : 11/18/2008
mgross : 8/28/2008
wwang : 7/3/2008
ckniffin : 6/26/2008
alopez : 4/12/2007
carol : 4/12/2007
wwang : 4/3/2007
mgross : 3/22/2007
alopez : 2/26/2007
terry : 2/21/2007
mgross : 2/5/2007
mgross : 2/5/2007
wwang : 1/23/2007
terry : 1/16/2007
terry : 11/3/2006
wwang : 6/26/2006
terry : 6/23/2006
alopez : 4/20/2006
terry : 4/19/2006
alopez : 3/20/2006
terry : 3/15/2006
wwang : 2/22/2006
terry : 2/15/2006
wwang : 2/2/2006
terry : 1/25/2006
mgross : 10/4/2005
terry : 9/22/2005
mgross : 2/3/2005
tkritzer : 9/1/2004
terry : 8/23/2004
alopez : 11/25/2003
alopez : 8/27/2003
alopez : 8/27/2003
alopez : 8/27/2003
terry : 8/26/2003
cwells : 1/15/2003
terry : 1/14/2003
cwells : 5/17/2002
cwells : 5/14/2002
carol : 4/8/2002
cwells : 2/19/2002
cwells : 2/13/2002
alopez : 2/4/2002
cwells : 1/7/2002
terry : 12/20/2001
carol : 10/5/2001
carol : 10/5/2001
mcapotos : 10/4/2001
terry : 9/27/2001
carol : 4/26/2001
alopez : 4/24/2001
alopez : 4/24/2001
cwells : 4/5/2001
cwells : 3/30/2001
terry : 3/20/2001
mgross : 3/6/2001
mgross : 3/6/2001
alopez : 3/2/2001
terry : 3/1/2001
carol : 12/20/2000
terry : 12/13/2000
alopez : 8/31/2000
mgross : 8/3/2000
mgross : 8/3/2000
alopez : 5/24/2000
alopez : 5/24/2000
alopez : 5/24/2000
alopez : 5/24/2000

* 300248

INHIBITOR OF NUCLEAR FACTOR KAPPA-B KINASE, REGULATORY SUBUNIT GAMMA; IKBKG


Alternative titles; symbols

INHIBITOR OF KAPPA LIGHT POLYPEPTIDE GENE ENHANCER IN B CELLS, KINASE OF, GAMMA
NF-KAPPA-B ESSENTIAL MODULATOR; NEMO
IKK-GAMMA
FIP3


HGNC Approved Gene Symbol: IKBKG

SNOMEDCT: 367520004;   ICD10CM: Q82.3;  


Cytogenetic location: Xq28     Genomic coordinates (GRCh38): X:154,541,238-154,565,046 (from NCBI)


Gene-Phenotype Relationships

Location Phenotype Phenotype
MIM number
Inheritance Phenotype
mapping key
Xq28 Autoinflammatory disease, systemic, X-linked 301081 X-linked 3
Ectodermal dysplasia and immunodeficiency 1 300291 X-linked recessive 3
Immunodeficiency 33 300636 X-linked recessive 3
Incontinentia pigmenti 308300 X-linked dominant 3

TEXT

Description

The IKBKG gene, also known as NEMO, encodes the regulatory gamma subunit of the IKB kinase (IKK) complex. Along with the other 2 subunits, IKBKA (CHUK; 600664) and IKBKB (603258), this IKK complex leads to the proteolytic degradation of the NFKB (see, e.g., 164011) inhibitor NFKBIA (164008), thus enabling NFKB to translocate to the nucleus and activate the transcription of cytokine-associated genes (summary by Heller et al., 2020).

IKBKG, or NEMO, is the founding member of an evolutionarily conserved family of NEMO-like kinases that function in numerous cell signaling pathways. NEMO-like kinases specifically phosphorylate serine or threonine residues that are followed by a proline residue (Chiu et al., 2011).


Cloning and Expression

Yamaoka et al. (1998) characterized a mutant cell line, 5R, originally isolated as a cellular flat variant of Rat-1 fibroblasts transformed by the Tax protein of human T-cell leukemia virus type 1 (HTLV-1). The 5R cell line was unresponsive to all tested NF-kappa-B (NFKB; see 164011)-activating stimuli. Using a genetic complementation approach, Yamaoka et al. (1998) cloned a component of the I-kappa-B kinase complex that they termed NEMO for 'NF-kappa-B essential modulator' from 5R cells. The 2.8-kb NEMO cDNA encodes a 412-amino acid protein that is acidic, rich in glutamic acid and glutamine residues (each 13%), and contains a leucine zipper motif at amino acids 315-342. Yamaoka et al. (1998) determined that the defective phenotype of 5R cells resulted from the absence of the NEMO protein. NEMO also complemented the 1.3E2 mutant cell line, in which NFKB is not activated in response to a large set of stimuli.

By using a monoclonal antibody against IKK-alpha, Rothwarf et al. (1998) purified the IKK complex to homogeneity from human cell lines. They determined that the IKK complex is composed of IKK-alpha, IKK-beta, and 2 other polypeptides of 50 and 52 kD which are differentially processed forms of a third subunit, IKK-gamma. Rothwarf et al. (1998) cloned IKK-gamma and identified it as the human homolog of mouse NEMO. The 419-amino acid IKK-gamma protein is composed of several potential coiled-coil motifs and a leucine zipper motif.

Using a yeast 2-hybrid assay to search for proteins that could interact with adenovirus protein E3-14.7K, known to prevent TNF-alpha (TNFA; 191160)-induced cytolysis, Li et al. (1999) cloned IKK-gamma, which they called FIP3 (14.7K-interacting protein). FIP3 contains leucine zippers and a zinc finger domain.

Using Western blot analysis and transfection studies in embryonic kidney cells, Puel et al. (2006) demonstrated that the IKBKG gene encodes a predominant 48-kD protein and an N-terminally truncated protein of 45 kD produced in smaller amounts and translated from methionine-38.


Gene Function

Yamaoka et al. (1998) found that NEMO interacted with IKK2 (IKK-beta; 603258), but not with IKK1 (IKK-alpha; 600664).

Rothwarf et al. (1998) determined that IKK-gamma forms dimers and trimers and interacts preferentially with IKK-beta. IKK-gamma was required for activation of the IKK complex. An IKK-gamma carboxy-terminal truncation mutant that binds IKK-beta blocked the activation of IKK and NF-kappa-B.

Li et al. (1999) determined that FIP3 inhibits both basal and induced transcriptional activity of NFKB and causes a late-appearing apoptosis with unique morphologic manifestations. E3-14.7K partially reversed apoptotic death induced by FIP3. FIP3 bound RIP (603453) and NIK (604655), which had been described as essential components of TNF-alpha-induced NFKB activation. FIP3 inhibited activation of NFKB induced by TNF-alpha, TNFR1 (191190), RIP, NIK, and IKK-beta, as well as basal levels of endogenous NFKB in 293 cells. FIP3 appeared both to activate a cell death pathway and to inhibit an NFKB-dependent survival mechanism.

Although in vitro studies had suggested that NEMO associates preferentially with IKK2 and not IKK1, Li et al. (1999) found that IKK1 immunoprecipitated with NEMO, suggesting that in vivo, IKK1 associates with NEMO through a component other than IKK2 in the IKK complex.

May et al. (2000) determined that an N-terminal alpha-helical region of NEMO associates with a region of IKKA and IKKB that they termed the NBD for 'NEMO-binding domain.' The NBD is a 6-amino acid C-terminal segment within the region denoted alpha-2 of IKKA and IKKB. A cell-permeable wildtype NBD peptide, but not a mutant, blocked the association of NEMO with the IKK complex and inhibited cytokine-induced NFKB activation and the expression of an NFKB-dependent gene, E-selectin (SELE; 131210), in endothelial cells in vitro. Wildtype NBD peptide also ameliorated acute inflammation, namely ear edema and peritonitis, in 2 experimental mouse models as effectively as dexamethasone. May et al. (2000) proposed that an NBD-type drug targeting the interaction of NEMO and the IKK complex might control inflammation yet maintain basal NFKB activity, thus avoiding toxic side effects.

Brummelkamp et al. (2003) designed a collection of RNA interference vectors to suppress 50 human deubiquitinating enzymes and used these vectors to identify deubiquitinating enzymes in cancer-relevant pathways. They demonstrated that inhibition of CYLD (605018) enhances activation of the transcription factor NF-kappa-B (164011). They showed that CYLD binds to the NEMO component of the IKK complex, and appears to regulate its activity through deubiquitination of TRAF2 (601895), as TRAF2 ubiquitination can be modulated by CYLD. Inhibition of CYLD increased resistance to apoptosis, suggesting a mechanism through which loss of CYLD contributes to oncogenesis. Brummelkamp et al. (2003) further demonstrated that this effect can be relieved by aspirin derivatives that inhibit NF-kappa-B activity.

Kovalenko et al. (2003) showed that CYLD interacts with NEMO and TRAF2. CYLD has a deubiquitinating activity that is directed toward the non-lys48-linked polyubiquitin chains and negatively modulates TRAF-mediated activation of IKK, strengthening the notion that ubiquitination is involved in IKK activation by TRAFs and suggesting that CYLD functions in this process.

Wu et al. (2006) demonstrated that NEMO associates with activated ATM (607585) after the induction of DNA double-strand breaks. ATM phosphorylates serine-85 of NEMO to promote its ubiquitin-dependent nuclear export. ATM is also exported in a NEMO-dependent manner to the cytoplasm, where it associates with and causes the activation of IKK in a manner dependent on another IKK regulator, a protein rich in glutamate, leucine, lysine, and serine (ELKS; 607127). Thus, Wu et al. (2006) concluded that regulated nuclear shuttling of NEMO links 2 signaling kinases, ATM and IKK, to activate NF-kappa-B by genotoxic signals.

Nenci et al. (2007) demonstrated that the transcription factor NFKB, a master regulator of proinflammatory responses, functions in gut epithelial cells to control epithelial integrity and the interaction between the mucosal immune system and gut microflora. Intestinal epithelial-specific inhibition of NFKB through conditional ablation of NEMO or both IKK1 and IKK2, IKK subunits essential for NFKB activation, spontaneously caused severe chronic intestinal inflammation in mice. NFKB deficiency led to apoptosis of colonic epithelial cells, impaired expression of antimicrobial peptides, and translocation of bacteria into the mucosa. Concurrently, this epithelial defect triggered a chronic inflammatory response in the colon, initially dominated by innate immune cells but later also involving T lymphocytes. Deficiency of the gene encoding the adaptor protein MyD88 (602170) prevented the development of intestinal inflammation, demonstrating that Toll-like receptor activation by intestinal bacteria is essential for disease pathogenesis in this mouse model. Furthermore, NEMO deficiency sensitized epithelial cells to TNF-induced apoptosis, whereas TNF receptor-1 (191190) inactivation inhibited intestinal inflammation, demonstrating that TNFR1 signaling is crucial for disease induction. Nenci et al. (2007) concluded that a primary NFKB signaling defect in intestinal epithelial cells disrupts immune homeostasis in the gastrointestinal tract, causing an inflammatory bowel disease-like phenotype. Their results further identified NFKB signaling in the gut epithelium as a critical regulator of epithelial integrity and intestinal immune homeostasis and have important implications for understanding the mechanisms controlling the pathogenesis of human inflammatory bowel disease.

Cytokine signaling is thought to require assembly of multicomponent signaling complexes at cytoplasmic segments of membrane-embedded receptors, in which receptor-proximal protein kinases are activated. Matsuzawa et al. (2008) reported that, upon ligation, CD40 (109535) formed a complex containing adaptor molecules TRAF2 and TRAF3 (601896), ubiquitin-conjugating enzyme UBC13 (UBE2N; 603679), cellular inhibitor of apoptosis protein-1 (CIAP1, or BIRC2; 601712) and -2 (CIAP2, or BIRC3; 601721), IKK-gamma, and MEKK1 (MAP3K1; 600982). TRAF2, UBC13, and IKK-gamma were required for complex assembly and activation of MEKK1 and MAP kinase cascades. However, the kinases were not activated unless the complex was translocated from the membrane to the cytosol upon CIAP1/CIAP2-induced degradation of TRAF3. Matsuzawa et al. (2008) proposed that this 2-stage signaling mechanism may apply to other innate immune receptors and may account for spatial and temporal separation of MAPK and IKK signaling.

By studying responses to the TLR4 (603030) ligand lipopolysaccharide (LPS) and to the bacterial chemoattractant fMLP in polymorphonuclear neutrophils (PMNs) from 1 patient with IRAK4 deficiency (607676) and 3 patients with NEMO deficiency causing X-linked hyper-IgM immunodeficiency with ectodermal dysplasia (300291), Singh et al. (2009) demonstrated reduced or absent superoxide production after impaired priming and activation of the oligomeric neutrophil NADPH oxidase (NOX; see 300481). The response was particularly weak or absent in IRAK4-deficient PMNs. NEMO-deficient PMNs had a phenotype intermediate between IRAK4-deficient PMNs and normal PMNs. Decreased LPS- and fMLP-induced phosphorylation of p38 (MAPK14; 600289) was observed in both deficiencies. Singh et al. (2009) proposed that decreased activation of NOX may contribute to increased risk of infection in patients with IRAK4 deficiency or NEMO deficiency.

One of the E3 ligases responsible for K63-linked NEMO polyubiquitination is TRAF6 (602355) which participates in several signaling pathways controlling immunity, osteoclastogenesis, skin development, and brain function. Gautheron et al. (2010) determined that a site at the N terminus of NEMO binds the coiled-coil domain of TRAF6 and apparently works in concert with NEMO's ubiquitin-binding domain to provide a dual mode of TRAF6 recognition. The E57K NEMO mutation, found in a mild form of incontinentia pigmenti (IP; 308300), resulted in impaired TRAF6 binding and IL1-beta (147720) signaling. In contrast, activation of NF-kappa-B (see 164011) by TNF-alpha (191160) was not affected. The authors concluded that the NEMO-TRAF6 interaction has physiologic relevance.

Gerlach et al. (2011) identified SHARPIN (611885) as a third component of the linear ubiquitin chain assembly complex (LUBAC), recruited to the CD40 and TNF receptor (see 191190) signaling complexes together with its other constituents, HOIL1 (RBCK1; 610924) and HOIP (RNF31; 612487). Mass spectrometry of TNFA signaling complexes revealed RIP1, also known as RIPK1 (603453), and NEMO to be linearly ubiquitinated. Tokunaga et al. (2011) also identified SHARPIN as a component of LUBAC, and showed that SHARPIN-containing complexes can linearly ubiquitinate NEMO and activate NK-kappa-B (see 164011). Ikeda et al. (2011) reported that SHARPIN functions as a novel component of LUBAC and that the absence of SHARPIN causes dysregulation of NF-kappa-B and apoptotic signaling pathways, explaining the severe phenotypes displayed by cpdm in Sharpin-deficient mice. Upon binding to the LUBAC subunit HOIP, SHARPIN stimulates the formation of linear ubiquitin chains in vitro and in vivo. Coexpression of SHARPIN and HOIP promotes linear ubiquitination of NEMO, an adaptor of the I-kappa-B kinases (IKKs; see 600664) and subsequent activation of NK-kappa-B signaling, whereas SHARPIN deficiency in mice causes an impaired activation of the IKK complex and NF-kappa-B in B cells, macrophages, and mouse embryonic fibroblasts. This effect is further enhanced upon concurrent downregulation of HOIL1L, another HOIP-binding component of LUBAC.

PER1 (602260) is a master regulator of circadian rhythm and functions in the nucleus to repress expression of central circadian clock genes. The periodicity of PER1 abundance, nuclear translocation, and transcriptional repression is regulated by PER1 phosphorylation, ubiquitination, and proteasomal degradation. Using Drosophila cells and various Per mutants, Chiu et al. (2011) found that Per was progressively phosphorylated by Nemo and doubletime (CSNK1E; 600863) during the circadian cycle. This progressive phosphorylation defined the length of the circadian cycle and the timing of proteasome-mediated Per degradation.


Gene Structure

The International Incontinentia Pigmenti Consortium (2000) sequenced the complete NEMO locus. NEMO is a 23-kb gene composed of 10 exons with 3 alternative noncoding first exons, 1a, 1b, and 1c (Jin and Jeang, 1999; Rothwarf et al., 1998; Li et al., 1999). The NEMO gene partially overlaps the G6PD (305900) gene, and it is transcribed in the opposite direction (Jin and Jeang, 1999).

Fusco et al. (2012) stated that the IKBKG gene has 9 coding exons (exons 2 through 10), 4 alternative noncoding first exons (exons 1A through 1D), and 2 promoters. Promoter A drives expression of IKBKG from exons 1D and 1A and is located within intron 2 of the more centromeric G6PD gene, which lies on the opposite strand. Promoter B is a bidirectional housekeeping promoter that drives expression of IKBKG from exons 1B and 1C, as well as G6PD. Fusco et al. (2012) determined that the region containing the G6PD gene and the 5-prime end of the IKBKG gene contains Alu elements.


Mapping

By sequence alignment, Jin and Jeang (1999) mapped the human NEMO gene to chromosome Xq28.

IKBKG Segmental Duplication

Aradhya et al. (2001) identified a truncated copy of NEMO (delta-NEMO), which maps 22 kb distal to NEMO and contains only exons 3 through 10. A sequence of 26 kb 3-prime of the NEMO coding sequence is also present in the same position relative to the delta-NEMO locus, bringing the total length of the duplication to 35.5 kb. The LAGE2 gene (CTAG1B; 300156) is also located within this duplicated region, and a similar but unique LAGE1 gene (CTAG2; 300396) is located just distal to the duplicated loci. Mapping and sequence information indicated that the duplicated regions are in opposite orientation. Analysis of the great apes suggested that the 35.5-kb NEMO/LAGE2 duplication occurred after divergence of the lineage leading to present day humans, chimpanzees, and gorillas, 10 to 15 million years ago. Despite this substantial evolutionary history, only 22 single-nucleotide differences exist between the 2 copies over the entire 35.5 kb, making the duplications more than 99% identical. This high sequence identity and the inverted orientations of the 2 copies, along with duplications of smaller internal sections within each copy, predispose this region to various genomic alterations. Aradhya et al. (2001) detected 4 rearrangements that involved NEMO, delta-NEMO, or LAGE1 and LAGE2. The authors hypothesized that the susceptibility of this complex genomic region to various types of pathogenic and polymorphic rearrangements may underlie the recurrent lethal deletion associated with incontinentia pigmenti (see MOLECULAR GENETICS).

Fusco et al. (2012) stated that the IKBKG gene and the more telomeric CTAG1A gene (300657) were involved in an inverted 35.7-kb segmental duplication on chromosome Xq28 that produced the CTAG1B gene and an IKBKG pseudogene, IKBKGP1, copied from IKBKG exons 3 through 10. Fusco et al. (2012) found that the duplicated regions and the regions between and near them contain a high number of short and long interspersed elements and long terminal repeats.


Molecular Genetics

X-Linked Dominant Incontinentia Pigmenti

The International Incontinentia Pigmenti Consortium (2000) demonstrated that heterozygous mutations in the IKBKG gene cause X-linked dominant incontinentia pigmenti (IP; 308300). The most common mutation in IP is a genomic rearrangement resulting in deletion of part of the NEMO gene (300248.0001). An 870-bp region of identity corresponding to an MER67B repeat exists both in intron 3 and 3-prime to exon 10; recombinations between the regions of identity delete exons 4 though 10 of NEMO. The International Incontinentia Pigmenti Consortium (2000) stated that this rearrangement, which occurs during paternal meiosis, causes 80% of new mutations. It was demonstrated that this mutation results in a lack of NF-kappa-B (see 164011) activation resulting in extreme susceptibility to apoptosis, leading to embryonic death in males and explaining the extremely skewed X inactivation seen in females with IP.

Aradhya et al. (2001) reported 2 families with IP with duplications in a 7-cytosine tract in exon 10 of the IKBKG gene (300248.0008 and 300248.0012). Aradhya et al. (2001) found that the duplication mutations of the IKBKG gene allowed males to survive and affected females to have random or slightly skewed X inactivation, whereas virtually all other mutations eliminate the production of NEMO, causing lethality in males and the typical skewing of X inactivation in females.

The International IP Consortium (2001) investigated 4 male patients with clinical hallmarks of IP. All 4 were found to carry the deletion in the NEMO gene normally associated with male lethality in utero. Survival in 1 patient was explained by a 47,XXY karyotype and skewed X inactivation. Three other patients possessed a normal 46,XY karyotype. It was demonstrated that these patients had both wildtype and deleted copies of the NEMO gene, indicating mosaicism for the common mutation. Therefore, the repeat-mediated rearrangement leading to the common deletion does not require meiotic division. Thus there are 3 mechanisms for survival of males carrying a NEMO mutation: hypomorphic alleles, a 47,XXY karyotype, and somatic mosaicism.

In an examination of families transmitting the recurrent deletion of exons 4 through 10 of the NEMO gene (300248.0001), Aradhya et al. (2001) revealed that the rearrangement occurred in the paternal germline in 70% of cases, indicating that it arises predominantly by intrachromosomal misalignment during meiosis. Expression analysis of human and mouse NEMO/Nemo showed that the gene becomes active early during embryogenesis and is expressed ubiquitously. The authors proposed a model to explain the pathophysiology of IP in terms of disruption of the NF-kappa-B signaling pathway.

Bardaro et al. (2003) presented instances in which the exon 4 to 10 delta-NEMO pseudogene deletion occurred in unaffected parents of 2 females with clinically characteristic IP, confusing the genetic analysis. They described a new PCR-based test that permitted unambiguous molecular diagnosis and proper familial genetic counseling for IP.

Fusco et al. (2004) identified NEMO mutations in 83 of 122 IP patients and measured the effects of NEMO point mutations on NFKB signaling in Nemo-deficient murine pre-B cells. A mutation in the N-terminal domain, required for IKK assembly, reduced but did not abolish NFKB activation following LPS stimulation. Mutations that disrupt the C-terminal domain (required for the recruitment of upstream factors) showed lower or no NFKB activation. There was no phenotype-genotype correlation observed; 64% of patients exhibited an extremely skewed X-inactivation pattern (80:20). Fusco et al. (2004) concluded that IP pathogenesis may depend on a combination of X inactivation and protein domain that recruits upstream factors and activates NFKB.

In a female infant who presented at birth with features of IP, Martinez-Pomar et al. (2005) identified a heterozygous mutation in the IKBKG gene (300248.0017). She also developed transient immunodeficiency that resolved spontaneously in the first years of life. The X-inactivation status of peripheral blood cells from the patient was evaluated at 24, 30, 38, and 48 months of age and was found to have progressed from random at 24 and 30 months to skewed at 38 and 48 months of age, at which point her immunodeficiency had disappeared. Martinez-Pomar et al. (2005) stated that this was the first time that selection against the mutated X chromosome in X-linked disease had been documented in vivo.

Ectodermal Dysplasia and Immunodeficiency 1

In affected males from 4 unrelated families with X-linked recessive hypohidrotic ectodermal dysplasia and immunodeficiency-1 (EDAID1; 300291), Zonana et al. (2000) identified hemizygous mutations in the IKBKG gene (300248.0007-300248.0010). All mutations occurred in exon 10 of the gene, which encodes the C-terminal zinc finger domain, and were predicted to result in a loss of NFKB activation. Since heterozygous mutations in this gene cause IP in females and are usually lethal in males, Zonana et al. (2000) hypothesized that the mutations identified in males with EDAID1 are hypomorphic; functional studies of the variants were not performed. Affected males showed dysgammaglobulinemia and, despite therapy, had significant morbidity and mortality from recurrent infections.

Doffinger et al. (2001) identified 5 novel mutations (see, e.g., 300248.0020) in IKBKG in affected males from 5 kindreds with anhidrotic ectodermal dysplasia and immune deficiency. Doffinger et al. (2001) also showed that the ectodysplasin receptor (EDAR; 604095) triggers NF-kappa-B through the NEMO protein, indicating that anhidrotic ectodermal dysplasia results from impaired NF-kappa-B signaling.

In 2 boys with EDAID1 associated with increased IgM, Jain et al. (2001) identified hemizygous missense mutations in the NEMO gene (C417R, 300248.0009 and D406V, 300248.0011). Both mutations occurred in the putative zinc finger domain, a potentially shared intracellular signaling component for EDAR and CD40L (300386). Although stimulation of patient monocytes with TNF and LPS resulted in responses similar to those of normal controls, stimulation with CD40L failed to induce phosphorylation of I-kappa-B-alpha (IKBA; 164008) and failed to upregulate expression of CD54 (ICAM1; 147840). CD40L stimulation also failed to induce class switching in patient B cells and failed to result in IL12 and TNF production. T-cell function and maturation appeared to be normal in the patients, and they had no history of opportunistic infections suggestive of T-cell dysfunction. Jain et al. (2001) concluded that NEMO has a regulatory function in NFKB activation and B-cell Ig class switching.

Jain et al. (2004) showed that 3 patients with EDAID1 due to missense mutations affecting cys417 of NEMO had markedly diminished levels of serum IgG and IgE. Activation of patient B cells with soluble CD40L and IL4 (147780) induced normal levels of I-epsilon and C-epsilon germline transcripts and expression of AID (AICDA; 605257), suggesting that expression of additional genes, possibly regulated by CD40-mediated REL (164910) activation, are necessary for Ig class switch recombination. Microarray analysis of EDAID1 B cells showed impairments in the expression of RAD50 (604040), LYL1 (151440), LIG4 (601837), and other factors relating to nonhomologous recombination but not previously linked to B-cell differentiation. Jain et al. (2004) proposed that IL4 augments some (e.g., RELA; 164014) but not all NEMO-dependent NFKB signaling.

In 3 unrelated boys with EDAID1, Orange et al. (2002) identified hemizygous mutations in the IKBKG gene. Two mutations occurred in the zinc finger domain (C417R, 300248.0009 and Q403X, 300248.0015), and 1 occurred in the first coiled-coil domain (L153R; 300248.0014). Patient cells showed defective CD40 signaling, as well as impaired natural killer (NK) cell activity, in spite of normal levels of peripheral blood NK numbers. Addition of IL2 (147680) induced NFKB activation and partially reversed the NK activity defect in vitro. Intravenous administration of IL2 to one of the patients, who had the L153R mutation and persistent cytomegalovirus infection, resulted in enhanced NK cell activity. Orange et al. (2002) proposed that NEMO is important for NFKB activation as well as NK cell cytotoxicity, and that IL2 may benefit patients with NEMO mutations.

In a family with ectodermal dysplasia and immunodeficiency previously reported by Lie et al. (1978), Orstavik et al. (2006) identified a splice site mutation in the NEMO gene (300248.0016). The family had 3 stillborn males, 3 affected males who were small for gestational age and died within 8 months, and 1 male who died at age 5 years. The 5-year-old boy had cone-shaped teeth, oligodontia, serious bacterial infections, and inflammatory bowel disease. Isolated subtle tooth anomalies were found in 3 female carriers examined, of whom 2 had random X inactivation and 1 had extreme skewing. Orstavik et al. (2006) stated that this was the first report of random X inactivation in carriers of EDAID.

Takada et al. (2010) analyzed the NEMO gene in a family in which a boy with EDAID1 died at 9 years of age from gastrointestinal bleeding, and his 6-year-old sister and mother were found to have incontinentia pigmenti and entero-Behcet disease (see 109650); all 3 carried the D406V (300248.0011) mutation. Takada et al. (2010) noted that these patients exhibited several unusual features, including the occurrence of incontinentia pigmenti and EDAID1 in a family; the presence of hypopigmented skin lesions in early infancy in the mother and daughter, since they are usually observed in the second decade to adulthood; and a lack of extremely skewed X-chromosome inactivation in the mother and daughter.

In a boy with EDAID1, Roberts et al. (2010) identified a hemizygous 2-bp deletion (c.1182_1183delTT; 300248.0027) in the IKBKG gene, predicted to result in a frameshift and premature termination. His mother, who carried the mutation in heterozygous state, showed signs of IP since childhood. Functional studies of the variant were not performed.

In a male infant with lethal EDAID1, Johnston et al. (2016) identified a hemizygous splice site mutation in the IKBKG gene (300248.0028). Western blot analysis of patient cells showed reduced size of the IKBKG protein, consistent with a frameshift and premature termination.

In a boy (patient 1) with EDAID1, Heller et al. (2020) identified a hemizygous splice site mutation (300248.0031) in the IKBKG gene, resulting in a frameshift and premature termination with loss of the zinc finger domain. Patient cells showed decreased IKBKG levels compared to controls; in vitro studies showed impaired degradation of IKBA and impaired IL6 production upon stimulation with IL1B and TNFA. His mother and sister, who were heterozygous for the mutation, had incontinentia pigmenti.

Immunodeficiency 33

Two unrelated male patients with immunodeficiency-33 (IMD33; 300636) without ectodermal dysplasia were described by Niehues et al. (2004) and Orange et al. (2004). The patient reported by Orange et al. (2004) was found to have a hemizygous splice mutation that resulted in the skipping of exon 9 of the NEMO gene, affecting the LZ domain (300248.0018) but leaving the zinc finger domain intact. In the patient reported by Niehues et al. (2004), Puel et al. (2006) identified a hemizygous 1-bp insertion in exon 2 of the NEMO gene (300248.0019). Puel et al. (2006) showed that a Kozakian methionine codon located immediately downstream from the insertion allowed the reinitiation of translation. The residual production of an NH2-truncated NEMO protein was sufficient for normal fetal development and for the subsequent normal development of skin appendages, but was insufficient for the development of protective immune responses.

In 2 unrelated boys with IMD33, Ku et al. (2005) identified hemizygous mutations in the IKBKG gene: an 18-bp deletion (300248.0025) and a missense mutation (L80P, 300248.0026). The mutations occurred in the coiled-coil domains of the protein. Western blot analysis of patient cells showed the presence of the IKBKG protein, but fibroblasts had poor IL6 production in response to TNFA and IL1B compared to controls. The findings were consistent with a hypomorphic allele. The patients had hypodontia and conical teeth, but no other features of ectodermal dysplasia. One carrier mother also had conical teeth.

In a Belgian boy with IMD33, Ku et al. (2007) identified a hemizygous splice site mutation in the IKBKG gene (300248.0023). Western blot analysis showed decreased levels of the protein compared to controls, consistent with partial IKBKG deficiency. The mother was heterozygous for this mutation.

In affected males from 3 unrelated kindreds with IMD33 manifest as susceptibility to mycobacterial infection, Filipe-Santos et al. (2006) identified 2 hemizygous missense mutations in the NEMO gene (E315A, 300248.0021 and R319Q, 300248.0022), both of which occurred in the leucine zipper (LZ) domain and were predicted to disrupt a salt bridge. Western blot and flow cytometric analyses showed normal expression of the mutant NEMO proteins. Patient mononuclear cells responded normally to most stimuli, but IFNG (147570) and IL12 (see 161561) production in response to PHA mitogen was impaired due to defective CD40 (109535) signaling in monocytes and dendritic cells. Filipe-Santos et al. (2006) concluded that mutations in NEMO that disrupt the leucine zipper domain provide a genetic etiology to X-linked recessive immunodeficiency with a particular susceptibility to mycobacterial disease.

In a 2-year-old boy with IMD33, Frans et al. (2017) identified a hemizygous missense mutation in the IKBKG gene (E57K; 300248.0029). The mutation affected the N-terminal domain of the protein. His mother, who also carried the mutation, had a history of recurrent sinorespiratory infections, but no signs of IP. The variant was present at a low frequency (0.001) in the ExAC database. Patient peripheral blood cells showed mildly decreased IL6 production after stimulation with IL1B compared to controls. However, NEMO expression in patient fibroblasts was normal, and IKBA was degraded normally upon stimulation with IL1B or TNFA, suggesting a specific effect of the mutation. In vitro functional expression studies in IKBKG-null cells transfected with the mutation showed mildly impaired IL6 production after stimulation with TNFA or IL1B compared to controls. Frans et al. (2017) noted that mutations affecting the N terminus of NEMO tend to lead to decreased production of immunoglobulins; the authors postulated a hypomorphic effect of this variant.

In 4 adult males from 3 unrelated families with IMD33 manifest as disseminated mycobacterial infections, Hsu et al. (2018) identified hemizygous splice site mutations in the 5-prime untranslated region of the IKBKG gene. Three patients from 2 families (families A and B) carried a c.1-16G-C transversion (300248.0030) at the last base of the first untranslated exon. The patient from family C carried a hemizygous c.1-16+G-T in the adjacent intron. Analysis of cells from the patient with the c.1-16G-C mutation showed a splicing abnormality, resulting in a 110-bp deletion at the 3-prime end of exon 1. This molecular defect resulted in decreased transcript and protein levels compared to controls (about 30%). Cells derived from the patients in families A and B failed to upregulate cytokines in response to certain TLR agonists, suggesting that this IKBKG mutation is hypomorphic.

In 3 boys from 2 unrelated families of European and Japanese descent with fatal IMD33, Boisson et al. (2019) identified a deep intronic mutation in the IKBKG gene (IVS4+866C-T; 300248.0024) that created a new splice donor site and resulted in a 44-nucleotide pseudoexon that produced a frameshift. The boy in the European family inherited the mutation from his mother, who had mild incontinentia pigmenti. The mutation in the Japanese boy occurred de novo. The variant was not found in the 1000 Genomes Project or gnomAD databases.

In a boy (patient 1) with IMD33, Abbott et al. (2014) identified a hemizygous missense mutation in the first coiled-coil domain of NEMO (D113N; 300248.0032). The patient had previously been reported in detail by Salt et al. (2008); his unaffected mother also carried the mutation.

Heller et al. (2020) identified hemizygosity for the D113N mutation in a boy with IMD33. His mother and grandmother, who were presumably unaffected, were heterozygous for the mutation. However, the proband's 40-year-old male cousin, who did not have recurrent infections and had normal response to polysaccharide antibodies, was hemizygous for D113N. Heller et al. (2020) noted that the allele frequency for this variant is rather high (0.009572), and that some suggest it may be a polymorphism (see Fusco et al., 2004).

X-linked Systemic Autoinflammatory Disease

In 3 unrelated boys (P1-P3) and a girl (P4) with X-linked systemic autoinflammatory disease (SAIDX; 301081), de Jesus et al. (2020) identified de novo hemizygous or heterozygous mutations in the IKBKG, all of which were predicted to disrupt splicing and result in the deletion of exon 5 (300248.0033-300248.0036). Functional studies of the variants were not performed. The patients were ascertained from a cohort of patients with autoinflammatory disease and a moderately elevated type I interferon signature.

In 3 unrelated boys with SAIDX, Lee et al. (2022) identified de novo hemizygous mutations in the IKBKG gene (300248.0033-300248.0035). The mutations, which were confirmed by Sanger sequencing, all led to overexpression of a NEMO protein lacking the domain encoded by exon 5 (NEMOdelEx5). In vitro studies showed that this splice isoform failed to associate with TANK binding kinase-1 (TBK1; 604834). Dermal fibroblasts from affected patients activated NFKB in response to TNF (191160), but showed impaired NFKB activation to TLR3 (603029) or RIGI-like receptor (RLR) stimulation with poly(I:C) positively correlated with levels of the NEMOdelEx5 splice isoform. By contrast, T cells, monocytes, and macrophages that expressed the splice site variant exhibited increased NFKB activation and IFN production. Blood cells from these patients expressed a strong IFN and NFKB transcriptional signature. Immune cells and TNF-stimulated dermal fibroblasts upregulated the inducible IKK protein (IKKi; 605048) that was stabilized by the splice variant, promoting type I IFN induction and antiviral responses. These data showed that IKBKG mutations that lead to alternative splicing of skipping exon 5 cause an autoinflammatory disorder, which the authors termed 'NEMO deleted exon 5 autoinflammatory syndrome (NDAS),' noting that it is distinct from the immunodeficiency syndrome resulting from loss-of-function IKBKG mutations.


Genotype/Phenotype Correlations

In general, males with NKBKG mutations affecting the C-terminal zinc finger domain have a more severe clinical course with immunodeficiency and ectodermal dysplasia, whereas patients with mutations affecting the leucine zipper domain or the more N-terminal coiled-coil domains have a less severe clinical course and do not show features of ectodermal dysplasia, although isolated hypotonia and/or conical teeth may be present (Orange et al., 2004, Heller et al., 2020).


Animal Model

Rudolph et al. (2000) generated NEMO/IKK-gamma-deficient mice by gene targeting. Mutant embryos died at embryonic day 12.5-13 from severe liver damage due to apoptosis. NEMO/IKK-gamma-deficient primary murine embryonic fibroblasts (MEFs) lacked detectable NF-kappa-B DNA-binding activity in response to TNF-alpha, IL1 (see 147760), bacterial lipopolysaccharide, and poly(IC) and did not show stimulus-dependent I-kappa-B kinase activity, which correlated with a lack of phosphorylation and degradation of I-kappa-B-alpha (164008). Consistent with these data, mutant MEFs showed increased sensitivity to TNF-alpha-induced apoptosis. Rudolph et al. (2000) concluded that their data provided in vivo evidence that NEMO is the first essential noncatalytic component of the IKK complex.

Makris et al. (2000) showed that female mice heterozygous for Ikbkg deficiency develop a unique dermatopathy characterized by keratinocyte hyperproliferation, skin inflammation, hyperkeratosis, and increased apoptosis. Although Ikbkg +/- females eventually recovered, IKBKG -/- males died in utero. The symptoms and inheritance pattern were very similar to those of IP. Indeed, biopsies and cells from IP patients exhibited defective IKBKG expression but normal expression of IKK catalytic subunits. The authors proposed that the IKBKG-deficient cells trigger an inflammatory reaction that eventually leads to their death.

Schmidt-Supprian et al. (2000) found that disruption of the mouse Ikbkg gene produces male embryonic lethality, completely blocks NF-kappa-B activation by proinflammatory cytokines, and interferes with the generation and/or persistence of lymphocytes. Heterozygous female mice developed patchy skin lesions with massive granulocyte infiltration and hyperproliferation and increased apoptosis of keratinocytes. Diseased animals presented severe growth retardation and early mortality. Surviving mice recovered almost completely, presumably through clearing the skin of Ikbkg-deficient keratinocytes. The authors stated that male lethality and strikingly similar skin lesions in heterozygous females are hallmarks of the human genetic disorder IP.

Using a chemical mutagenesis screen in mice, Siggs et al. (2010) identified an X-linked recessive phenotype, termed 'pan resistance-2' (panr2), marked by diminished secretion of Tnf and other Nfkb-dependent cytokines by macrophages in response to ligands for Tlr3 (603029), Tlr4, Tlr7 (300365), and Tlr9 (605474), as well as for the TLR heterodimers Tlr1 (601194)/Tlr2 (603028) and Tlr2/Tlr6 (605403). They identified the panr2 mutation as a 473T-C transition in exon 4 of the Ikbkg gene, which results in a leu153-to-pro (L153P) substitution within the first coiled-coil domain of the protein. Panr2 mutant males were viable but azoospermic, and they usually lacked inguinal lymph nodes. Other tissues were normal, although serum lacked all Ig isotypes. Panr2 mutant mice had an impairment of MAPK phosphorylation and Nfkb p65 translocation, but not of Ikba degradation. Siggs et al. (2010) concluded that IKBKG-regulated pathways are of immunologic importance beyond IKBA degradation and may account for other immunodeficiencies.


ALLELIC VARIANTS 36 Selected Examples):

.0001   INCONTINENTIA PIGMENTI

IKBKG, EX4-10DEL
ClinVar: RCV000012200

In male and female fetuses with incontinentia pigmenti (IP; 308300), The International Incontinentia Pigmenti Consortium (2000) found a genomic rearrangement resulting in deletion of part of the NEMO gene. An 870-bp region of identity corresponding to an MER67B repeat exists in the NEMO gene both in intron 3 and 3-prime to exon 10; recombinations between the regions of identity delete exons 4 though 10 of NEMO. The International Incontinentia Pigmenti Consortium (2000) stated that this rearrangement accounts for 80% of new IP mutations. This rearrangement would result in a truncated molecule carrying the 133 N-terminal amino acids of NEMO plus at least 26 novel amino acids. It was predicted that this molecule would contain part of the first coiled-coil domain, and may still interact with IKK2 (603258), but would be unlikely to respond to upstream signals. Failure of I-kappa-B-alpha (164008) degradation and lack of NF-kappa-B (see 164011) activation were demonstrated in IP embryonic fibroblasts.

Aradhya et al. (2001) identified 277 mutations in 357 unrelated IP patients. The recurrent deletion of exons 4 to 10 accounted for 90% of the identified mutations. The remaining mutations were small duplications, substitutions, and deletions.


.0002   ECTODERMAL DYSPLASIA AND IMMUNODEFICIENCY 1, MALE-RESTRICTED

INCONTINENTIA PIGMENTI, INCLUDED
IKBKG, TER420TRP
SNP: rs137853321, ClinVar: RCV000012203, RCV000170521

In a male infant (patient IP85) with ectodermal dysplasia and immunodeficiency-1 (EDAID1; 300291), The International Incontinentia Pigmenti Consortium (2000) identified a hemizygous c.1259A-G transition in exon 10 of the IKBKG gene, resulting in a stop420-to-trp (X420W) substitution. This change results in the addition of 27 residues to the C terminus of the mature protein. The mutation was also found in heterozygous state in his mother, who had incontinentia pigmenti (IP; 308300). In addition to recurrent infections, the boy also had osteopetrosis and lymphedema. He died at age 2.5 years from a tuberculosis infection.

Doffinger et al. (2001) studied patient IP85 reported by The International Incontinentia Pigmenti Consortium (2000) as well as another boy, of French descent, with the hemizygous X420W mutation and classified both as having anhidrotic ectodermal dysplasia with immunodeficiency, osteopetrosis, and lymphedema (OLEDAID; see 300291), which is within the phenotypic spectrum of EDAID1. Infections in the French boy, who died at the age of 1.5 years, included atypical mycobacteria and pneumococcus; he also had osteopetrosis and lymphedema, indicating that these additional features are associated with this specific mutation. His mother, who carried the mutation in heterozygous state, had mild IP. Detailed in vitro functional expression studies showed that the X420W mutation resulted in a 50 to 60% reduction of NF-kappa-B activation. Cells from both male patients with this mutation showed impaired cellular responses to LPS, IL1B, and TNFA. The findings indicated that the X420W mutation impairs, but does not abolish, NFKB activation, consistent with a hypomorphic allele and postnatal survival of the boys.


.0003   INCONTINENTIA PIGMENTI

IKBKG, 10-BP INS, NT127
SNP: rs2070949441, ClinVar: RCV000012204

In an incontinentia pigmenti (IP; 308300) proband and her affected mother, but not in unaffected sibs, The International Incontinentia Pigmenti Consortium (2000) identified a 10-bp insertion in exon 2 of the IKBKG gene, between nucleotides 127 and 128. The insertion, an exact duplication of the previous 10 nucleotides, shifts the reading frame of the protein to add 8 amino acids after residue 43 and then truncates the protein at the next in-frame stop codon.


.0004   INCONTINENTIA PIGMENTI

IKBKG, 1-BP INS, 1110C
SNP: rs1569556615, ClinVar: RCV000012201, RCV003162240

In a family with incontinentia pigmenti (IP; 308300), The International Incontinentia Pigmenti Consortium (2000) identified a single C insertion between nucleotides 1110 and 1111 that segregated with the disease. This frameshift mutation would putatively append 23 new amino acids onto proline residue 370 in the translated protein.


.0005   INCONTINENTIA PIGMENTI

IKBKG, MET407VAL
SNP: rs137853322, gnomAD: rs137853322, ClinVar: RCV000012202, RCV001582474

In a female with incontinentia pigmenti (IP; 308300), The International Incontinentia Pigmenti Consortium (2000) identified an A-to-G transition in exon 10 of the IKBKG gene that changed methionine to valine at codon 407. SSCP analysis showed that this relatively conservative change segregated with the disease through a 3-generation pedigree.


.0006   INCONTINENTIA PIGMENTI

IKBKG, PRO62TER
SNP: rs137853323, ClinVar: RCV000012205, RCV000256164

In a family with incontinentia pigmenti (IP; 308300), The International Incontinentia Pigmenti Consortium (2000) identified a C-to-T transition at nucleotide 184 of the IKBKG gene resulting in a proline-to-stop mutation in exon 2.


.0007   ECTODERMAL DYSPLASIA AND IMMUNODEFICIENCY 1

IKBKG, GLU391TER
SNP: rs137853324, ClinVar: RCV000012206, RCV000760425

In 3 affected boys from a family (family 2) with hypohidrotic ectodermal dysplasia and immunodeficiency (EDAID1; 300291), Zonana et al. (2000) identified a hemizygous c.1171G-T transversion in exon 10 of the IKBKG gene, resulting in a glu391-to-ter (E391X) substitution within the C-terminal end of the protein. The truncated protein was predicted to lack the putative zinc finger domain. Western blot analysis of patient cells showed that a protein was expressed. Additional functional studies were not performed, but the authors postulated that the mutation preserves some function. Two female carriers in the family had conical teeth, some variable hyperpigmented skin lesions, and increased IgA, but no immune defects.


.0008   ECTODERMAL DYSPLASIA AND IMMUNODEFICIENCY 1

INCONTINENTIA PIGMENTI, INCLUDED
IKBKG, 1-BP DUP, 1167C
SNP: rs782178147, gnomAD: rs782178147, ClinVar: RCV000012209, RCV000413717, RCV001172473, RCV002506000, RCV003992285

In 2 brothers (family 4) with lethal hypohidrotic ectodermal dysplasia with immune deficiency-1 (EDAID1; 300291), Zonana et al. (2000) identified a hemizygous 1-bp duplication (c.1167_1168dupC) in exon 10 of the IKBKG gene, predicted to result in a frameshift, the addition of novel amino acids at codons 390-393, and premature termination at codon 394. The duplication resulted from an insertion of a cytosine within a wildtype run of 7 cytosines in exon 10. The mutation was predicted to delete the putative zinc finger domain. The patients also had osteopetrosis and died of mycobacterial infection.

Kosaki et al. (2001) described an unusual family in which a girl with this heterozygous mutation had skin abnormalities with streaky hyperpigmented macules, features of ectodermal dysplasia, including decreased sweating, thin hair, and hypodontia, and onset of recurrent infections around 4 years of age. She later developed pulmonary and renovascular hypertension, and died at age 11 years after cardiac catheterization. IgD and IgE were increased. X-inactivation studies of peripheral blood leukocytes showed a random pattern. The mother had hyperpigmented macules in a streaky configuration on the trunk and limbs at several weeks of age, but never developed immunodeficiency; DNA from the mother was not studied. The family had previously been reported by Akiyama et al. (1994) as having 'linear and whorled nevoid hypermelanosis (see 614323). Zonana and Ferguson (2001) noted that the findings of random X inactivation in the girl reported by Kosaki et al. (2001) was unusual, and postulated that other molecular mechanisms may have been at play, including the possibility of skewed X inactivation in different leukocyte subsets. These authors emphasized that this phenotype in females is a very rare occurrence, and that generally, only males are affected.

Aradhya et al. (2001) identified the same frameshift mutation, referred to as a 1-bp duplication in the 7-cytosine tract (1161_1162dupC) of the IKBKG gene, in a male patient (family XL344) with EDAID1. Female members of the family, who were heterozygous for the mutation, had typical signs of incontinentia pigmenti (IP; 308300). The affected male exhibited skin pigmentation, dental problems, and immune dysfunction. He suffered multiple episodes of infection, including meningitis and pneumonia, due to poor lymphocyte function and remarkably low levels of circulating IgG. He also exhibited heat intolerance with hyperthermia, anhidrosis, eczema, and fine sparse hair, which led to a diagnosis of ectodermal dysplasia. At the age of 3 years he was receiving routine supplements of IgG to prevent recurrent infections. He showed hepatosplenomegaly and had contracted Mycobacterium avium intracellulare, an infection common among patients with AIDS. Aradhya et al. (2001) identified a different mutation involving the same C(7) tract in another family; see 300248.0012.


.0009   ECTODERMAL DYSPLASIA AND IMMUNODEFICIENCY 1

IKBKG, CYS417ARG
SNP: rs137853325, ClinVar: RCV000012207

In 2 brothers (family 1) with hypohidrotic ectodermal dysplasia and immunodeficiency-1 (EDAID1; 300291), Zonana et al. (2000) identified a hemizygous c.1249T-C transition in exon 10 of the IKBKG gene, resulting in a cys417-to-arg (C417R) substitution in the putative zinc finger domain. The mother, who carried the mutation in heterozygous state, was unaffected.

Orange et al. (2002) identified the C417R mutation in a 16-year-old boy (patient 3) with EDAID1 with recurrent sinopulmonary infections and bacteremia. Tetanus vaccination failed to confer detectable serologic or delayed hypersensitivity responses. The patient also had reduced natural killer cell function.

In a male patient with EDAID1, Jain et al. (2001) identified hemizygosity for the C417R mutation, which occurred in the putative zinc finger domain.


.0010   ECTODERMAL DYSPLASIA AND IMMUNODEFICIENCY 1

IKBKG, CYS417PHE
SNP: rs137853326, ClinVar: RCV000012211, RCV000059068

In a boy (family 3) with hypohidrotic ectodermal dysplasia and immunodeficiency-1 (EDAID1; 300291), Zonana et al. (2000) identified a hemizygous c.1250G-T transversion in exon 10 of the IKBKG gene, resulting in a cys417-to-phe (C417F) substitution in the zinc finger domain. The patient's mother, who carried the mutation, had mild tooth anomalies and a 'large patch of skin with neither hair nor sweating response.'


.0011   ECTODERMAL DYSPLASIA AND IMMUNODEFICIENCY 1, MALE-RESTRICTED

INCONTINENTIA PIGMENTI, INCLUDED
IKBKG, ASP406VAL
SNP: rs137853327, ClinVar: RCV000024285, RCV001172474

In a male patient with ectodermal dysplasia and immunodeficiency-1 (EDAID1; 300291), Jain et al. (2001) identified a hemizygous c.1217A-T transversion in exon 10 of the IKBKG gene, resulting in an asp406-to-val (D406V) substitution. The mutation occurred in the putative zinc finger domain.

In a family in which a boy with ectodermal dysplasia with immune deficiency-1 died at 9 years of age from gastrointestinal bleeding and his sister and mother were found to have incontinentia pigmenti (IP; 308300) and to fulfill the criteria for entero-Behcet disease (see 109650), Takada et al. (2010) identified the D406V mutation in the NEMO gene. The 6-year-old sister developed oral and perianal ulcers at 5.5 years of age and had hypopigmented lesions without atrophy in the abdominal area and extremities that had been present since early infancy. Endoscopy showed multiple ulcerative lesions in the colon, which had no reactive changes at the margins and showed chronic active inflammation on histology. The authors stated that the patient's findings fulfilled diagnostic criteria for the enteric form of Behcet disease. The 42-year-old mother developed oral and perianal ulcers at 8 years of age and was diagnosed with Behcet disease at age 12. She also had hypopigmented skin lesions in the abdominal area and extremities that had been observed since early infancy. Skewed X-chromosome inactivation was not seen in the peripheral blood mononuclear cells or in oral or intestinal mucosa of these patients.


.0012   INCONTINENTIA PIGMENTI

IKBKG, 13-BP DUP, NT1166
SNP: rs2071167272, ClinVar: RCV000012213

In affected women from a family (family XL320) with incontinentia pigmenti (IP; 308300), Aradhya et al. (2001) identified a heterozygous 13-bp duplication (c.1166_1178dup) at the end of a cytosine tract (a C(7) tract) in exon 10 of the IKBKG gene. This duplication was predicted to cause a frameshift after amino acid P393 and protein truncation after addition of 4 novel amino acids, resulting in absence of the zinc finger domain. The family had previously been studied by Roberts et al. (1998), who reported that an affected male carrying the hemizygous mutation was carried to term, but died from severe hemorrhage 24 hours after birth. In a subsequent pregnancy, the mother experienced severe bleeding during an elective abortion, suggesting that the mutation in this family disrupted hemostasis.


.0013   ECTODERMAL DYSPLASIA AND IMMUNE DEFICIENCY 1

IKBKG, 4.4-KB DUP
ClinVar: RCV000012214

In a 2-year-old boy with hypohidrotic ectodermal dysplasia and immune deficiency-1 (EDAID1; 300291), Nishikomori et al. (2004) identified a hemizygous 4.4-kb duplication of genomic sequence of a segment extending from intron 3 to exon 6 in the IKBKG gene, resulting in reduced expression of the protein. The patient had atypical features of very few naive T cells and defective mitogen-induced proliferation of peripheral blood mononuclear cells. Specific cell lineages (monocytes and neutrophils) expressed reduced levels of IKBKG, but 2 populations of T, B, and NK cells were detected, 1 with normal and 1 with reduced expression of IKBKG. This was thought most likely to be due to postzygotic reversion. The patient had hypohidrosis, coarse hair, delayed eruption of teeth, and transient lower extremity lymphedema, but did not develop the skin lesions typical of incontinentia pigmenti. At age 3 months, he was admitted to hospital because of interstitial pneumonitis. Thereafter he had frequent bacterial infections and febrile episodes due to skin abscess, pneumonia, pneumococcal sepsis, otitis media, and sepsis. He also suffered from intractable diarrhea that resulted in failure to thrive. One oral live polio vaccination resulted in no detectable anti-poliovirus titers.


.0014   ECTODERMAL DYSPLASIA AND IMMUNODEFICIENCY 1

IKBKG, LEU153ARG
SNP: rs137853328, ClinVar: RCV000012215

In a patient (patient 1) under 24 months of age who had ectodermal dysplasia with immunodeficiency-1 (EDAID1; 300291), Orange et al. (2002) identified a hemizygous c.458T-G transversion in exon 4 of the IKBKG gene, resulting in a leu153-to-arg (L153R) substitution within the first coiled-coil domain of the protein. In vitro functional expression studies showed impaired CD40-mediated activation of NFKB compared to controls. The patient had bacterial and viral sepsis, as well as Streptococcal bovis meningitis and cytomegalovirus colitis. He was noted to have absent sweat glands on skin biopsy in the 'Methods' section, but additional features of ectodermal dysplasia were not noted. Orange et al. (2004) also reported this patient and noted that he had 'characteristics' of ectodermal dysplasia; his mother carried the mutation and had oligodontia, some alopecia, and skin hyperpigmentation, consistent with mild features of incontinentia pigmenti.


.0015   ECTODERMAL DYSPLASIA AND IMMUNODEFICIENCY 1

IKBKG, GLN403TER
SNP: rs137853329, ClinVar: RCV000012216

In a 17-year-old patient (patient 2) with hypohidrotic ectodermal dysplasia with immunodeficiency-1 (EDAID1; 300291), Orange et al. (2002) identified a hemizygous c.1207C-T transition in exon 10 of the IKBKG gene, resulting in a gln403-to-ter (Q403X) truncation within the zinc finger domain of the protein. The patient had a history of cutaneous granulomas and the presence of Mycobacterium avium-intracellulare in blood, bone marrow, and skin, as well as deficient specific antibody and lymphoproliferative responses and reduced natural killer cell function.


.0016   ECTODERMAL DYSPLASIA AND IMMUNODEFICIENCY 1

IKBKG, IVS6DS, G-A, +5
SNP: rs1569556603, ClinVar: RCV000012217

In affected members and obligate carriers of a family with ectodermal dysplasia and immunodeficiency-1 (EDAID1; 300291), originally reported by Lie et al. (1978), Orstavik et al. (2006) identified a +5G-A transition in the splice donor site of exon 6 of the IKBKG gene. The mutation was not found in unaffected family members or 140 control chromosomes. RT-PCR analysis of fibroblast RNA from an aborted affected male fetus demonstrated skipping of exons 4, 5, and 6, which resulted in a truncated protein of about 35 kD. IKBA degradation was strongly impaired in the fetal fibroblasts, suggesting impaired NFKB signaling. One healthy carrier female had a completely skewed X-inactivation pattern with the normal X active, whereas the 2 other female carriers had a random X-inactivation pattern.


.0017   INCONTINENTIA PIGMENTI

IKBKG, 1-BP DUP, 1409A
SNP: rs2148385141, ClinVar: RCV000012218

In a female infant with incontinentia pigmenti (IP; 308300) and transient immunodeficiency (see 300291), Martinez-Pomar et al. (2005) identified a 1-bp duplication (c.1049dupA) in exon 7 of the IKBKG gene, within a run of 3 adenosines. The mutation resulted in a frameshift and a premature stop codon at position 284. The X-inactivation status of peripheral blood cells from the patient was evaluated at 24, 30, 38, and 48 months of age and was found to have progressed from random at 24 and 30 months to skewed at 38 and 48 months of age, at which point her immunodeficiency had disappeared. Martinez-Pomar et al. (2005) stated that this was the first time that selection against the mutated X chromosome in X-linked disease had been documented in vivo.


.0018   IMMUNODEFICIENCY 33

IKBKG, IVS8AS, G-A, -1
SNP: rs2148385355, ClinVar: RCV003151721

In a 15-year-old boy with immunodeficiency-33 (IMD33; 300636), Orange et al. (2004) identified a de novo hemizygous G-to-A transition in intron 8 of the IKBKG gene (c.1056-1G-A), resulting in the skipping of exon 9 and the deletion of 19 residues (353_373). Analysis of patient peripheral blood and buccal epithelial cells showed presence of both mutant and wildtype transcripts. Western blot analysis using an antibody against the LZ domain showed barely detectable IKBKG protein levels in patient cells, whereas an antibody against the zinc finger domain showed normal protein levels. Presence of some normal transcripts may explain immunodeficiency without signs of ectodermal dysplasia in this patient. In vitro functional expression studies showed impaired, but not absent, nuclear translocation of NFKB in stimulated patient B cells compared to controls, as well as variable response to TNFA. Orange et al. (2004) postulated that exon 9 may be dispensable for ectodermal development.


.0019   IMMUNODEFICIENCY 33

IKBKG, 1-BP INS, 110C
SNP: rs1569556522, ClinVar: RCV000012220

In a patient with immunodeficiency-33 (IMD33; 300636), originally reported by Niehues et al. (2004), Puel et al. (2006) identified a hemizygous 1-bp insertion in exon 2 of the IKBKG gene (c.110_111insC) that resulted in the most-upstream premature translation termination codon of any NEMO mutation described to that time (Met38fsTer48). An almost canonically Kozakian methionine AUG start codon (Kozak, 2002) is located at nucleotide positions 112-114, corresponding to methionine-38 in the NEMO protein. The likelihood of this AUG codon acting as a translation initiation site was estimated at 0.27 and increased to 0.51 in the sequence context found in the patient studied by Puel et al. (2006), versus 0.63 for the first AUG codon. The reinitiation of translation would result in the synthesis of a protein approximately 45 kD in size. This protein was the only isoform detected in the patient's cells.


.0020   ECTODERMAL DYSPLASIA AND IMMUNODEFICIENCY 1

IKBKG, ALA288GLY
SNP: rs137853330, ClinVar: RCV000012221

In a 2-year-old boy (patient 10) with anhidrotic ectodermal dysplasia and immunodeficiency-1 (EDAID1; 300291), Doffinger et al. (2001) identified a hemizygous c.863C-G transversion in the IKBKG gene, resulting in an ala288-to-gly (A288G) substitution in the second coiled-coil domain. The mutation was not found in 200 control chromosomes, and the patient's mother, who carried the mutation, showed skewed X inactivation in her blood cells. Functional studies of the variant were not performed.

Using temperature-induced unfolding, Vinolo et al. (2006) demonstrated that the A281G murine mutation in NEMO, corresponding to the human A288G mutation, causes an important loss in oligomer stability. Fluorescence studies showed that the tyrosine located in the adjacent zinc finger domain exhibits an alteration in its spectral properties. Functional complementation assays using NEMO-deficient pre-B and T lymphocytes showed that the pathogenic mutation reduces TNF-alpha (191160) and lipopolysaccharide-induced NFKB (see 164011) activation by altering the assembly of the IKK complex.


.0021   IMMUNODEFICIENCY 33

IKBKG, GLU315ALA
SNP: rs137853331, ClinVar: RCV000012222

In 4 affected members of an American family with immunodeficiency-33 (IMD33; 300636), Filipe-Santos et al. (2006) identified a hemizygous c.944A-C transversion in the IKBKG gene, resulting in a glu315-to-ala (E315A) substitution in the leucine zipper domain of the protein. The proband was a non-BCG-vaccinated adolescent with severe, persistent M. avium infection who did not respond well to antibiotic treatment.


.0022   IMMUNODEFICIENCY 33

IKBKG, ARG319GLN
SNP: rs137853332, ClinVar: RCV000012223

In affected males from 2 unrelated French and German families with immunodeficiency-33 (IMD33; 300636), Filipe-Santos et al. (2006) identified a hemizygous c.956G-A transition in the IKBKG gene, resulting in an arg319-to-gln (R319Q) substitution in the leucine zipper domain of the protein. The proband of the French family was a child with disseminated BCG who was well at age 8 years after treatment with antituberculosis antibiotics. The proband of the German family was a young non-BCG-vaccinated boy diagnosed with mycobacterial disease.


.0023   IMMUNODEFICIENCY 33

IKBKG, ARG173GLY
SNP: rs179363866, ClinVar: RCV000012224, RCV000059072

In a 4.5-year-old boy, born to unrelated Belgian parents, with immunodeficiency-33 (IMD33; 300636), Ku et al. (2007) identified a hemizygous c.518C-G transversion at the end of exon 4 of the NEMO gene, predicted to result in an arg173-to-gly (R173G) substitution at a conserved residue in the first coiled-coil domain. The mother was heterozygous for this mutation. RT-PCR analysis of patient cells showed the presence of 2 abnormal splicing products corresponding to the skipping of exons 4-6 and exons 5-6. Since the mutation occurred 2 nucleotides away from the end of the exon, it had abnormal splicing effects. Western blot analysis of patient cells showed decreased levels of NEMO compared to controls, suggesting partial NEMO deficiency. Patient fibroblasts showed impaired responses to IL1B and TNFA, suggesting impaired activation of NF-kappa-B. There was also an impaired cellular response to interleukin-1 receptor (IL1R; 147810), Toll-like receptor (TLR; see 601194), and tumor necrosis factor receptor (TNFR; see 191190) stimulation.


.0024   IMMUNODEFICIENCY 33, MALE-RESTRICTED

INCONTINENTIA PIGMENTI, INCLUDED
IKBKG, IVS4DS, C-T, +866
SNP: rs2071101767, ClinVar: RCV000757923, RCV001172475

In 3 boys from 2 unrelated families of European and Japanese descent with fatal immunodeficiency-33 (IMD33; 300636), Boisson et al. (2019) identified a deep intronic mutation (IVS4+866C-T) in the IKBKG gene (chrX.153,787,731C-T, GRCh37). The boy in the European family inherited the mutation from his mother, who had mild features of incontinentia pigmenti (IP; 308300). The mutation in the Japanese boy occurred de novo. The mutation was shown to create a new splicing donor site that resulted in a 44-nucleotide pseudoexon that produced a frameshift. Patient fibroblasts showed decreased levels of mutant transcript due to nonsense-mediated mRNA decay; there were also decreased levels of the mutant protein, suggesting a quantitative defect. The amount of aberrant splicing varied between cell types in the affected boys. In leukocytes, the abnormal splice variant predominated at approximately 97%, while in fibroblasts and iPSC-derived neuronal precursor cells, the abnormal RNA was measured at about 65%. The splicing factor SRSF6 (601944) was shown to bind to the pseudoexon, resulting in its inclusion in the mature mRNA. By knocking down SFSF6 or CLK (see 601951), a protein that phosphorylates SR proteins, the authors were able to restore wildtype expression in mutant cells. The variant segregated with the disorder in both families and was not found in the 1000 Genomes Project or gnomAD databases.


.0025   IMMUNODEFICIENCY 33

IKBKG, 18-BP DEL, NT811
SNP: rs2071141016, ClinVar: RCV001172476

In a 6.5-year-old French boy (patient 1) with immunodeficiency-33 (IMD33; 300636), Ku et al. (2005) identified a hemizygous 18-bp in-frame deletion (c.811_828del) in exon 7 of the IKBKG gene, resulting in the deletion of residues 271 to 276 in the coiled-coil 2 domain. The patient's mother, who had conical teeth, was heterozygous for the mutation; blood analysis showed random X inactivation. Western blot analysis of patient cells showed presence of the IKBKG protein, but fibroblasts responded weakly to stimulation with TNFA and IL1B compared to controls. The findings were consistent with a hypomorphic allele. The patient had hypodontia and conical teeth, but no other features of ectodermal dysplasia.


.0026   IMMUNODEFICIENCY 33

IKBKG, LEU80PRO
SNP: rs2071059718, ClinVar: RCV001172477

In an 11-year-old German boy (patient 2) with ectodermal dysplasia and immunodeficiency-33 (IMD33; 300636), Ku et al. (2005) identified a hemizygous c.239T-C transition in exon 3 of the IKBKG gene, resulting in a leu80-to-pro (L80P) substitution in the first coiled-coil domain. The patient's unaffected mother was heterozygous for the mutation; blood analysis showed skewed X-inactivation. Western blot analysis of patient cells showed presence of the IKBKG protein, but fibroblasts responded weakly to stimulation with TNFA and IL1B compared to controls. The findings were consistent with a hypomorphic allele. The patient had hypodontia and conical teeth, but no other features of ectodermal dysplasia.


.0027   ECTODERMAL DYSPLASIA AND IMMUNODEFICIENCY 1, MALE-RESTRICTED

INCONTINENTIA PIGMENTI, INCLUDED
IKBKG, 2-BP DEL, 1182TT
ClinVar: RCV001172478, RCV001172479

In a 6-year-old boy with ectodermal dysplasia and immunodeficiency-1 (EDAID1; 300291), Roberts et al. (2010) identified a hemizygous 2-bp deletion (c.1182_1183delTT) in the IKBKG gene, predicted to result in a frameshift and premature termination affecting the zinc finger domain. The mutation was inherited from the patient's mother, who had features of incontinentia pigmenti (IP; 308300). Functional studies of the variant and studies of patient cells were not performed. The boy also had the additional features of osteopetrosis and edema, as well as some skin features of IP. Roberts et al. (2010) described the patient as having OLEDAID.


.0028   ECTODERMAL DYSPLASIA AND IMMUNODEFICIENCY 1, MALE-RESTRICTED

INCONTINENTIA PIGMENTI, INCLUDED
IKBKG, IVS8DS, G-C, +5
SNP: rs1557236796, ClinVar: RCV000523987, RCV001172480, RCV001172481

In a male infant with lethal ectodermal dysplasia and immunodeficiency-1 (EDAID1; 300291), Johnston et al. (2016) identified a hemizygous G-to-C transversion (c.1117+5G-C) in intron 8 of the IKBKG gene, predicted to result in a splice site alteration, the skipping of exon 9, and a frameshift with premature termination of the C-terminal region (Arg352SerfsTer22). The mutation was inherited from his mother who had features of incontinentia pigmenti (IP; 308300); X-chromosome inactivation studies showed nonrandom maternal X inactivation with expression only of the wildtype allele. Western blot analysis of the proband's cells showed reduced-sized mutant IKBKG, consistent with the predicted truncated protein.


.0029   IMMUNODEFICIENCY 33

IKBKG, GLU57LYS ({dbSNP rs148695964})
SNP: rs148695964, gnomAD: rs148695964, ClinVar: RCV000059069, RCV001172482, RCV003493432, RCV003905029

In a 2-year-old boy with immunodeficiency-33 (IMD33; 300636), Frans et al. (2017) identified a hemizygous c.169G-A transition (c.169G-A, NM_003639) in the IKBKG gene, resulting in a glu57-to-lys (E57K) substitution in the N-terminal domain. His mother, who also carried the mutation, had a history of recurrent sinorespiratory infections but no signs of IP. The variant was present at a frequency of 0.001 in the European population in the ExAC database (August, 2016). Patient peripheral blood cells showed mildly decreased IL6 production after stimulation with IL1B compared to controls. However, NEMO expression in patient fibroblasts was normal, and IKBA was degraded normally upon stimulation with IL1B or TNFA, suggesting a specific effect of the mutation. In vitro functional expression studies in IKBKG-null cells transfected with the mutation showed impaired IL6 production after stimulation with TNFA or IL1B compared to controls. Frans et al. (2017) noted that mutations affecting the N terminus of NEMO tend to lead to decreased production of immunoglobulins; the authors postulated a hypomorphic effect of this variant.


.0030   IMMUNODEFICIENCY 33

IKBKG, c.1-16G-C
SNP: rs2070806148, ClinVar: RCV001172483

In 3 adult males from 2 unrelated families (families A and B) with immunodeficiency-33 (IMD33; 300636) manifest as disseminated mycobacterial infections, Hsu et al. (2018) identified a hemizygous c.1-16G-C transversion at the last base of the first untranslated exon. Analysis of patient cells showed a splicing abnormality, resulting in a 110-bp deletion at the 3-prime end of exon 1. This molecular defect resulted in decreased transcript and protein levels compared to controls (about 30%). Cells derived from the patients in families A and B failed to upregulate cytokines in response to certain TLR agonists, suggesting that this IKBKG mutation is hypomorphic.


.0031   ECTODERMAL DYSPLASIA AND IMMUNODEFICIENCY 1, MALE-RESTRICTED

INCONTINENTIA PIGMENTI, INCLUDED
IKBKG, IVS9DS, G-A, +1
SNP: rs2071161458, ClinVar: RCV001172484, RCV001172485

In a 2-year-old boy (P1) with ectodermal dysplasia and immunodeficiency-1 (EDAID1; 300291), Heller et al. (2020) identified a hemizygous G-to-A transition in intron 9 of the IKBKG gene. The mutation caused the skipping of exon 9, a frameshift, and premature termination (Arg352Serfs373Ter), with loss of the zinc finger domain. Patient cells showed decreased IKBKG levels compared to controls; in vitro studies showed impaired degradation of IKBA and impaired IL6 production upon stimulation with IL1B and TNFA. The patient had a severe phenotype with functional T- and B-cell deficiency. He underwent hematopoietic stem cell transplant. His mother and sister, who were heterozygous for the mutation, had incontinentia pigmenti (IP; 308300).


.0032   IMMUNODEFICIENCY 33

IKBKG, ASP113ASN
SNP: rs179363896, gnomAD: rs179363896, ClinVar: RCV000059070, RCV001172486, RCV002470750

In a boy (patient 1) with immunodeficiency-33 (IMD33; 300636), Abbott et al. (2014) identified a hemizygous c.337G-A transition in the IKBKG gene, resulting in an asp113-to-asn (D113N) substitution in the first coiled-coil domain. The patient had impaired NK cell function and impaired T-cell receptor signaling with decreased NFKB activity, but Toll-like receptor signaling appeared to be intact. He underwent successful bone marrow transplant. The patient had previously been reported in detail by Salt et al. (2008); his unaffected mother also carried the mutation.

Heller et al. (2020) identified a hemizygous D113N mutation in a boy with IMD33. His mother and grandmother, who were presumably unaffected, carried the heterozygous mutation. However, the proband's 40-year-old male cousin, who did not have recurrent infections and had normal response to polysaccharide antibodies, was hemizygous for D113N. Heller et al. (2020) noted that the allele frequency for this variant is rather high (0.009572), and that some suggest it may be a polymorphism (see Fusco et al., 2004).


.0033   AUTOINFLAMMATORY DISEASE, SYSTEMIC, X-LINKED

IKBKG, 597G-A
SNP: rs2148382665, ClinVar: RCV001762996, RCV002251769

In a boy (P1) with X-linked systemic autoinflammatory disease (SAIDX; 301081), de Jesus et al. (2020) identified a de novo hemizygous c.597G-A transition in exon 5 of the IKBKG gene, predicted to result in a silent substitution (V199V), but also predicted to disrupt an exonic splicing enhancer and an exon-identity element in exon 5, resulting in alternative splicing. The patient had lymphohistiocytic panniculitis, chorioretinitis, progressive B cell lymphopenia, hypogammaglobulinemia, and conical teeth, prompting targeted sequencing of the IKBKG gene. Functional studies of the variant were not performed.

In a 9-year-old boy (P1) with SAIDX, Lee et al. (2022) identified a de novo heterozygous c.597G-A transition in the IKBKG gene. Analysis of patient cells detected a shorter NEMO mRNA splice variant lacking the 153 nucleotides of exon 5. Patient cells showed a higher ratio of mutant to full-length cDNA compared to healthy controls. The mutation, which was found by targeted sequencing of NEMO and further confirmed by Sanger sequencing of genomic DNA, was not present in 1,150 healthy individuals. In vitro functional expression studies in patient peripheral blood cells showed that stimulation with poly(I:C) or viral infection resulted in enhanced type I IFN production likely due to a stabilized complex of mutant IKBKG and IKKi (605048). The patient had panniculitis, optic neuritis, chorioretinitis, hypogammaglobulinemia, conical teeth, and CNS hemorrhage.


.0034   AUTOINFLAMMATORY DISEASE, SYSTEMIC, X-LINKED

IKBKG, IVS5DS, T-G, +2
SNP: rs2148382857, ClinVar: RCV002250446

In a boy (P3) with X-linked systemic autoinflammatory disease (SAIDX; 301081), de Jesus et al. (2020) identified a de novo hemizygous T-to-G transversion in intron 5 of the IKBKG gene (c.671+2T-G), predicted to result in the deletion of exon 5. Functional studies of the variant were not performed. The patient had panniculitis, progressive B cell lymphopenia, thrombocytopenia, and hypogammaglobulinemia without major infections.

In a 5-year-old boy (P2) with SAIDX, Lee et al. (2022) identified a de novo hemizygous T-to-G transversion in intron 5 of the IKBKG gene (chrX:153,788,776T-G), disrupting a splice site and resulting in the production of a mutant transcript lacking exon 5. Analysis of patient cells detected a shorter NEMO mRNA splice variant lacking the 153 nucleotides of exon 5. Patient cells showed a higher ratio of mutant to full-length cDNA compared to healthy controls. The mutation, which was found by targeted sequencing of NEMO and further confirmed by Sanger sequencing of genomic DNA, was not present in 1,150 healthy individuals. In vitro functional expression studies in patient peripheral blood cells showed that stimulation with poly(I:C) or viral infection resulted in enhanced type I IFN production likely due to a stabilized complex of mutant IKBKG and IKKi (605048). The patient had lymphocytic panniculitis, hypogammaglobulinemia, and a CNS cortical bleed.


.0035   AUTOINFLAMMATORY DISEASE, SYSTEMIC, X-LINKED

IKBKG, IVS5DS, G-A, +5
SNP: rs2148382861, ClinVar: RCV002250447

In a boy (P2) with X-linked systemic autoinflammatory disease (SAIDX; 301081), de Jesus et al. (2020) identified a de novo hemizygous G-to-A transition in intron 5 of the IKBKG gene (c.671+5G-A), predicted to result in the deletion of exon 5. Functional studies of the variant were not performed. The patient had panniculitis, uveitis, progressive B cell lymphopenia, and hypogammaglobulinemia without major infections.

In a 3-year-old boy (P3) with SAIDX, Lee et al. (2022) identified a de novo heterozygous G-to-A transition (chrX:153,788,779G-A) in the IKBKG gene, resulting in a splice site alteration and the deletion of exon 5. Analysis of patient cells detected a shorter NEMO mRNA splice variant lacking the 153 nucleotides of exon 5. Patient cells showed a higher ratio of mutant to full-length cDNA compared to healthy controls. The mutation, which was found by targeted sequencing of NEMO and further confirmed by Sanger sequencing of genomic DNA, was not present in 1,150 healthy individuals. In vitro functional expression studies in patient peripheral blood cells showed that stimulation with poly(I:C) or viral infection resulted in enhanced type I IFN production likely due to a stabilized complex of mutant IKBKG and IKKi (605048). The patient had panniculitis, uveitis, lymphopenia, hypogammaglobulinemia, and a CNS hemorrhage.


.0036   AUTOINFLAMMATORY DISEASE, SYSTEMIC, X-LINKED

IKBKG, IVS5AS, A-G, -2
SNP: rs2071121693, ClinVar: RCV002250444

In a girl (P4) with X-linked systemic autoinflammatory disease (SAIDX; 301081), de Jesus et al. (2020) identified a de novo heterozygous A-to-G transition in intron 5 of the IKBKG gene (c.519-2A-G), predicted to result in the deletion of exon 5. Functional studies of the variant were not performed. The patient had panniculitis, lipodystrophy, anemia, myositis, and progressive B cell lymphopenia without hypogammaglobulinemia.


REFERENCES

  1. Abbott, J. K., Quinones, R. R., de la Morena, M. T., Gelfand, E. W. Successful hematopoietic cell transplantation in patients with unique NF-kappa-B essential modulator (NEMO) mutations. (Letter) Bone Marrow Transplant. 49: 1446-1447, 2014. [PubMed: 25068423] [Full Text: https://doi.org/10.1038/bmt.2014.157]

  2. Akiyama, M., Aranami, A., Sasaki, Y., Ebihara, T., Sugiura, M. Familial linear and whorled nevoid hypermelanosis. J. Am. Acad. Derm. 30: 831-833, 1994. [PubMed: 8169255] [Full Text: https://doi.org/10.1016/s0190-9622(94)70090-7]

  3. Aradhya, S., Bardaro, T., Galgoczy, P., Yamagata, T., Esposito, T., Patlan, H., Ciccodicola, A., Munnich, A., Kenwrick, S., Platzer, M., D'Urso, M., Nelson, D. L. Multiple pathogenic and benign genomic rearrangements occur at a 35 kb duplication involving the NEMO and LAGE2 genes. Hum. Molec. Genet. 10: 2557-2567, 2001. [PubMed: 11709543] [Full Text: https://doi.org/10.1093/hmg/10.22.2557]

  4. Aradhya, S., Courtois, G., Rajkovic, A., Lewis, R. A., Levy, M., Israel, A., Nelson, D. L. Atypical forms of incontinentia pigmenti in male individuals result from mutations of a cytosine tract in exon 10 of NEMO (IKK-gamma). Am. J. Hum. Genet. 68: 765-771, 2001. [PubMed: 11179023] [Full Text: https://doi.org/10.1086/318806]

  5. Aradhya, S., Woffendin, H., Jakins, T., Bardaro, T., Esposito, T., Smahi, A., Shaw, C., Levy, M., Munnich, A., D'Urso, M., Lewis, R. A., Kenwrick, S., Nelson, D. L. A recurrent deletion in the ubiquitously expressed NEMO (IKK-gamma) gene accounts for the vast majority of incontinentia pigmenti mutations. Hum. Molec. Genet. 10: 2171-2179, 2001. [PubMed: 11590134] [Full Text: https://doi.org/10.1093/hmg/10.19.2171]

  6. Bardaro, T., Falco, G., Sparago, A., Mercadante, V., Molins, E. G., Tarantino, E., Ursini, M. V., D'Urso, M. Two cases of misinterpretation of molecular results in incontinentia pigmenti, and a PCR-based method to discriminate NEMO/IKK-gamma gene deletion. Hum. Mutat. 21: 8-11, 2003. [PubMed: 12497627] [Full Text: https://doi.org/10.1002/humu.10150]

  7. Boisson, B., Honda, Y., Ajiro, M., Bustamante, J., Bendavid, M., Gennery, A. R., Kawasaki, Y., Ichishima, J., Osawa, M., Nihira, H., Shiba, T., Tanaka, T., and 16 others. Rescue of recurrent deep intronic mutation underlying cell type-dependent quantitative NEMO deficiency. J. Clin. Invest. 129: 583-597, 2019. [PubMed: 30422821] [Full Text: https://doi.org/10.1172/JCI124011]

  8. Brummelkamp, T. R., Nijman, S. M. B., Dirac, A. M. G., Bernards, R. Loss of the cylindromatosis tumour suppressor inhibits apoptosis by activating NF-kappa-B. Nature 424: 797-801, 2003. [PubMed: 12917690] [Full Text: https://doi.org/10.1038/nature01811]

  9. Chiu, J. C., Ko, H. W., Edery, I. NEMO/NLK phosphorylates PERIOD to initiate a time-delay phosphorylation circuit that sets circadian clock speed. Cell 145: 357-370, 2011. Note: Erratum: Cell 145: 635 only, 2011. [PubMed: 21514639] [Full Text: https://doi.org/10.1016/j.cell.2011.04.002]

  10. de Jesus, A. A., Hou, Y., Brooks, S., Malle, L., Biancotto, A., Huang, Y., Calvo, K. R., Marrero, B., Moir, S., Oler, A. J., Deng, Z., Montealegre Sanchez, G. A., and 75 others. Distinct interferon signatures and cytokine patterns define additional systemic autoinflammatory diseases. J. Clin. Invest. 130: 1669-1682, 2020. [PubMed: 31874111] [Full Text: https://doi.org/10.1172/JCI129301]

  11. Doffinger, R., Smahi, A., Bessia, C., Geissmann, F., Feinberg, J., Durandy, A., Bodemer, C., Kenwrick, S., Dupuis-Girod, S., Blanche, S., Wood, P., Rabia, S. H., and 16 others. X-linked anhidrotic ectodermal dysplasia with immunodeficiency is caused by impaired NF-kappa-B signaling. Nature Genet. 27: 277-285, 2001. [PubMed: 11242109] [Full Text: https://doi.org/10.1038/85837]

  12. Filipe-Santos, O., Bustamante, J., Haverkamp, M. H., Vinolo, E., Ku, C.-L., Puel, A., Frucht, D. M., Christel, K., von Bernuth, H., Jouanguy, E., Feinberg, J., Durandy, A., and 25 others. X-linked susceptibility to mycobacteria is caused by mutations in NEMO impairing CD40-dependent IL-12 production. J. Exp. Med. 203: 1745-1759, 2006. [PubMed: 16818673] [Full Text: https://doi.org/10.1084/jem.20060085]

  13. Frans, G., van der Werff Ten Bosch, J., Moens, L., Gijsbers, R., Changi-Ashtiani, M., Rokni-Zadeh, H., Shahrooei, M., Wuyts, G., Meyts, I., Bossuyt, X. Functional evaluation of an IKBK variant suspected to cause immunodeficiency without ectodermal dysplasia. J. Clin. Immun. 37: 801-810, 2017. [PubMed: 28993958] [Full Text: https://doi.org/10.1007/s10875-017-0448-9]

  14. Fusco, F., Bardaro, T., Fimiani, G., Mercadante, V., Miano, M. G., Falco, G., Israel, A., Courtois, G., D'Urso, M., Ursini, M. V. Molecular analysis of the genetic defect in a large cohort of IP patients and identification of novel NEMO mutations interfering with NF-kappa-B activation. Hum. Molec. Genet. 13: 1763-1773, 2004. [PubMed: 15229184] [Full Text: https://doi.org/10.1093/hmg/ddh192]

  15. Fusco, F., Paciolla, M., Napolitano, F., Pescatore, A., D'Addario, I., Bal, E., Lioi, M. B., Smahl, A., Miano, M. G., Ursini, M. V. Genomic architecture at the incontinentia pigmenti locus favours de novo pathological alleles through different mechanisms. Hum. Molec. Genet. 21: 1260-1271, 2012. [PubMed: 22121116] [Full Text: https://doi.org/10.1093/hmg/ddr556]

  16. Gautheron, J., Pescatore, A., Fusco, F., Esposito, E., Yamaoka, S., Agou, F., Ursini, M. V., Courtois, G. Identification of a new NEMO/TRAF6 interface affected in incontinentia pigmenti pathology. Hum. Molec. Genet. 19: 3138-3149, 2010. [PubMed: 20529958] [Full Text: https://doi.org/10.1093/hmg/ddq222]

  17. Gerlach, B., Cordier, S. M., Schmukle, A. C., Emmerich, C. H., Rieser, E., Haas, T. L., Webb, A. I., Rickard, J. A., Anderton, H., Wong, W. W.-L., Nachbur, U., Gangoda, L., Warnken, U., Purcell, A. W., Silke, J., Walczak, H. Linear ubiquitination prevents inflammation and regulates immune signalling. Nature 471: 591-596, 2011. [PubMed: 21455173] [Full Text: https://doi.org/10.1038/nature09816]

  18. Heller, S., Kolsch, U., Magg, T., Kruger, R., Scheuern, A., Schneider, H., Eichinger, A., Wahn, V., Unterwalder, N., Lorenz, M., Schwarz, K., Meisel, C., Schulz, A., Hauck, F., von Bernuth, H. T cell impairment is predictive for a severe clinical course in NEMO deficiency. J. Clin. Immun. 40: 421-434, 2020. [PubMed: 31965418] [Full Text: https://doi.org/10.1007/s10875-019-00728-y]

  19. Hsu, A. P., Zerbe, C. S., Foruraghi, L., Iovine, N. L., Leiding, J. W., Mushatt, D. M., Wild, L., Kuhns, D. B., Holland, S. M. IKBKG (NEMO) 5-prime untranslated splice mutations lead to severe, chronic disseminated mycobacterial infections. Clin. Infect. Dis. 67: 456-459, 2018. [PubMed: 29534156] [Full Text: https://doi.org/10.1093/cid/ciy186]

  20. Ikeda, F., Deribe, Y. L., Skanland, S. S., Stieglitz, B., Grabbe, C., Franz-Wachtel, M., van Wijk, S. J. L., Goswami, P., Nagy, V., Terzic, J., Tokunaga, F., Androulidaki, A., Nakagawa, T., Pasparakis, M., Iwai, K., Sundberg, J. P., Schaefer, L., Rittinger, K., Macek, B., Dikic, I. SHARPIN forms a linear ubiquitin ligase complex regulating NF-kappa-B activity and apoptosis. Nature 471: 637-641, 2011. [PubMed: 21455181] [Full Text: https://doi.org/10.1038/nature09814]

  21. Jain, A., Ma, C. A., Liu, S., Brown, M., Cohen, J., Strober, W. Specific missense mutations in NEMO result in hyper-IgM syndrome with hypohydrotic (sic) ectodermal dysplasia. Nature Immun. 2: 223-228, 2001. [PubMed: 11224521] [Full Text: https://doi.org/10.1038/85277]

  22. Jain, A., Ma, C. A., Lopez-Granados, E., Means, G., Brady, W., Orange, J. S., Liu, S., Holland, S., Derry, J. M. J. Specific NEMO mutations impair CD40-mediated c-Rel activation and B cell terminal differentiation. J. Clin. Invest. 114: 1593-1602, 2004. [PubMed: 15578091] [Full Text: https://doi.org/10.1172/JCI21345]

  23. Jin, D. Y., Jeang, K. T. Isolation of full-length cDNA and chromosomal localization of human NF-kappaB modulator NEMO to Xq28. J. Biomed. Sci. 6: 115-120, 1999. [PubMed: 10087442] [Full Text: https://doi.org/10.1007/BF02256442]

  24. Johnston, A. M., Niemela, J., Rosenzweig, S. D., Fried, A. J., Delmonte, O. M., Fleisher, T. A., Kuehn, H. A novel mutation in IKBKG/NEMO leads to ectodermal dysplasia with severe immunodeficiency (EDA-ID). (Letter) J. Clin. Immun 36: 541-543, 2016. [PubMed: 27368913] [Full Text: https://doi.org/10.1007/s10875-016-0309-y]

  25. Kosaki, K., Shimasaki, N., Fukushima, H., Hara, M., Ogata, T., Matsuo, N. Female patient showing hypohidrotic ectodermal dysplasia and immunodeficiency (HED-ID). (Letter) Am. J. Hum. Genet. 69: 664-665, 2001. [PubMed: 11484156] [Full Text: https://doi.org/10.1086/323003]

  26. Kovalenko, A., Chable-Bessia, C., Cantarella, G., Israel, A., Wallach, D., Courtois, G. The tumour suppressor CYLD negatively regulates NF-kappa-B signalling by deubiquitination. Nature 424: 801-805, 2003. [PubMed: 12917691] [Full Text: https://doi.org/10.1038/nature01802]

  27. Kozak, M. Pushing the limits of the scanning mechanism for initiation of translation. Gene 299: 1-34, 2002. [PubMed: 12459250] [Full Text: https://doi.org/10.1016/s0378-1119(02)01056-9]

  28. Ku, C.-L., Dupuis-Girod, S., Dittrich, A.-M., Bustamante, J., Santos, O. F., Schulze, I., Bertrand, Y., Couly, G., Bodemer, C., Bossuyt, X., Picard, C., Casanova, J.-L. NEMO mutations in 2 unrelated boys with severe infections and conical teeth. Pediatrics 115: e615-e619, 2005. Note: Electronic Article. [PubMed: 15833888] [Full Text: https://doi.org/10.1542/peds.2004-1754]

  29. Ku, C.-L., Picard, C., Erdos, M., Jeurissen, A., Bustamante, J., Puel, A., von Bernuth, H., Filipe-Santos, O., Chang, H.-H., Lawrence, T., Raes, M., Marodi, L., Bossuyt, X., Casanova, J.-L. IRAK4 and NEMO mutations in otherwise healthy children with recurrent invasive pneumococcal disease. J. Med. Genet. 44: 16-23, 2007. [PubMed: 16950813] [Full Text: https://doi.org/10.1136/jmg.2006.044446]

  30. Lee, Y., Wessel, A. W., Xu, J., Reinke, J. G., Lee, E., Kim, S. M., Hsu, A. P., Zilberman-Rudenko, J., Cao, S., Enos, C., Brooks, S. R., Deng, Z., and 11 others. Genetically programmed alternative splicing of NEMO mediates an autoinflammatory disease phenotype. J. Clin. Invest. 132: e128808, 2022. [PubMed: 35289316] [Full Text: https://doi.org/10.1172/JCI128808]

  31. Li, Q., Van Antwerp, D., Mercurio, F., Lee, K.-F., Verma, I. M. Severe liver degeneration in mice lacking the I-kappa-B kinase 2 gene. Science 284: 321-325, 1999. [PubMed: 10195897] [Full Text: https://doi.org/10.1126/science.284.5412.321]

  32. Li, Y., Kang, J., Friedman, J., Tarassishin, L., Ye, J., Kovalenko, A., Wallach, D., Horwitz, M. S. Identification of a cell protein (FIP-3) as a modulator of NF-kappa-B activity and as a target of an adenovirus inhibitor of tumor necrosis factor alpha-induced apoptosis. Proc. Nat. Acad. Sci. 96: 1042-1047, 1999. [PubMed: 9927690] [Full Text: https://doi.org/10.1073/pnas.96.3.1042]

  33. Lie, S. O., Froland, S., Brandtzaeg, P., Vandvik, B., Steen-Johnsen, J. Transient B cell immaturity with intractable diarrhoea: a possible new immunodeficiency syndrome. J. Inherit. Metab. Dis. 1: 137-143, 1978. [PubMed: 117248] [Full Text: https://doi.org/10.1007/BF01805582]

  34. Makris, C., Godfrey, V. L., Krahn-Senftleben, G., Takahashi, T., Roberts, J. L., Schwarz, T., Feng, L., Johnson, R. S., Karin, M. Female mice heterozygous for IKK-gamma/NEMO deficiencies develop a dermatopathy similar to the human X-linked disorder incontinentia pigmenti. Molec. Cell 5: 969-979, 2000. [PubMed: 10911991] [Full Text: https://doi.org/10.1016/s1097-2765(00)80262-2]

  35. Martinez-Pomar, N., Munoz-Saa, I., Heine-Suner, D., Martin, A., Smahi, A., Matamoros, N. A new mutation in exon 7 of NEMO gene: late skewed X-chromosome inactivation in an incontinentia pigmenti female patient with immunodeficiency. Hum. Genet. 118: 458-465, 2005. [PubMed: 16228229] [Full Text: https://doi.org/10.1007/s00439-005-0068-y]

  36. Matsuzawa, A., Tseng, P.-H., Vallabhapurapu, S., Luo, J.-L., Zhang, W., Wang, H., Vignali, D. A. A., Gallagher, E., Karin, M. Essential cytoplasmic translocation of a cytokine receptor-assembled signaling complex. Science 321: 663-668, 2008. Note: Erratum: Science 322: 375 only, 2008. [PubMed: 18635759] [Full Text: https://doi.org/10.1126/science.1157340]

  37. May, M. J., D'Acquisto, F., Madge, L. A., Glockner, J., Pober, J. S., Ghosh, S. Selective inhibition of NF-kappa-B activation by a peptide that blocks the interaction of NEMO with the I-kappa-B kinase complex. Science 289: 1550-1554, 2000. [PubMed: 10968790] [Full Text: https://doi.org/10.1126/science.289.5484.1550]

  38. Nenci, A., Becker, C., Wullaert, A., Gareus, R., van Loo, G., Danese, S., Huth, M., Nikolaev, A., Neufert, C., Madison, B., Gumucio, D., Neurath, M. F., Pasparakis, M. Epithelial NEMO links innate immunity to chronic intestinal inflammation. Nature 446: 557-561, 2007. [PubMed: 17361131] [Full Text: https://doi.org/10.1038/nature05698]

  39. Niehues, T., Reichenbach, J., Neubert, J., Gudowius, S., Puel, A., Horneff, G., Lainka, E., Dirksen, U., Schroten, H., Doffinger, R., Casanova, J. L., Wahn, V. Nuclear factor kappa-B essential modulator-deficient child with immunodeficiency yet without anhidrotic ectodermal dysplasia. J. Allergy Clin. Immun. 114: 1456-1462, 2004. [PubMed: 15577852] [Full Text: https://doi.org/10.1016/j.jaci.2004.08.047]

  40. Nishikomori, R., Akutagawa, H., Maruyama, K., Nakata-Hizume, M., Ohmori, K., Mizuno, K., Yachie, A., Yasumi, T., Kusunoki, T., Heike, T., Nakahata, T. X-linked ectodermal dysplasia and immunodeficiency caused by reversion mosaicism of NEMO reveals a critical role for NEMO in human T-cell development and/or survival. Blood 103: 4565-4572, 2004. [PubMed: 14726382] [Full Text: https://doi.org/10.1182/blood-2003-10-3655]

  41. Orange, J. S., Brodeur, S. R., Jain, A., Bonilla, F. A., Schneider, L. C., Kretschmer, R., Nurko, S., Rasmussen, W. L., Kohler, J. R., Gellis, S. E., Ferguson, B. M., Strominger, J. L., Zonana, J., Ramesh, N., Ballas, Z. K., Geha, R. S. Deficient natural killer cell cytotoxicity in patients with IKK-gamma/NEMO mutations. J. Clin. Invest. 109: 1501-1509, 2002. [PubMed: 12045264] [Full Text: https://doi.org/10.1172/JCI14858]

  42. Orange, J. S., Jain, A., Ballas, Z. K., Schneider, L. C., Geha, R. S., Bonilla, F. A. The presentation and natural history of immunodeficiency caused by nuclear factor kappa-B essential modulator mutation. J. Allergy Clin. Immun. 113: 725-733, 2004. [PubMed: 15100680] [Full Text: https://doi.org/10.1016/j.jaci.2004.01.762]

  43. Orange, J. S., Levy, O., Brodeur, S. R., Krzewski, K., Roy, R. M., Niemela, J. E., Fleisher, T. A., Bonilla, F. A., Geha, R. S. Human nuclear factor kappa-B essential modulator mutation can result in immunodeficiency without ectodermal dysplasia. J. Allergy Clin. Immun. 114: 650-656, 2004. [PubMed: 15356572] [Full Text: https://doi.org/10.1016/j.jaci.2004.06.052]

  44. Orstavik, K. H., Kristiansen, M., Knudsen, G. P., Storhaug, K., Vege, A., Eiklid, K., Abrahamsen, T. G., Smahi, A., Steen-Johnsen, J. Novel splicing mutation in the NEMO (IKK-gamma) gene with severe immunodeficiency and heterogeneity of X-chromosome inactivation. Am. J. Med. Genet. 140A: 31-39, 2006. [PubMed: 16333836] [Full Text: https://doi.org/10.1002/ajmg.a.31026]

  45. Puel, A., Reichenbach, J., Bustamante, J., Ku, C.-L., Feinberg, J., Doffinger, R., Bonnet, M., Filipe-Santos, O., de Beaucoudrey, L., Durandy, A., Horneff, G., Novelli, F., Wahn, V., Smahi, A., Israel, A., Niehues, T., Casanova, J.-L. The NEMO mutation creating the most-upstream premature stop codon is hypomorphic because of a reinitiation of translation. Am. J. Hum. Genet. 78: 691-701, 2006. [PubMed: 16532398] [Full Text: https://doi.org/10.1086/501532]

  46. Roberts, C. M. L., Angus, J. E., Leach, I. H., McDermott, E. M., Walker, D. A., Ravenscroft, J. C. A novel NEMO gene mutation causing osteopetrosis, lymphoedema, hypohidrotic ectodermal dysplasia and immunodeficiency (OL-HED-ID). Europ. J. Pediat. 169: 1403-1407, 2010. [PubMed: 20499091] [Full Text: https://doi.org/10.1007/s00431-010-1206-7]

  47. Roberts, J. L., Morrow, B., Vega-Rich, C., Salafia, C. M., Nitowsky, H. M. Incontinentia pigmenti in a newborn male infant with DNA confirmation. Am. J. Med. Genet. 75: 159-163, 1998. [PubMed: 9450877] [Full Text: https://doi.org/10.1002/(sici)1096-8628(19980113)75:2<159::aid-ajmg7>3.0.co;2-o]

  48. Rothwarf, D. M., Zandi, E., Natoli, G., Karin, M. IKK-gamma is an essential regulatory subunit of the I-kappa-B kinase complex. Nature 395: 297-300, 1998. [PubMed: 9751060] [Full Text: https://doi.org/10.1038/26261]

  49. Rudolph, D., Yeh, W.-C., Wakeham, A., Rudolph, B., Nallainathan, D., Potter, J., Elia, A. J., Mak, T. W. Severe liver degeneration and lack of NF-kappa-B activation in NEMO/IKK-gamma-deficient mice. Genes Dev. 14: 854-862, 2000. [PubMed: 10766741]

  50. Salt, B. H., Niemela, J. E., Pandey, R., Hanson, E. P., Deering, R. P., Quinones, R., Jain, A., Orange, J. S., Gelfand, E. W. IKBKG (nuclear factor-kappa-B essential modulator) mutation can be associated with opportunistic infection without impairing Toll-like receptor function. J. Allergy Clin. Immun. 121: 976-982, 2008. [PubMed: 18179816] [Full Text: https://doi.org/10.1016/j.jaci.2007.11.014]

  51. Schmidt-Supprian, M., Bloch, W., Courtois, G., Addicks, K., Israel, A., Rajewsky, K., Pasparakis, M. NEMO/IKK-gamma-deficient mice model incontinentia pigmenti. Molec. Cell 5: 981-992, 2000. [PubMed: 10911992] [Full Text: https://doi.org/10.1016/s1097-2765(00)80263-4]

  52. Siggs, O. M., Berger, M., Krebs, P., Arnold, C. N., Eidenschenk, C., Huber, C., Pirie, E., Smart, N. G., Khovananth, K., Xia, Y., McInerney, G., Hedestam, G. B. K., Nemazee, D., Beutler, B. A mutation of Ikbkg causes immune deficiency without impairing degradation of I-kappa-B-alpha. Proc. Nat. Acad. Sci. 107: 3046-3051, 2010. [PubMed: 20133626] [Full Text: https://doi.org/10.1073/pnas.0915098107]

  53. Singh, A., Zarember, K. A., Kuhns, D. B., Gallin, J. I. Impaired priming and activation of the neutrophil NADPH oxidase in patients with IRAK4 or NEMO deficiency. J. Immun. 182: 6410-6417, 2009. [PubMed: 19414794] [Full Text: https://doi.org/10.4049/jimmunol.0802512]

  54. Takada, H., Nomura, A., Ishimura, M., Ichiyama, M., Ohga, S., Hara, T. NEMO mutation as a cause of familial occurrence of Behcet's disease in female patients. Clin. Genet. 78: 575-579, 2010. [PubMed: 20412081] [Full Text: https://doi.org/10.1111/j.1399-0004.2010.01432.x]

  55. The International Incontinentia Pigmenti Consortium. Genomic rearrangement in NEMO impairs NF-kappa-B activation and is a cause of incontinentia pigmenti. Nature 405: 466-472, 2000. [PubMed: 10839543] [Full Text: https://doi.org/10.1038/35013114]

  56. The International IP Consortium. Survival of male patients with incontinentia pigmenti carrying a lethal mutation can be explained by somatic mosaicism or Klinefelter syndrome. Am. J. Hum. Genet. 69: 1210-1217, 2001. [PubMed: 11673821] [Full Text: https://doi.org/10.1086/324591]

  57. Tokunaga, F., Nakagawa, T., Nakahara, M., Saeki, Y., Taniguchi, M., Sakata, S., Tanaka, K., Nakano, H., Iwai, K. SHARPIN is a component of the NF-kappa-B-activating linear ubiquitin chain assembly complex. Nature 471: 633-636, 2011. [PubMed: 21455180] [Full Text: https://doi.org/10.1038/nature09815]

  58. Vinolo, E., Sebban, H., Chaffotte, A., Israel, A., Courtois, G., Veron, M., Agou, F. A point mutation in NEMO associated with anhidrotic ectodermal dysplasia with immunodeficiency pathology results in destabilization of the oligomer and reduces lipopolysaccharide- and tumor necrosis factor-mediated NF-kappa-B activation. J. Biol. Chem. 281: 6334-6348, 2006. [PubMed: 16379012] [Full Text: https://doi.org/10.1074/jbc.M510118200]

  59. Wu, Z.-H., Shi, Y., Tibbetts, R. S., Miyamoto, S. Molecular linkage between the kinase ATM and NF-kappaB signaling in response to genotoxic stimuli. Science 311: 1141-1146, 2006. [PubMed: 16497931] [Full Text: https://doi.org/10.1126/science.1121513]

  60. Yamaoka, S., Courtois, G., Bessia, C., Whiteside, S. T., Weil, R., Agou, F., Kirk, H. E., Kay, R. J., Israel, A. Complementation cloning of NEMO, a component of the I-kappa-B kinase complex essential for NF-kappa-B activation. Cell 93: 1231-1240, 1998. [PubMed: 9657155] [Full Text: https://doi.org/10.1016/s0092-8674(00)81466-x]

  61. Zonana, J., Elder, M. E., Schneider, L. C., Orlow, S. J., Moss, C., Golabi, M., Shapira, S. K., Farndon, P. A., Wara, D. W., Emmal, S. A., Ferguson, B. M. 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. 67: 1555-1562, 2000. [PubMed: 11047757] [Full Text: https://doi.org/10.1086/316914]

  62. Zonana, J., Ferguson, B. Reply to Kosaki et al. Am. J. Hum. Genet. 69: 665-666, 2001.


Contributors:
Cassandra L. Kniffin - updated : 05/24/2022
Cassandra L. Kniffin - updated : 06/03/2020
Alan F. Scott - updated : 02/19/2019
Matthew B. Gross - updated : 9/8/2014
George E. Tiller - updated : 8/28/2013
Patricia A. Hartz - updated : 7/11/2013
Marla J. F. O'Neill - updated : 5/18/2012
Patricia A. Hartz - updated : 4/6/2012
Paul J. Converse - updated : 3/15/2012
Ada Hamosh - updated : 7/19/2011
Paul J. Converse - updated : 2/28/2011
Paul J. Converse - updated : 8/28/2008
Ada Hamosh - updated : 4/12/2007
Victor A. McKusick - updated : 2/21/2007
Paul J. Converse - updated : 2/5/2007
George E. Tiller - updated : 1/16/2007
Marla J. F. O'Neill - updated : 6/23/2006
Ada Hamosh - updated : 4/19/2006
Victor A. McKusick - updated : 3/15/2006
Marla J. F. O'Neill - updated : 2/15/2006
Marla J. F. O'Neill - updated : 1/25/2006
Paul J. Converse - updated : 9/22/2005
Paul J. Converse - updated : 2/3/2005
Victor A. McKusick - updated : 8/23/2004
Ada Hamosh - updated : 8/27/2003
Ada Hamosh - updated : 8/26/2003
Victor A. McKusick - updated : 1/14/2003
George E. Tiller - updated : 5/14/2002
George E. Tiller - updated : 2/13/2002
Victor A. McKusick - updated : 12/20/2001
Victor A. McKusick - updated : 9/27/2001
Victor A. McKusick - updated : 3/20/2001
Paul J. Converse - updated : 3/6/2001
Carol A. Bocchini - updated : 12/20/2000
Victor A. McKusick - updated : 12/13/2000
Paul J. Converse - updated : 8/31/2000
Stylianos E. Antonarakis - updated : 8/3/2000
Ada Hamosh - updated : 5/24/2000

Creation Date:
Ada Hamosh : 5/24/2000

Edit History:
carol : 12/14/2023
carol : 06/02/2022
carol : 06/01/2022
alopez : 05/27/2022
ckniffin : 05/24/2022
carol : 06/23/2020
mgross : 06/22/2020
carol : 06/10/2020
carol : 06/09/2020
ckniffin : 06/03/2020
carol : 02/20/2019
carol : 02/19/2019
carol : 11/15/2018
carol : 11/09/2018
carol : 09/13/2017
carol : 05/19/2015
mgross : 9/8/2014
carol : 9/16/2013
tpirozzi : 8/30/2013
tpirozzi : 8/29/2013
tpirozzi : 8/28/2013
mgross : 7/16/2013
mgross : 7/11/2013
terry : 3/15/2013
carol : 5/22/2012
terry : 5/18/2012
mgross : 5/15/2012
terry : 5/1/2012
terry : 4/6/2012
mgross : 3/21/2012
terry : 3/15/2012
terry : 7/25/2011
alopez : 7/19/2011
joanna : 6/6/2011
mgross : 3/1/2011
terry : 2/28/2011
alopez : 11/18/2008
mgross : 8/28/2008
wwang : 7/3/2008
ckniffin : 6/26/2008
alopez : 4/12/2007
carol : 4/12/2007
wwang : 4/3/2007
mgross : 3/22/2007
alopez : 2/26/2007
terry : 2/21/2007
mgross : 2/5/2007
mgross : 2/5/2007
wwang : 1/23/2007
terry : 1/16/2007
terry : 11/3/2006
wwang : 6/26/2006
terry : 6/23/2006
alopez : 4/20/2006
terry : 4/19/2006
alopez : 3/20/2006
terry : 3/15/2006
wwang : 2/22/2006
terry : 2/15/2006
wwang : 2/2/2006
terry : 1/25/2006
mgross : 10/4/2005
terry : 9/22/2005
mgross : 2/3/2005
tkritzer : 9/1/2004
terry : 8/23/2004
alopez : 11/25/2003
alopez : 8/27/2003
alopez : 8/27/2003
alopez : 8/27/2003
terry : 8/26/2003
cwells : 1/15/2003
terry : 1/14/2003
cwells : 5/17/2002
cwells : 5/14/2002
carol : 4/8/2002
cwells : 2/19/2002
cwells : 2/13/2002
alopez : 2/4/2002
cwells : 1/7/2002
terry : 12/20/2001
carol : 10/5/2001
carol : 10/5/2001
mcapotos : 10/4/2001
terry : 9/27/2001
carol : 4/26/2001
alopez : 4/24/2001
alopez : 4/24/2001
cwells : 4/5/2001
cwells : 3/30/2001
terry : 3/20/2001
mgross : 3/6/2001
mgross : 3/6/2001
alopez : 3/2/2001
terry : 3/1/2001
carol : 12/20/2000
terry : 12/13/2000
alopez : 8/31/2000
mgross : 8/3/2000
mgross : 8/3/2000
alopez : 5/24/2000
alopez : 5/24/2000
alopez : 5/24/2000
alopez : 5/24/2000