Вы находитесь на странице: 1из 21

Leading Edge

Review

30 Years of NF-kB: A Blossoming


of Relevance to Human Pathobiology
Qian Zhang,1 Michael J. Lenardo,2,* and David Baltimore3
1Department of Translational Medicine, Sidra Medical and Research Center, Doha, Qatar
2Molecular Development of the Immune System Section and NIAID Clinical Genomics Program, National Institute of Allergy and Infectious
Diseases, National Institutes of Health, Bethesda, MD 20892, USA
3Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA

*Correspondence: lenardo@nih.gov
http://dx.doi.org/10.1016/j.cell.2016.12.012

NF-kB was discovered 30 years ago as a rapidly inducible transcription factor. Since that time, it
has been found to have a broad role in gene induction in diverse cellular responses, particularly
throughout the immune system. Here, we summarize elaborate regulatory pathways involving
this transcription factor and use recent discoveries in human genetic diseases to place specific
proteins within their relevant medical and biological contexts.

Thirty years ago, Ranjan Sen and one of the authors did a series from a pool of five monomers, yielding up to 15 NF-kB com-
of experiments that identified a protein binding to a specific, plexes (Smale, 2012).
conserved DNA sequence in nuclei of activated B lymphocytes The remarkable capability of NF-kB to alter a cell’s biology re-
(Sen and Baltimore, 1986). We named it for the cell type in which sults from hundreds of target genes that it activates or represses.
we identified it and the gene it affected: nuclear factor binding Even with 15 potential forms, this is a daunting task because
near the k light-chain gene in B cells, or NF-kB. Had we realized each component is needed at a characteristic concentration
that NF-kB would have a wide role in inflammation and other nat- and time following inflammatory challenge. We know now that
ural and pathological processes, we might have found a simpler there is specificity information all around the target genes, from
designation for ease of typing. which other transcription factors may be bound along with
Because NF-kB was induced during B cell maturation, we sus- NF-kB, to the specific binding site sequence and its effect on
pected that it had a major role in B cell activation and develop- the detailed structure of NF-kB, to the particular form of NF-kB
ment. This has proved true, but it greatly underestimated the that best binds to a particular site, to the intrinsic half-life of the
impact of NF-kB on biological systems (Baltimore, 2011; Hayden induced mRNA and to the rate of splicing of the induced
and Ghosh, 2008). First of all, it constitutes a paradigm of a rapid pre-mRNA (Hayden and Ghosh, 2012). This is a rich field of
response factor, one held in latency in the cell (Figure 1). When biology, not yet fully plumbed.
an inflammatory or other insult impinges on a cell, it activates a In the following presentation, we will review the detailed mo-
whole pathway of orderly responses, starting immediately after lecular biology of NF-kB-mediated signaling. Against this back-
stimulation. Once resolution of the stimulus is achieved, the ground of understanding, we will then discuss mutational evi-
pathway resets to latency. Second, not just B cells are activated; dence for NF-kB’s function in particular situations, contrasting
perhaps more importantly, cells of the innate immune system are human genetic data with mouse studies and highlighting the dif-
the first to be activated through NF-kB at a wound or site of infec- ferences between these two mammalian species. With such
tion (Ben-Neriah, 2002). These cells provide the first line of de- interesting differences, we can confidently predict that all
fense, especially to bacterial invaders. Many minor incursions species will, over time, have adapted the proteins we describe
are cleared solely by innate immunity in mammals; for lower or- to their individual biology and ecological challenges, leaving
ganisms, the innate system may be their only defense. More much detail to be uncovered.
extensive inflammation will activate B and T cells of the adaptive
immune system, again with the heavy involvement of NF-kB. Biochemistry
As time has passed, what seemed a simple induction process Hundreds of genes utilize NF-kB through variations of a
mediated by such proinflammatory molecules as TNF or lipo- nearly palindromic DNA sequence with a consensus of
polysaccharide has turned out to involve many intermediate 50 -GGGRNWYYCC-30 (N, any base; R, purine; W, adenine or
factors and processes including protein-protein dimerization, thymine; Y, pyrimidine), termed kB, found in their enhancers
phosphorylation of serine, threonine or tyrosine residues, and or promoters (http://www.bu.edu/nf-kb/gene-resources/target-
polyubiquitin modifications (Hayden and Ghosh, 2008, 2012). genes/) (Sen and Baltimore, 1986; Lenardo et al., 1987). The
Many inducers are known, each seemingly working through its site binds dimers of five proteins comprising the Rel transcription
individual pathway. Also, we have appreciated that NF-kB is factor family: p50, p52, Rel A (p65), Rel B, and c-Rel that share
not a single transcription protein but rather a family of related N-terminal homology with the v-Rel oncogene (Hayden and
protein complexes working as hetero- and homodimers, drawing Ghosh, 2008) (Figure 2A). The Rel homology domain (RHD) is

Cell 168, January 12, 2017 Published by Elsevier Inc. 37


Figure 1. The NF-kB Paradigm of Timely
and Flexible Biochemical Control of Cell
Behavior

expression, and (2) it enables the pre-


formed NF-kB transcription complexes
to be launched into action instantaneously
providing a rapid molecular switch for re-
300 amino acids and has three functions: sequence-specific sponses to pathogens or inflammatory stimuli. NF-kB also em-
DNA binding, dimerization, and inhibitory protein binding (Hay- bodies an interesting developmental paradigm at least for B cells.
den and Ghosh, 2012; Smale, 2012). The RHD contains two One conceptual problem with tissue-specific gene expres-
subdomains with beta-barrel immunoglobulin (Ig)-like folds. sion is an infinite regression paradox. If a tissue-specific tran-
The N-terminal subdomain resembles the p53 DNA-binding scription factor (TSTF) is required for a set of tissue-specific
domain and specifies DNA recognition. The C-terminal sub- genes, then does TSTF itself require a TSTF and so on? The
domain has hydrophobic residues that form dimerization and NF-kB paradigm shows that lineage and developmental tran-
inhibitory protein interfaces. The domains are joined at a hinge scriptional regulators can be induced biochemically, thereby
that clamps onto both sides of the cognate kB site across the resolving the paradox.
major groove of the DNA. Following the RHD is a nuclear locali- The first step of NF-kB activation involves post-translational
zation sequence (NLS) (Figure 2A). The five Rel family proteins modification of IkB inhibitors. This occurs by a canonical
fall into two classes. First, p50 and p52 arise from precursor pro- pathway (CP) and an alternative pathway (AP) (Hayden and
teins, p105 (NFKB1) and p100 (NFKB2), respectively, from which Ghosh, 2008). For the CP, a kinase complex called IkB kinase
a C-terminal region containing ankyrin repeats (AnkR) is post- (IKK) specifically phosphorylates IkB proteins leading to their
translationally cleaved. Second, Rel A, Rel B, and c-Rel are syn- degradation (Ben-Neriah, 2002). This causes NF-kB dimers,
thesized as mature proteins with transcription transactivation which actively shuttle between the nucleus and cytosol, to stay
domains (TADs). The two paradigmatic dimers are p50:p65 nuclear and induce gene expression. The cytosolic IKK holoen-
and p52:RelB, although other combinations including p50:p50 zyme, which we’ll call ‘‘classical’’ IKK, contains a regulatory sub-
and p52:p52 homodimers or p52:RELA or p50:RELB hetero- unit, ‘‘NF-kB essential modifier’’ (NEMO, also called IKKg/Fip-3/
dimers exist with distinct functions (Smale, 2012). All in all, IKKAP), and two kinase subunits, IKKa and IKKb (Figure 2D).
13 of the 15 potential NF-kB family complexes have been NEMO is a non-catalytic subunit that tethers IKKa and IKKb
demonstrated, the other two being undescribed and possibly into a regulatory holocomplex and is required for ubiquitination
unfavored (S. Ghosh, personal communication). TAD-containing reactions that beget protein oligomerization and signaling (Hay-
heterodimers are transcriptional activators, whereas p50 or p52 den and Ghosh, 2008). Activation involves signaling assemblies
homodimers are repressors unless bound to secondary proteins. connecting upstream signal generation apparatuses to IKK using
Since dimer combinations may recognize distinct variants of the ubiquitin modifications as the mortar. The TNF receptor (TNFR)
kB site with different affinities, many gene regulatory patterns induces a well-understood IKK induction process. TNF stimula-
can be generated from this handful of proteins. tion of TNFR recruits the RIP-1, TRADD, TRAF2, cIAP, TAB, and
In unstimulated cells, such as ‘‘resting’’ lymphocytes before TAK1 proteins into a megacomplex with the linear ubiquitin as-
antigen encounter, NF-kBs are mainly cytoplasmic due to the sembly complex (LUBAC) stabilized by linear and K63-linked
binding of a dedicated set of inhibitory proteins comprising the polyubiquitin chains (Dondelinger et al., 2016). Then IKK is incor-
‘‘Inhibitor of kB’’ (IkB) family (Hayden and Ghosh, 2008). The intri- porated, causing ubiquitination of NEMO and phosphorylation of
cate biochemical pathways controlling NF-kB/inhibitor com- IKK that induce its kinase activity (Hayden and Ghosh, 2008). The
plexes have been reviewed authoritatively, and we will provide composition of the megacomplex depends on specific ubiquitin
only a brief summary (Hayden and Ghosh, 2012). The inhibitors, modifications. This, in turn, determines whether TNFR engage-
IkBa, IkBb, IkBε, BCL-3, IkBz, IkBNS, and the C-terminal por- ment causes NF-kB induction and survival or an alternative
tions of the precursor proteins p105 (IkBg) and p100 (IkBd), all fate in which recruitment of RIP kinases 1 and 3 induce cell death
contain five to seven tandem ankyrin repeats (AnkRs) instead of NF-kB (Dondelinger et al., 2016). The RIP protein fam-
(Figure 2B). AnkRs are 33 amino acid ankyrin-like protein-protein ily, RIPs 1–7, are adaptors and kinases that transmit signals from
association domains that extend as helices capable of binding to a wide variety of surface receptors and intracellular stress sen-
NF-kB covering the NLS. The C-terminal AnkR-containing, sors to transcriptional mediators including Jnk, Erk, and p38
IkB-like regions of p100 and p105 undergo limited proteolysis and NF-kB as well as mediators of necrotic death (Meylan and
to make p52 and p50 which transit to the nucleus. For p50, Tschopp, 2005). Generally, these transduction complexes can
this occurs constitutively, but p52 is only generated after specific send pleiotropic parallel signals besides NF-kB to coordinate
signaling events. The canonical p50/p65 heterodimer is regu- necessary responses.
lated principally by IkBa (Figure 2C). Biochemical control of nu- B and T lymphocytes induce NF-kB in adaptive immune re-
clear localization via IkB has two strategic consequences: (1) it sponses through the CARD11:Bcl10:MALT1 (CBM) complex
becomes a principal control point for NF-kB-induced gene (Hayden and Ghosh, 2008). Newly expressed genes promote

38 Cell 168, January 12, 2017


Figure 2. Components of the NF-kB System
(A) Rel homology domain proteins in the NF-kB protein family and the v-Rel oncoprotein. The N-terminal regions of REL family proteins share the Rel homology
domain (RHD), which has amino acid similarity to the product of the v-rel oncogene from the Reticuloendotheliosis virus REV-T and a nuclear localization
sequence (NLS). RelB has a leucine zipper (LZ), and RelA, RelB, and c-Rel harbor a transactivation domain (TAD). p105 and p100 have five to seven tandem
ankyrin repeats (AnkR) and a death domain (DD). Regulatory phosphorylation sites (P) and ubiquitination sites (Ub) are shown.
(B) Inhibitors of NF-kB (IkB) family of proteins. The eight described IkBs are characterized by their five to seven tandem ankyrin repeats (AnkR), which mediate
binding to NF-kB dimers. The N-terminal regions of the classical IkBs (IkBa, IkBb, IkBε) contain two serine residues (P), which permit the accelerated ubiq-
uitination (Ub) and degradation of the protein when phosphorylated. BCL3 has similar modification residues but IkBz and IkBNS do not. The C-terminal regions of
p105 and p100 function as IkBs for their linked RHD even before processing by forming large complexes with Rel proteins, including p50 and p52. PEST, region
rich in the amino acids proline, glutamic acid, serine, and threonine; DD, death domain.
(C) Structure of the IkB:NF-kB latent complex (PBD 1NFI). Ribbon diagram of the crystal structure of the isolated complex including IkBa (blue), showing its
anchor and repeat domain (ARD) and PEST sequence rich in the amino acids proline, glutamic acid, serine, and threonine that acts as a signal for protein
degradation, p50 (red), and the RelA (green), inter-domain linker, and N-terminal domain (NTD).
(D) IKK complex. Shown are the components of the IKK complex including two kinases: IKKa, IKKb, and the non-enzymatic subunit NEMO. CC1, coiled-coil
domain 1; CC2, coiled-coil domain 2; LZ, leucine zipper; Zn, zinc-finger; Kinase, kinase domain; HLH, helix-loop-helix region; NBD, NEMO-binding domain.
Regulatory phosphorylation sites (P) and ubiquitination sites (Ub) are shown. The number of amino acids (aa) of each protein is shown at right. Adapted from
Hayden and Ghosh (2008, 2012).

Cell 168, January 12, 2017 39


lymphocyte proliferation and specific immune functions recapture DNA-bound NF-kB and halt transcription (Hayden
including antibody production by B cells and the generation of and Ghosh, 2008). Moreover, NF-kB induces the IkBa gene
cytokines and other anti-pathogen responses by T cells. Abnor- establishing a cybernetic negative feedback (Hayden and
malities of immune activation can foster immunodeficiency, Ghosh, 2012).
autoimmune diseases, or lymphoid malignancies. The CBM The physiological role of NF-kB is best delineated in the immune
complex is the shared central conduit of T cell receptor (TCR) system. Knockout mice for the RHD proteins show predominantly
and B cell receptor (BCR) signaling essential for NF-kB (Blonska immunological defects (Gerondakis et al., 2006). Also, a cornu-
and Lin, 2011; Hayden and Ghosh, 2012). After receptor engage- copia of human genetic disorders affecting NF-kB show promi-
ment, CARD11 (CARMA1) is recruited and phosphorylated by nent immune pathology (Courtois et al., 2016). Correspondingly,
PKC-q in T cells and PKC-b in B cells. In T cells, CARD11 nucle- CP inducers, mainly p50:RelA dimers, include all sorts of pro-in-
ates the tripartite CBM on the inner leaflet of the membrane by flammatory cytokines such as tumor necrosis factor (TNF) and
attracting BCL10 and the paracaspase MALT1 (Hayden and interleukin-1 (IL-1), conventional humoral and cellular antigens,
Ghosh, 2012). Caspase-8 is recruited as a full-length, unpro- pathogen-associated molecular patterns (PAMPs) including
cessed, but weakly active form and ropes in IKK to the many bacterial and viral products, cell-bound and soluble immune
CBM. MALT1-dependent tethering of TRAF6 stimulates poly- mediators, and effector molecules. These potently trigger NF-kB
ubiquitination of NEMO and the phosphorylation of BCL10, in lymphocytes, macrophages, and dendritic cells and other minor
IKKa, and IKKb. Then, casein kinase 1a brings IKKb into the immune cell types (Hayden and Ghosh, 2008, 2012). Inducing
signaling complex and phosphorylates CARD11 to temper NF-kB turns on thousands of genes that re-sculpt the transcrip-
further signaling (Bidère et al., 2009). CARD9, CARD10 tional program of immune cells from quiescence to a proliferating
(CARMA3), and CARD14 (CARMA2) can form analogous effector phenotype capable of fighting pathogens. The NF-kB-
biochemical complexes to activate NF-kB during specialized induced genes include nearly the entire armamentarium of im-
immunity (Blonska and Lin, 2011). Which specific CARD protein mune guardians: chemokines, cytokines, adhesion molecules,
depends on the receptor and downstream effect. The BCL10/ inflammatory mediators, and apoptosis inhibitors, giving NF-kB
MALT1 dimer is shared by all CARDs. In addition to NF-kB, a pivotal role in global immunity (Lenardo et al., 1989; http://
different CBM complexes can stimulate c-Jun N-terminal kinase www.bu.edu/nf-kb/gene-resources/targetgenes/).
(JNK), mechanistic target of rapamycin (mTOR), and other signal The AP of NF-kB activation is not regulated by classical IKK or
pathways (Blonska and Lin, 2011). However, lymphocytes IkBs and therefore doesn’t respond to CP inducers (Nishikori,
require NF-kB. For B lymphocytes, BAFF/BAFF receptor 2005). Whereas the CP responds rapidly to immediate infectious
(BAFFR) causes maturation and survival through NF-kB (Geron- threats, the AP has slow and sustained kinetics dependent on
dakis and Siebenlist, 2010; Mackay and Cancro, 2006). For new protein synthesis consistent with a role in organogenesis
T lymphocytes, many cytokines and their receptors—ones that (Baltimore, 2011; Hayden and Ghosh, 2008). Many inducers
control and are controlled by NF-kB—govern proliferation, are a subgroup of the TNFR superfamily, perhaps any receptor
specialization into functional subsets, and survival (Gerondakis that signals through TRAFs, including BAFFR, CD40, RANK,
and Siebenlist, 2010). 4-1BB, HVEM, OX40, GITR, Fn14, TNFR2, and CD30, each of
The rapid and irreversible demolition of IkB proteins in the which likely relates to a different function of the AP (Gerondakis
proteasome—the toggle for NF-kB—is triggered by phosphory- and Siebenlist, 2010; Mackay and Cancro, 2006). For example,
lation and ubiquitination. In the CP, IKKb phosphorylates IkBa B cell-activating factor (BAFF) drives peripheral B cell differ-
on Ser 32 and Ser 36, or IkBb on Ser 19 and Ser 23. Phosphor- entiation and survival (Mackay and Cancro, 2006). CD40 is
ylated IkBs are then polyubiquitinated by SCFbTrCP E3 ubiquitin expressed broadly on immune cells and uses both the AP and
ligases, the signal for proteasome deposition (Ben-Neriah, CP to control B cell activation, maturation, germinal center for-
2002). Mutation of any of these serines will block IkB degradation mation, somatic mutation, and class switching. Lymphotoxin
and prevent signaling. By contrast, the AP utilizes a dimer of (LT) and LIGHT trigger the LTb receptor (LTbR) to mediate devel-
IKKa to phosphorylate p100 on serines 176 and 180 causing pro- opment of peripheral lymph nodes and Peyer’s patches (Sun,
teasomal processing to p52 (Hayden and Ghosh, 2008, 2012). 2011). Finally, RANK regulates bone formation and dendritic
p100 is mainly complexed with RelB with its AnkR domain as cell functions through AP signals (Hayden and Ghosh, 2008).
an IkB (sometimes called IkBd) to retain nascent p52 and RelB How each of these distinct responses is conveyed by the
in the cytoplasm. Phosphorylation of the p100 AnkR domain is p52:RelB dimer is unknown. Specific microRNAs, miR-223,
independent of IKKb, IKKg, or the classical IKK complex. miR-15a, and miR-16 are negative regulators of IKKa, showing
Instead, it requires the NF-kB-inducing kinase (NIK). NIK is un- that there exist even more sophisticated regulatory mechanisms
stable and a TRAF complex controls its ubquitination and pro- for gene control (Ma et al., 2011).
teasomal turnover. Receptors induce the AP by pulling away As these 30 years have gone on, the field has witnessed dis-
the TRAF complex, thereby stabilizing NIK so it can phosphory- covery of an increasing diversity of activators, signaling compo-
late IKKa. This causes IKKa to specifically phosphorylate the nents, and responding genes for the NF-kB system that are
cytoplasmic p100:RelB complex on serines 866 and 870 causing updated continuously on the web (http://www.bu.edu/nf-kb/
ubiquitination and proteolysis of the C-terminal AnkR domain gene-resources/target-genes/). The central question persists:
and nuclear ingress of p52:RelB (Sun, 2011). Transcription by how this fairly simple regulatory system can respond to an enor-
NF-kB is also enhanced by Rel protein phosphorylation (Hayden mous group of inducers and faithfully transpose those into
and Ghosh, 2008). Also, IkBa and IkBε can go to the nucleus and appropriate patterns of gene expression in different tissues.

40 Cell 168, January 12, 2017


Figure 3. The Major Monogenic Diseases in the Core Pathways of Both the Classical and Alternative Pathways of NF-kB Signaling
Color-coded boxes indicate the main feature of diseases. CP, classical; AP, alternative.

A relatively new approach is to examine genetic defects affecting in approximately 65% of IP patients. Only heterozygous female
NF-kB in humans. Clinicians provide an extraordinarily detailed survive. The prominent skin lesions suggest a vital role of
phenotype that can now be combined with whole-genome anal- NEMO and NF-kB in skin cells. Due to X inactivation/lyonization,
ysis together with exhaustive systems analysis using transcrip- female heterozygotes have half of their skin cells with normal
tomics and proteomics. We now review the human genetic NEMO expression and the other half deficient. The defective
data and compare them with our current understanding from half die quickly after birth and leave behind damaged skin and
biochemistry and mouse data. skewed X inactivation in the residual keratinocytes. Skewed
X inactivation in leukocytes also implies that NEMO is essential
Human Genetics for their survival.
There are human genetic diseases affecting a large number of Hypomorphic NEMO Mutations. Milder disease is caused by
NF-kB components, and these are summarized in Figure 3 and hypomorphic NEMO mutations manifesting as anhidrotic ecto-
Table 1. dermal dysplasia with immunodeficiency (EDA-ID) (Döffinger
IkB Regulatory Complex et al., 2001). EDA-ID studies also revealed a requirement for
NEMO. NEMO/IKBKG mutations were the first genetic defects NF-kB in keratinocyte proliferation and differentiation. A paradox
discovered in the NF-kB pathway and showed unique pheno- of NEMO deficiency is that the B, T, and natural killer (NK) sub-
types caused by different mutations. Because IKBKG is on the sets develop normally in most EDA-ID patients, but Ig production
X chromosome, disease was more severe in males. Complete is often low and severe gram-positive cocci, gram-negative
loss-of-function (LOF), i.e., null or amorphic, mutations cause bacilli, or mycobacterial infections lead to high mortality at an
incontinentia pigmenti (IP) in females and prenatal lethality in early age. Studies in patient’s cells showed that defective
males except when the latter show mosaicism or an XXY karyo- NEMO impairs the CP response to many immune mediators
type. Thus, it is an essential gene. Hypomorphic mutations cause including IL-1b, IL-18, TNF, or lipopolysaccharide (LPS) (Döffin-
a different skin disease, anhidrotic ectodermal dysplasia with im- ger et al., 2001; Hubeau et al., 2011). TNF signaling stimulates
munodeficiency (EDA-ID) in males, and mild IP in females. The both apoptosis and activation pathways. Achieving the appro-
severity of phenotypes dictates that most mutations are de novo. priate immune response requires the NF-kB induction of
Amorphic NEMO Mutations. IP patients have skin inflamma- apoptosis inhibitory genes (Karin and Lin, 2002). Deficiencies
tion combined with varying abnormalities of hair, nails, teeth, of NEMO and other pathway components that disrupt NF-kB
retinal vascularization, and the CNS. The disease was clinically cause TNF-induced apoptosis to predominate, which impairs
recognized in the 1920s and genetically linked to Xq28 in the immunity. Interestingly, NEMO-deficient patient cells also re-
late 1980s (Wieacker et al., 1985). In 2000, only 2 years after it sponded poorly to CD40 ligand (CD40L/CD154) that activates
was cloned and mapped to the same region, NEMO was identi- both CP and AP through multiple TRAFs, possibly predisposing
fied as the causal gene of IP (Smahi et al., 2000). All IP mutations to mycobacterial infections. The NEMO-dependent CP rapidly
are amorphic. A deletion of exons 4 through 10 of NEMO occurs activates NF-kB, as an immediate response to inflammatory

Cell 168, January 12, 2017 41


42 Cell 168, January 12, 2017

Table 1. Features of Monogenic Diseases in the Core NF-kB Pathways in Order of Discovery
Year of Protein Method of
Discovery Name Gene Name Discovery Type of Disorder Inheritance Model Defects in Patients Defects in Mouse Models Reference
2000 CYLD CYLD linkage study familial AD cell hyperproliferation T cell developmental defect, Bignell et al. (2000);
cylindromatosis susceptibility to induced Massoumi et al. (2006);
colonic inflammation and Zhang et al. (2006)
increased incidence of tumors
2000 NEMO IKBKG candidate familial incontinentia XL female failures of NF-kB induction in dermatopathy similar to the Schmidt-Supprian
gene pigmenti (IP) integument and ectoderm- human X-linked disorder et al. (2000); Smahi
sequencing derived appendages incontinentia pigmenti et al. (2000)
(female mouse)
2001 NEMO IKBKG candidate anhidrotic XL male impaired CP responses to reduced number of CD8+ but Döffinger et al. (2001);
gene ectodermal dysplasia many inducers including IL- not CD4+ thymocytes; Gerondakis et al. (2006)
sequencing with 1b, IL-18, TNF, or LPS; complete absence of
immunodeficiency normally developed T, B, and peripheral T cells (T linage
(EDA-ID) NK cells conditional ko)
2002 Caspase-8 CASP8 candidate autoimmune AR defective CD95/FAS- embryonic lethal; lymphopenia Chun et al. (2002);
gene lymphoproliferative induced apoptosis and TCR and impaired responses to Salmena et al. (2003)
sequencing syndrome with signaling antigen (T cell specific casp8
immunodeficiency deficiency)
2003 IkBa NFKBIA biochemistry anhidrotic AD poor T cell responses to similar EDA phenotype plus Courtois et al. (2003);
ectodermal dysplasia antigen stimuli, poor lacking lymph nodes, Peyer’s Mooster et al. (2015)
with responses to TNF patches, splenic marginal
immunodeficiency and CD40L zones, and germinal centers
(EDA-ID) and exhibit significantly
decreased responses to TLRs,
TNF, LTbR, and BAFF
2009 CARD9 CARD9 linkage study chronic AR fungal infections, especially impaired NF-kB signals from Blonska and Lin (2011);
mucocutaneous rare deep dermatophytoses, Dectin-1 and Dectin-2 Glocker et al. (2009);
candidiasis; deep blunted GM-CSF responses, Lanternier et al. (2013)
dermatophytosis decreased Th17 cells
2010 IKKa IKBKA microarray severe fetal AR embryonic lethal die at birth, skeletal and Gerondakis et al.
encasement epidermal defects (2006); Lahtela
malformation et al. (2010)
2012 HOIL-1 HOIL1 NGS immunodeficiency, AR NEMO ubiquitination in spontaneous amylopectin-like Boisson et al. (2012);
autoinflammation and response to TNF or IL-1 deposits in the myocardium, MacDuff et al. (2015)
amylopectinosis susceptibility to Listeria
monocytogenes, Toxoplasma
gondii, and Citrobacter
rodentium infections, but not
spontaneous autoinflammation
2012 CARD14 CARD14 linkage study psoriasis AD increased expression and N/A Fuchs-Telem et al.
hyperactivation of NF-kB in (2012); Jordan
endothelial cells et al. (2012)
(Continued on next page)
Table 1. Continued
Year of Protein Method of
Discovery Name Gene Name Discovery Type of Disorder Inheritance Model Defects in Patients Defects in Mouse Models Reference
2012 CARD11 CARD11 RNA-seq congenital B cell AD (GOF) B cell lymphocytosis, hyper- N/A Snow et al. (2012)
lymphocytosis responsiveness of B cells to
BCR stimulation, CD40L,
and BAFF, spontaneously
aggregated CARD11 and
NF-kB activation in T cells
2013 CARD11 CARD11 NGS severe combined AR (LOF) absent degradation of IkBa defective TCR, BCR, CD40 Egawa et al. (2003);
immunodeficiency or phosphorylation of p65 in signaling, blockage of Treg Greil et al. (2013); Hara
(SCID) response to TCR or BCR development et al. (2003); Stepensky
stimulation et al. (2013)
2013 IKKb IKBKB linkage study severe combined AR defective T and B cell embryonic lethal, TNF Pannicke et al. (2013);
immunodeficiency activation/proliferation, induced hepatocyte apoptosis, Pasparakis et al. (2002);
(SCID) reduced B cell response liver degeneration, lung Tanaka et al. (1999)
to CD40L deformation, skin inflammation
2013 P100/52 NFKB2 NGS common variable AD poor antibody responses, defective secondary lymphoid Chen et al. (2013);
immune deficiency predominantly unswitched organ development, impaired Gerondakis et al. (2006)
(CVID) with adrenal naive B cells in the periphery; B cell development; enhanced
insufficiency abrogated processing of DC function
p100 to p52 in patient cells
before or after CD40-CD40L
signaling
2013 MALT1 MALT1 linkage study combined AR impaired NF-kB responses similar to human without Treg Jabara et al. (2013);
+ NGS immunodeficiency to PMA + ionomycin, PHA, disfunction Ruefli-Brasse
(CID) + inflammatory and anti-CD3 stimulation, et al. (2003)
cell infiltration severely decreased Treg
counts
2014 BCL10 BCL10 NGS combined AR recurrent respiratory viral defective B cell maturation, Torres et al. (2014); Xue
immunodeficiency infections, and oral decreased marginal zone B et al. (2003)
(CID) + autoimmunity candidiasis; normal cells, susceptibility to blood-
lymphocyte counts with borne bacteria
overwhelmingly naive
Cell 168, January 12, 2017 43

phenotypes and
undetectable Tregs;
impaired TLR signaling
2014 NIK MAP3K14 linkage study combined AR defective maturation and severe structural defects in Willmann et al. (2014);
+ NGS immunodeficiency functions of T, B, and lymph nodes, Peyer’s patches, Yin et al. (2001)
(CID) NK cells splenic and thymic structures
found in the latter
(Continued on next page)
44 Cell 168, January 12, 2017

Table 1. Continued
Year of Protein Method of
Discovery Name Gene Name Discovery Type of Disorder Inheritance Model Defects in Patients Defects in Mouse Models Reference
2015 P105/50 NFKB1 NGS common variable haploinsufficiency? B cell dysfunctions no phenotype in heterozygous Fliegauf et al. (2015);
immune mouse; homozygous mouse Sha et al. (1995)
deficiency (CVID) showed B cell dysfunction
2015 RelB RELB linkage study combined AR increased p65 nuclear inflammatory phenotype and Gerondakis et al.
+ NGS immunodeficiency translocation and hematopoietic abnormalities (2006); Sharfe
(CID) hyperactivation in response et al. (2015)
to TCR stimulation,
decreased c-REL nuclear
translocation after CD40
stimulation in B cells
2015 HOIP HOIP linkage study immunodeficiency, AR absent response to CD40L, embryonic lethal Boisson et al. (2015);
+ NGS autoinflammation and impaired responses to IL-1b Peltzer et al. (2014)
amylopectinosis and TNF-a
2016 A20 TNFAIP3 NGS Behcet-like haploinsufficiency increased K63-ubiquitinated spontaneous NLRP3 Vande Walle et al.
autoimmunity NEMO, increased inflammasome activity to LPS (2014); Zhou
phosphorylation of IkBa and et al. (2016)
IKKs, and prolonged nuclear
residence of NF-kB;
increased pro IL-1b, NLRP3
and activated caspase-1 and
increased production of
mature IL-1b after LPS
stimulation
2016 OTULIN OTULIN NGS inflammatory AR persisting IKK activity and embryonic lethal Damgaard et al. (2016)
syndromes IkBa phosphorylation in
response to TNF stimulation
stemming from a lack of
NEMO deubiquitination
AR, autosomal recessive; AD, autosomal dominant; XL, X-linked; GOF, gain of function; LOF, loss of function; NGS, next-generation exome sequencing.
stimuli. Conversely, the NEMO-independent AP acts slowly and that are catalytically inactive and fail to elicit NF-kB (Kalay et al.,
continuously to promote cell maturation and differentiation. 2012). Thus, IKKa mutations and RIP4 mutations likely affect
Thus, dendritic cells (DCs) from NEMO-deficient patients ex- NF-kB, but the prominent skin manifestations no doubt also
hibited poor production of IL-12 in response to CD40L, while pe- reflect effects on non-NF-kB pathways. For example, IKKa defi-
ripheral B cell differentiation dependent on CD40L was relatively ciency also potentially reveals NF-kB-independent functions.
normal (Filipe-Santos et al., 2006). Moreover, CD40L activates IKKa can translocate to the nucleus and phosphorylate histone
the MAPK pathway in a NEMO- and ubiquitination-dependent H3. It also acts on chromatin independently of its kinase activity
fashion, revealing NF-kB-independent functions for NEMO altering the expression of specific genes, for example 14-3-3d,
(Elgueta et al., 2009). which influences the G2/M checkpoint. Also, both IKKa and
NEMO mutations found in EDA-ID are heterozygous and hypo- IKKb regulate b-catenin stability in the Wnt pathway. Moreover,
morphic and have varying phenotypes due to effects on different nuclear IKKa induces cell-cycle arrest and terminal differentia-
protein domains as well as non-coding regions involved in tion by regulating Smad2/3 target genes (Liu et al., 2012).
splicing and mRNA stability. There are two broad classes of dis- IKKb/IKK2/IKBKB. IKKb-deficient patients appear normal at
ease: EDA-ID or immunodeficiency without EDA. Mutations birth but develop SCID. This is strikingly different than knockout
causing decreased protein expression, folding, or stability, mice, which die as embryos due to liver degeneration, lung
such as mutations in the 50 untranslated region or exon 1B, usu- deformation, skin inflammation, and many other dysmorphic
ally cause immunodeficiency without EDA; the A288G mutation, features. IKKb-deficient patients lack signs of developmental
which affects protein stability, leads to EDA-ID. By contrast, mu- or gene regulatory defects in other organs (Gerondakis et al.,
tations that do not affect protein levels, but abolish one of the 2006). This mouse-human discordance indicates a true differ-
ubiquitin binding sites, lead to EDA-ID (e.g., D306N, D311G, ence in physiological roles because human fibroblasts and pe-
D311N, C417R) (Hubeau et al., 2011). Thus, abnormal ubiquitina- ripheral blood cells from patients had no detectable IKKb protein
tion patterns may cause aberrant signaling complexes that affect equivalent to a mouse ‘‘knockout.’’ Curiously, patient cells also
keratinocyte differentiation in ways that go beyond a simple lack had reduced NEMO and IKKa protein levels despite normal
of NEMO protein. mRNA levels (Mousallem et al., 2014; Pannicke et al., 2013), sug-
IKKa/IKK1/IKBKA. The two enzymatic components of the IKK gesting that IKKb confers stability on all three proteins in the
complex, IKKa and IKKb, share 50% overall amino acid classical IKK complex in humans. This was not seen in ikkb-de-
sequence identity and 65% kinase domain identity but cause ficient mice (Tanaka et al., 1999).
drastically different human diseases, neither of which resembles IKKb-deficient patients reveal the biological differences be-
NEMO deficiency (Hayden and Ghosh, 2008). IKKa deficiency tween IKKa and IKKb. Normally, purified classical IKK contains
leads to lethal fetal malformations, whereas IKKb deficiency NEMO:IKKa:IKKb in a 2:1:1 stoichiometry, although an alternate
causes severe combined immunodeficiency (SCID) (Lahtela IKK composed of NEMO and IKKb homodimers occurs in T cells
et al., 2010; Pannicke et al., 2013). (Hayden and Ghosh, 2008). IKKa homodimers complexed with
Autosomal recessive LOF mutations of IKKa lead to fetal NEMO can phosphorylate IkB in vitro, although it is uncertain
encasement malformation (also called Cocoon syndrome whether they exist in vivo. In IKKb-deficient cells, however, IKK
because of the appearance of the fetuses), the most severe incorporates IKKa homodimers, which alters responses to
and fatal phenotype of any NF-kB gene deficiency. IKKa defi- certain stimuli. Phosphorylation and degradation of IkBa and
ciency was confirmed by gene expression profiling in aborted fe- IL-6 induction were absent to TLR5 stimulation by flagellin, dras-
tuses. The severe phenotype of IKKa deficiency is surprising tically reduced in response to TNF stimulation, and only margin-
because in vitro binding assays show IKKb has higher affinity ally affected in response to IL-1b (Pannicke et al., 2013). Thus,
for NEMO than IKKa and IKKb can form homodimers and bind IKKa homodimers can mediate CP signaling for certain NF-kB-
to NEMO without IKKa. So, in theory, IKKa deficiency should activating receptors, perhaps because IKKa associates more
impair the IKK complex less than IKKb deficiency. It is surprising weakly with NEMO than IKKb. Receptor selectivity has also
that IKKb cannot compensate for the lack of IKKa. However, been confirmed by IKKb knockdowns in human fibroblasts.
consistent with the in vitro assays, ikka knockout mouse died Hence, IKKa and IKKb homodimers might serve different recep-
soon after birth, a slightly milder phenotype than the embryonic tors. Moreover, the full response to TNF was reconstituted by
lethal phenotype of the ikkb knockout mouse (Gerondakis et al., restoring IKKb in patient cells, confirming that IKKb has unique
2006). Even more confounding, mice with an unactivatable functions not compensated by IKKa (Pannicke et al., 2013).
mutant but not complete loss of ikka are viable. Although NF-kB was first discovered for k light-chain expres-
Interestingly, LOF mutations of receptor-interacting protein ki- sion, it has proved indispensable for many lymphocyte responses
nase 4 (RIP4) cause the same but less severe spectrum of (Hayden and Ghosh, 2012). Patients with IKKb deficiency had
craniofacial, skin, and limb deformities as IKKa LOF mutations, normal T and B cell development, but the activation and prolifer-
in a disease called Bartsocas-Papas syndrome or Popliteal ation of mature T and B cells were severely impaired, fostering a
Pterygium syndrome (Kalay et al., 2012). RIP4 is an adaptor for SCID phenotype (Mousallem et al., 2014). Normal activation and
upstream signaling cassettes to the IKK complex with the partic- differentiation into effector or memory cells are blocked and
ipation of TRAF proteins (Hayden and Ghosh, 2008). Among the mostly naive lymphocytes remain. The activation block can be
seven human RIPs, each of which signals to distinct gene induc- partly overcome by stronger stimulation with phorbol myristate
tion programs, RIP4 regulates keratinocyte differentiation and acetate (PMA) or phytohemagglutinin (PHA). Patient B cells also
wound healing through NF-kB. RIP4 mutations produce proteins showed reduced responses to CD40L stimulation with IL-21.

Cell 168, January 12, 2017 45


IkB Family (IkBa)/NFKBIA. IkBa (NFKBIA) is the prototype of (reflecting deficient Ig isotype class-switching), Epstein-Barr
the IkB family and remains the only one identified in a human virus (EBV)-associated lymphoproliferation, low peripheral
monogenic disease (Hayden and Ghosh, 2008). All human B cell counts, and, occasionally, other symptoms such as gran-
IkBa mutations are heterozygous gain-of-function (GOF) muta- ulomatous lung disease, neutropenia, and idiopathic thrombo-
tions that augment NF-kB inhibition, causing autosomal domi- cytopenic purpura (ITP). The disease has variable expressivity,
nant disease that blends the clinical features of NEMO and a feature commonly observed in haploinsufficiency conditions.
IKKb deficiency (Courtois et al., 2003; Schimke et al., 2013). For example, the father of a young woman with severe respira-
As in hypomorphic NEMO mutations, EDA is observed. Like tory infections and EBV-lymphoproliferative disease had the
IKKb deficiency, peripheral T cells respond poorly to antigen same mutation but only mild respiratory symptoms not requiring
(Courtois et al., 2003). TNF or CD40L evoke little nuclear trans- medical attention (Boztug et al., 2016). In large kindreds, hetero-
location of NF-kB. Thus, the GOF mutations in IkBa have a zygous individuals exhibited widely varying antibody responses,
dominant-negative effect. A mouse model was generated infection, lymphoma, or inflammatory bowel diseases (Fliegauf
recently to mimic the heterozygous human S32I IkBa mutation. et al., 2015). In haploinsufficient immunological diseases,
Heterozygous mice develop EDA comparable to human pa- whether this variation in clinical phenotype is due to independent
tients but also lack lymph nodes, Peyer’s patches, splenic mar- genetic variants or environmental influences remains a fasci-
ginal zones, and germinal centers and exhibit significantly nating question.
decreased responses to TLRs, TNF, LTbR, and BAFF. Thus, NFKB1 mutations cause partial reduction, i.e., haploinsuffi-
mice, unlike humans, show impairment of both the CP and ciency, of both p105 and p50 in the resting state and severely
AP (Mooster et al., 2015). reduced p105 phosphorylation after stimulation (Boztug et al.,
Most dominant interfering NFKBIA mutations are either substi- 2016; Fliegauf et al., 2015). Several multiplex kindreds with
tutions affecting the phosphorylation of residues Ser32 and splicing or frameshift mutations in NFKB1 caused haploinsuffi-
Ser36 or N-terminal protein truncations removing these serines. ciency in a dominant inheritance pattern due to reduced levels
The mutant IkBa proteins are expressed but cannot undergo of p105 and p50 protein indicating that the WT allele cannot
phosphorylation-driven degradation. Disregarding induction sig- ramp up expression to compensate for the mutant allele.
nals, they remain constitutively bound to NF-kB and freeze the Considering that varied RHD dimers exist in lymphocytes,
pathway. This has been elegantly verified by overexpressing changes in p105 or p50 levels might alter the balance of other di-
mutant IkBa proteins encoded by patient alleles and showing mers causing surprising outcomes. For example, in the family
that they override the wild-type protein and block NF-kB- mentioned above, the father with less severe disease had sub-
induced gene expression (Courtois et al., 2003). Certain trunca- stantially greater levels of p50 and p105 protein than the
tion mutations have variable blocking effects perhaps due to dif- daughter for undiscovered reasons (Boztug et al., 2016). A new
ferential protein stability or the efficiency of utilizing alternative study of Finnish families with heterozygous LOF mutations in
ATG start sites downstream of premature mutant stop codons. NFKB1 revealed variants affecting protein stability, subunit
For example, a very early truncating mutation, E14X, showed phosphorylation, or nuclear translocation and causing a wide
strong dominant inhibition due to a very efficient internal ATG range of dominant immune disorders (M. Seppänen, personal
that facilitated high mutant protein expression (Lopez-Granados communication).
et al., 2008). However, another heterozygous early truncating Completely different clinical phenotypes are found with
mutation, W11X, showed no dominant-negative effect. The genetic alterations of the different RHD proteins. A heterozy-
IkBa protein from the wild-type allele was normally phosphory- gous de novo nonsense mutation in RELA/p65 is associated
lated and degraded so that there was only haploinsufficiency with lymphadenopathy and idiopathic thrombocytopenic pur-
of NF-kB, resulting in a milder disease phenotype (McDonald pura. The mutation results in roughly one-half of the normal
et al., 2007). Further studies might reveal how the kinetics of protein expression, but how haploinsufficiency leads to
IkBa degradation, resynthesis, and accumulation of protein abnormal lymphocyte homeostasis and autoimmunity is still
from the wild-type (WT) allele is affected by the presence of a under investigation (W. Comrie, A. Faroqi, and M.J.L., unpub-
mutant protein. Recently, a homozygous IkBNS/NFKBID muta- lished data).
tion has been found in a patient with combined immunodefi- CARD-BCL10-MALT1 Signalosome: MALT1, BCL10, CARD9,
ciency, and marked B and CD8+ T cell lymphopenia (V. Bryant, CARD11, and CARD14. The CBM complex is affected by
personal communication). There are no disease associations many genetic defects causing immune dysregulation and malig-
yet for IkBb or IkBε. nancy. Interestingly, BCL10 and MALT1 deficiencies are similar
Rel Homology Domain Protein Family. Mutations in three of the since they generally work together, whereas the clinical disor-
five RHD protein genes: p50 (NFKB1), p65 (RELA), and c-Rel ders caused by deficiencies with CARD9, 11, and 14 are different
(REL), have been chiefly associated with humoral immunodefi- because each plays a different role in immunity.
ciency. The p52 and RelB components of the AP will be dis- MALT1. MALT1 (MALT lymphoma translocation protein 1) was
cussed further below. Heterozygous NFKB1 (p105/p50) gene first discovered in mucosa-associated lymphoid tissue (MALT)
mutations cause common variable immunodeficiency (CVID) lymphoma in 1999 (Dierlamm et al., 1999), in which a chromo-
(Fliegauf et al., 2015). CVID is milder than SCID, prevails in older some translocation spawns a cIAP2-MALT1 fusion gene that
patients, and manifests as defective antibody responses. Pa- constitutively activates NF-kB. Members of three families with
tients with NFKB1 mutations experience recurrent respiratory in- LOF MALT1 mutations show CID, enteropathy, and severe
fections, hypogammaglobulinemia with low IgG, IgM, and IgA eczema together with dysmorphic features. Most patients have

46 Cell 168, January 12, 2017


hypogammaglobulinemia but paradoxically increased levels of NF-kB signals from Dectin-1 and Dectin-2, two receptors that
IgE, termed hyper-IgE. T regulatory cell (Treg) counts were recognize Candida albicans in unicellular and hyphal forms,
decreased causing autoimmune features. Patient cells showed respectively. If either receptor is activated, signaling through
impaired NF-kB responses to PMA + ionomycin, PHA, and the SYK kinase leads to sequential assembly of the CARD9
anti-CD3 stimulation marked by decreased BCL10 cleavage CBM complex and downstream NF-kB signaling (Blonska and
and IkBa degradation leading to immunodeficiency (Charbit- Lin, 2011). Cells from a patient with a p.Y92H variant in the
Henrion et al., 2016; Jabara et al., 2013). How specific gene CARD domain showed impaired phosphorylation of IkBa after
expression changes cause particular confluences of immunode- stimulation with zymosan, a Dectin-1 inducer. However, when
ficiency and autoimmunity has not been explained. mutant and wild-type CARD9 proteins were co-expressed with
MALT1 harbors a death domain (DD) and is distantly homolo- BCL10 and MALT1, they both assembled CBM complexes.
gous to caspases in its C-terminal domain, which confers cas- Therefore, it is still controversial whether the NF-kB signaling
pase-like proteolytic function. Its substrates include BCL10, defect in the CARD9-deficient patient cells is CBM dependent
NIK, RELB, CYLD, and A20, all of which are key NF-kB regula- or not. Moreover, CARD9-deficient cells showed blunted
tors. MALT1 enzymatic activation is induced by dimerization GM-CSF responses mediated by the ERK signaling pathway,
and substrate engagement, rather than processing as a reminiscent of defects in neutrophil killing and recruitment
zymogen like conventional caspases. Caspase function both defects reported in CARD9-deficient patients (Gavino et al.,
promotes and inhibits specific NF-kB functions (Hachmann 2016). CARD9-deficient patients also have decreased Th17
and Salvesen, 2016). Human mutations are found throughout T cells, which are crucial for controlling mucosal, but not sys-
the CARD, paracaspase, and Ig-like domains, and they are all temic, candidiasis (Glocker et al., 2009).
LOF (Charbit-Henrion et al., 2016; Jabara et al., 2013). These de- CARD11/CARMA1. In 2006, CARD11, together with MALT1,
fects resemble the phenotypes found in the malt1 knockout were found to be targeted by somatic mutations that induce
mouse (Ruefli-Brasse et al., 2003). However, five out of six constitutive activation of NF-kB and survival signals for a partic-
MALT1-deficient patients also had inflammatory disorders espe- ular subtype of human Diffuse Large B Cell Lymphoma (DLBCL)
cially enteropathy (Charbit-Henrion et al., 2016). Inflammation is (Ngo et al., 2006) (see below). CARD11 missense mutations are
absent from malt1 knockout mice but emerged in genetically en- found in 9.6% of DLBCL tumor biopsies. Introducing the mutant
gineered paracaspase-dead mice indicating enzyme deficiency CARD11 into lymphoma cell lines constitutively induces NF-kB
is involved (Yu et al., 2015). (Lenz et al., 2008) (see below). Germline CARD11 mutations
BCL10. BCL10 deficiency, identified in one patient, resembles can cause very different disease phenotypes. GOF mutations
MALT1 deficiency (Torres et al., 2014). The consanguineous pa- cause BENTA disease (B cell expansion with NF-kB and T cell
tient exhibited CID and autoimmunity with gastroenteritis, recur- anergy), whereas LOF mutations cause SCID. Families with
rent respiratory viral infections, and oral candidiasis. Lympho- hypomorphic, dominant-negative CARD11 mutations also have
cyte counts were normal but exhibited overwhelmingly naive significant atopic disease, including atopic dermatitis, allergy,
phenotypes and undetectable Tregs, implying disturbed CP acti- high IgE and eosinophils, asthma, and immunodeficiency (J. Mil-
vation. Patient fibroblasts showed impaired NF-kB responses to ner, A. Snow, and E. Gelfand, personal communication). Each
LPS (TLR4), zymosan (TLR2/6), and poly(I:C) (TLR3), marked by disease provides a window into CBM signalosome function
decreased nuclear translocation and deficient IL-6 and IL-8 pro- and potential therapeutic strategies (Greil et al., 2013; Snow
duction, hence innate immunity also failed. Interestingly, patient et al., 2012).
mononuclear cells showed normal innate responses, which was The discovery of BENTA disease fits with the idea that
a surprising difference in wiring between humans and mice CARD11 was a bona fide oncogene in DLBCL. Four different
because bcl10 knockout mice display impaired NF-kB in macro- autosomal dominant GOF germline mutations of CARD11,
phages and DCs following innate stimuli. G116S, E127G, G123D, and C49Y, have been discovered in
CARD9. CARD9 deficiency illustrates how NF-kB connects four different families (Buchbinder et al., 2015; Snow et al.,
different receptors and adaptors to specific cellular responses 2012). Three affect the CC domain and one variant, C49Y, is
(Glocker et al., 2009). Homozygous nonsense (LOF) mutations located in the N-terminal CARD. The mutant proteins aggregate,
in CARD9 were first found in a large consanguineous family colocalize with MALT1, activate IKK, and increase nuclear local-
and caused recurrent fungal infections especially in the brain. ization of NF-kB and transcription (Buchbinder et al., 2015; Snow
More recently, a large variety of patients with CARD9 defi- et al., 2012). The germline mutations in the CC and CARD do-
ciencies have been reported, and they consistently show severe mains recapitulated or were identical to the somatic DLBCL mu-
fungal infections especially rare deep dermatophytoses and tations (Lenz et al., 2008). The germline GOF mutations resulted
CNS candidiasis. The patients carried LOF mutations such as in polyclonal, sometimes even monoclonal, B cell lymphocy-
stop-gain nonsense codons as well as missense mutations, tosis, expansion of transitional B cells, and hyper-responsive-
although not all missense mutations have been tested rigorously ness of B cells to BCR stimulation, CD40L, and BAFF, all through
for functional effects. Typically disease onset was in childhood; NF-kB. Similar to B cells, T cells from BENTA patients also
however, some patients presented in adulthood illustrating that showed spontaneously aggregated CARD11 and NF-kB activa-
there might be modifying genes or environmental influences tion. However, peripheral T cells were not expanded and poorly
involved (Lanternier et al., 2015). responsive to TCR stimulation, likely due to anergy mechanisms
A molecular understanding of the human phenotypes emerges (Snow et al., 2012). Although BENTA patients did not develop
from mouse knockout studies showing that CARD9 transduces T cell lymphocytosis and T cell lymphoma, GOF mutations of

Cell 168, January 12, 2017 47


CARD11 have been detected in adult T cell leukemia/lymphoma cyte and not immune cell expression. More intriguingly, the
(ATLL) and in a form of cutaneous T cell lymphoma (CTCL) called CARD14 mutations increase expression in skin causing NF-kB
Sézary syndrome (Juilland and Thome, 2016). Also, one BENTA hyperactivation in skin cells. Some patient mutations displayed
patient developed B cell chronic lymphocytic leukemia (Snow autonomous activation in luciferase assays, whereas others
et al., 2012). The somatic and germline GOF mutations appear showed increased TNF responses. However, CARD14 increases
to be driver mutations indicating that CARD11 may be a prom- and NF-kB hyperactivation have also been observed in psoriasis
ising therapeutic target for BENTA plus other lymphoid malig- without CARD14 mutations (Harden et al., 2015). Thus, these
nancies. features may be a cause or consequence of psoriatic skin lesions
However, the discovery of CARD11 LOF mutations tells a or both.
different story. Patients with homozygous recessive LOF LUBAC Complex and Its Regulators: HOIL1, HOIP, A20,
CARD11 mutations display severe immunodeficiency with pre- OTULIN, and CYLD. Ubiquitination is a crucial post-transla-
dominantly naive T and B lymphocytes and a block in B cell tional protein modification governing NF-kB pathways [8, 113].
maturation. Antibody production by B cells and cytokine produc- Many pathway components, including NEMO, IkBs, p100, and
tion and proliferation by T cells are severely impaired (Greil et al., adaptors such as MALT1 and BCL10 rely on specific types of
2013; Stepensky et al., 2013). Patient lymphocytes showed regulatory and degradative ubiquitination (Iwai, 2014). Specif-
essentially no degradation of IkBa or p65 phosphorylation in ically, non-degradative or ‘‘regulatory’’ ubiquitination is the
response to TCR or BCR stimulation (Stepensky et al., 2013). attachment of polyubiquitin (joined at K63 residues) to signaling
Overall, the clinical and biochemical features of MALT1, proteins. As mentioned above, this provides the reversible
BCL10, and CARD11 LOF mutations are similar, testimony to ‘‘glue’’ that facilitates the multiprotein complexes obligatory for
synergistic functions in the CBM. One characteristic feature of NF-kB activation. Certain ubiquitin reactions are facilitated by
SCID caused by CBM deficiency is that the patients can have TRAF proteins. The CP may also involve linear ubiquitin modifi-
normal peripheral T and B cell counts, yet a predominantly naive cations catalyzed by the ‘‘linear ubiquitin assembly complex’’
lymphocyte phenotype indicative of failed TCR and BCR re- (LUBAC) (Sasaki and Iwai, 2015). The enzyme holocomplex is
sponses. Thus, there appear to be different requirements for composed of three key proteins. HOIL-1 interacting protein
NF-kB signaling in lymphocyte ontogeny and mature lympho- (HOIP) is the E3 ligase. Haem-oxidized IRP2 ubiquitin ligase-1
cytes. The recent report of somatic reversion in a CARD11 LOF (HOIL1) and SHARPIN (SHANK-associated RH domain interact-
mutation patient, who presented as Omenn syndrome, under- ing protein) complete the active LUBAC complex that generates
scored this point. In this patient, a second-site somatic reversion Methionine 1-linked linear polyubiquitin chains in the presence of
changed the original germline mutation (p.C150*) to a missense several E2s to specifically during CP activation (Tokunaga,
mutation (p.C150L), which partly restored CARD11 function. 2013). NEMO is a LUBAC target, but there may be multiple,
The somatic mutation appeared only in a subclone of T cells perhaps overlapping, ubiquitination processes in NF-kB activa-
indicating a survival and expansion advantage conferred in a tion (Sasaki and Iwai, 2015). Negative feedback for NF-kB acti-
founder cell by reinstating NF-kB signaling. However, the expan- vation is arbitrated by several deubiquitinases, including CYLD
sion of a single T cell clone perturbed immune homeostasis and (CYLD lysine 63 deubiquitinase), OTULIN (OTU deubiquitinase
led to massive T cell infiltration into the skin causing progressive with linear linkage specificity), and A20/TNFAIP3 (TNF alpha
eczema and erythroderma, accompanied by lymphadenopathy induced protein 3) (Tokunaga, 2013). Except for SHARPIN, all
and hepatosplenomegaly. Furthermore, the oligoclonal T cells the other proteins in the LUBAC complex and its regulators are
did not provide sufficiently broad immune protection against un- associated with monogenic diseases.
controlled CMV infections and multiple bouts of sepsis caused HOIL1. HOIL1/RBCK1 deficiency was found in three patients
by Staphylococcus aureus, Enterococcus, and Pseudomonas from two unrelated families with immunodeficiency, autoinflam-
that eventually proved fatal to the child at 16 months of age mation and amylopectinosis, a glycogen storage disease that
(Fuchs et al., 2015). causes liver failure. CP impairment accounted for the invasive
CARD14. CARD14, a less well-studied member of the CARD- pyogenic bacterial infections but did not explain the autoinflam-
family, nucleates an alternative CBM complex and is the mation and amylopectinosis. The mutations were autosomal
apparent causative gene in two related diseases: psoriasis recessive LOF leading to no detectable HOIL1 protein and
2 (PSORS2) and Familial Pityriasis Rubra Pilaris (PRP), a psoria- decreased LUBAC complex stability. NEMO ubiquitination in
sis variant that chiefly affects hair follicles (Fuchs-Telem et al., response to TNF or IL-1 stimulation was defective. Conse-
2012; Jordan et al., 2012). Dominant GOF mutations lead to pso- quently, less NEMO was recruited to the RIP1 or IRAK-1 adaptor
riasis with or without arthritis. GOF mutations in CARD14 can proteins in cytokine receptor signaling complexes, IKK was
also lead to generalized pustular psoriasis (Sugiura, 2014). underphosphorylated, and IkB degradation was delayed.
A coding SNP in CARD14 (R820W) has been associated with HOIL1 overexpression in patient cells rescued the protein levels
sporadic (non-congenital) psoriasis in a meta-genome-wide as- of HOIP and SHARPIN and generated a durable LUBAC com-
sociation study (GWAS) (Tsoi et al., 2012). plex, which improved signaling (Boisson et al., 2012).
The discovery of heterozygous CARD14 mutations selectively The mechanisms of autoinflammation and amylopectinosis
in psoriatic diseases revealed an unexpected function of in HOIL1-deficient patients were further investigated using
CARD14 in keratinocytes and innate immunity. CARD14 was Hoil1 knockout mice. These showed spontaneous amylo-
thought to mediate NF-kB signaling like other CARD proteins. pectin-like deposits in the myocardium, susceptibility to Listeria
However, the specific skin manifestations are due to keratino- monocytogenes, Toxoplasma gondii, and Citrobacter rodentium

48 Cell 168, January 12, 2017


infections, but not spontaneous autoinflammation (MacDuff Nucleotide variants in A20 are associated in GWAS with mul-
et al., 2015). Immunodeficiency associated with this molecule tiple autoimmune diseases, including systemic lupus erythema-
affects innate immunity, because Hoil1 deficiency in Rag1-defi- tosus (SLE), rheumatoid arthritis, psoriasis, type 1 diabetes, ce-
cient mice with no lymphocytes still created a susceptibility liac disease, Crohn’s disease, coronary artery disease in type
to Listeria infection. Surprisingly, hoil1/ mice displayed 2 diabetes, and systemic sclerosis (Ma and Malynn, 2012). A20
enhanced control of MHV68 and Mycobacterium tuberculosis mutations were identified in a monogenic syndrome that resem-
infections compared to wild-type mice. Both serum IL-6 and bled Behçet’s disease, an autoinflammatory disease involving
TNF levels were increased in infected mice, which was also mucosal ulcers and eye inflammation (Zhou et al., 2016). All of
evident in HOIL1-deficient patients thereby providing insight the mutations were heterozygous and either frameshift or
into the autoinflammatory disorder. How defects in the LUBAC nonsense, which led to haploinsufficient A20 expression. After
complex lead to dramatically variable susceptibilities to different TNF stimulation, patient lymphocytes and fibroblasts showed
pathogens is unknown. The animal models showed that chronic increased K63-ubiquitinated NEMO, increased phosphorylation
viral infections in the hoil1/ mouse protected them from Liste- of IkBa and IKKs, and prolonged nuclear NF-kB. The patients
ria infection, possibly due to the increased production of the had elevated serum TNF, IP-10, IL-17, IL-9, IFN, and IL-6. Similar
cytokines IL-6, TNF, and IL-12, which enhanced the ability of to A20-deficient mice, the patients also had greater levels of pro
macrophages to kill Listeria (MacDuff et al., 2015). Therefore, IL-1b, NLRP3, and activated caspase-1 and increased produc-
HOIL1 deficiency in mice and humans revealed a homeostatic tion of mature IL-1b after LPS stimulation. Treatment with an
balance between inflammation and immune protection. IL-1 inhibitor, Anakinra, reversed systemic inflammation (Zhou
HOIP. A single case of autosomal recessive deficiency of HOIP et al., 2016). Additional patients with heterozygous missense
has been reported, which provides unique information regarding mutations were discovered shortly after the first report, which
the biological functions of HOIP since the hoip/ mouse is em- raised the possibility that the mutant protein was dominant nega-
bryonic lethal (Boisson et al., 2015; Peltzer et al., 2014). The tive, but this is not yet verified (Ohnishi et al., 2016; Shigemura
HOIP patient has a homozygous missense mutation, which et al., 2016).
severely impaired LUBAC stability. Consequently, this patient OTULIN. OTULIN is another deubiquitinase that cleaves the
phenocopies the HOIL-1-deficient patients with susceptibility N-terminal methionine-linked polyubiquitination chains gener-
to infections, amylopectinosis, and autoinflammation. ated by LUBAC (Keusekotten et al., 2013). Four families with
The biochemical defects in the HOIP-deficient patient were homozygous mutations of OTULIN had autosomal recessive in-
different from the HOIL-1-deficient patients, although it might flammatory disease with recurrent nodular panniculitis with fever
be difficult to draw conclusions based on a single patient with and neutrophil infiltration, lipodystrophy, diarrhea, joint swelling,
a missense mutation. The HOIP-deficient patient showed and failure to thrive. The mutations severely decreased protein
severely decreased linear ubiquitination, decreased phosphory- levels and deubiquitinase activity. Patient cells showed persist-
lation of IKK, and slightly delayed IkB degradation in response to ing IKK activity and IkBa phosphorylation in response to TNF
IL-1b and TNF. However, when stimulated with CD40L, the pa- stemming from a lack of NEMO deubiquitination. Patients had
tient B cells showed no NF-kB signaling, indicated by a failure increased cytokine profiles resembling the A20-deficient pa-
of IKK phosphorylation and IkBa degradation. As a result, the pa- tients. Interestingly, conditional deletion of otulin in myeloid cells,
tient’s B cells failed to induce CD27 or CD38 when activated but not T or B cells, causes systemic inflammation, neutrophil
with CD40L/IL-21. Interestingly, the defects in the patient infiltration, and multi-organ inflammation in mouse models.
B cells resembled CD40-deficient patients, suggesting that the Treatment with anti-TNF-neutralizing antibodies ameliorated
CD40-CD40L signaling, at least in the CP, requires LUBAC. inflammation in patients and rescues mouse phenotypes, indi-
Although both IL-1b and TNF and induced NF-kB signaling cating that TNF is a key driver of the systemic disease (Dam-
were impaired in HOIP-deficient cells, the defect in IL-1b was gaard et al., 2016). Considering the similar biochemical defects
more severe than TNF, a phenomenon also observed in HOIL- and cytokine profiles in A20-deficient and OTULIN-deficient
1-deficient cells (Boisson et al., 2015). patients, TNF or IL-1 blockers may be an effective treatment.
A20/TNFAIP3. A20/TNFAIP3 is a ubiquitin removal enzyme CYLD. CYLD (cylindromatosis protein) was first mapped to
regulating NF-kB [123-125]. It contains a C-terminal OTU chromosome 16q12-13 in 1995 by the familial skin cylidromato-
(Ovarian Tumor) domain, which mediates its de-ubiquitylating sis phenotype (Biggs et al., 1995). Later, a large cohort of cylidro-
(DUB) activity and seven zinc fingers (ZFs) that harbor its matosis patients helped to identify CYLD as a tumor suppressor
E3 ubiquitin binding and cleavage activities. A20 expression is gene and NF-kB regulator (Bignell et al., 2000). CYLD contains
induced by NF-kB and then decreases NF-kB signaling as a three cytoskeletal-associated protein-glycine-conserved (CAP-
negative feedback loop. A20 reverses multiple NF-kB signaling GLY) domains that function as a deubiquitinating enzyme that
pathways by cleaving the K63-linked polyubiquitin chains on targets key molecules such as NEMO. Moreover, CYLD contains
activated RIP1, RIP2, TRAF6, TBK1, and NEMO. A20 also in- a USP catalytic domain that acts on ubiquitin at specific lysine
hibits ubiquitin chain synthesis by blocking the binding of E2 residues and a molecular triumvirate of Cys601-His871-
and E3 proteins (Ma and Malynn, 2012). A20 also negatively reg- Asp889 in the a1 helical subdomain that mounts a nucleophilic
ulates Nlrp3 inflammasome signaling by suppressing Nlrp3, attack on K63 residues in polyubiquitin.
proIL-1b, and proIL-18 production (Duong et al., 2015; Vande The major biological function revealed by CYLD-deficient pa-
Walle et al., 2014). Thus, A20 connects NF-kB regulation to a tients is the control of cell growth as indicated by profound cylin-
broader spectrum of inflammation functions. dromatosis (Brooke-Spiegler syndrome), a benign tumor of the

Cell 168, January 12, 2017 49


sweat glands around the head, face, and neck (Mathis et al., 10 gene that can function during embryogenesis in humans,
2015). CYLD mutations are also associated with another skin which has been lost in evolution in the mouse genome (Su and
disease, multiple familial trichoepithelioma, type 1 (MFT1), char- Lenardo, 2008).
acterized by numerous firm skin papules that are trichoepithe- Alternative Pathway
liomas (follicular tumors) (Farkas et al., 2016). Why so many NIK. Two NIK/MAP3K14-deficient patients were identified in
different CYLD mutations produce tumors of skin origin is not one consanguineous family using next-generation sequencing
known. CYLD was first found to deubiquitinate NEMO, which in- (NGS). The patients suffered recurrent and severe bacterial, viral,
hibits the CP (Kovalenko et al., 2003). CYLD has been proposed and Cryptosporidium infections. Immune phenotyping showed
to shape both adaptive and innate immunity by regulating TCR, multiple lymphocyte defects, including B cell lymphopenia,
BCR, and TLR signaling (Mathis et al., 2015). However, CYLD reduced class-switched memory B cells, impaired B cell sur-
deficiency yields very different phenotypes compared to insuffi- vival, impaired ICOSL expression, defects in follicular helper
ciency of A20 and OTULIN even though both CYLD and OTULIN T cells (Tfh) and memory T cells, and malfunctioning NK cells.
can operate together to regulate the LUBAC complex. This indicates that the triggering kinase for the AP greatly influ-
The difference may lie in the ability of CYLD to govern cell ences key lymphocyte functions.
proliferation, which may not relate to its NF-kB regulatory func- The LOF mutation found in these patients abolished kinase ac-
tion. For example, CYLD deficiency may cause overexpression tivity against IKKa (Willmann et al., 2014). The phenotype of pa-
of the c-myb oncogene as a feature of cylindromas (Corda and tients was different from the NIK knockout mouse because it
Sala, 2016). However, in addition to NEMO, activated CYLD lacked the severe structural defects in lymph nodes, Peyer’s
can remove the lysine 63-linked polyubiquitin chains of BCL-3 patches, and splenic and thymic structures found in the latter
(an atypical IkB), thus preventing BCL-3 translocation into the (Yin et al., 2001). Nonetheless, bone marrow chimeras and con-
nucleus together with p52 and p50, which may also promote ditional lymphocyte knockout mouse models recapitulated the
cell proliferation. Keratinocytes defective in CYLD showed patient phenotype, including decreased memory T cells, disrup-
increased nuclear translocation of BCL-3, p52, and p50 after ted B cell maturation and activation, and defective DCs and NK
chemical or UV treatment (Massoumi et al., 2006). Therefore, cells (Brightbill et al., 2015; Katakam et al., 2015; Noma et al.,
CYLD not only regulates the activation of canonical NF-kB in 2015; Rowe et al., 2013). These results imply that NIK action
conditions of infection and inflammation, it also suppresses in non-hematopoietic tissues is crucially different in humans
cell proliferation under stress conditions. and mice.
Caspase-8. Caspase-8 (CASP8) deficiency revealed a surpris- NFKB2/p100/p52. Heterozygous NFKB2/p100/p52 mutations
ing role for this enzyme in the NF-kB pathway. This was verified have been identified in an autosomal-dominant type of CVID,
only when conditional knockouts in lymphocytes were made complicated with autoimmunity and endocrine deficiency. The
since the germline deficiency in mice is embryonic lethal (Lem- patients exhibited recurrent respiratory infections, hypogamma-
mers et al., 2007). CASP8 belongs to the cysteine-aspartic globulinemia, alopecia areata, an autoimmune infiltration of the
acid protease (caspase) family and has a well-established role scalp that causes hair loss, and, frequently, adrenal insuffi-
in the apoptosis signaling cascade from death receptors such ciency. They manifest poor antibody responses, predominantly
as Fas (Price et al., 2014). However, rare patients with autosomal unswitched naive B cells in the periphery, and some reports of
recessive disease were found to have homozygous LOF CASP8 T and NK cell activation defects (Chen et al., 2013; Lee et al.,
mutations (Chun et al., 2002). The disease involved lymphade- 2014; Lougaris et al., 2015). In resting lymphocytes, the NFKB2
nopathy, splenomegaly, and defective CD95/FAS-induced protein predominantly exists as the p100 precursor form.
apoptosis of peripheral blood lymphocytes. In addition, unlike Upstream signaling, especially TRAF3, activates NIK and IKKa
related patients with the autoimmune lymphoproliferative syn- dimers, which, in turn, phosphorylate the two regulatory serines
drome, the disease also included immunodeficiency with recur- in the NRD domain (S866 and S870). Consequently, the canon-
rent sinopulmonary and herpes simplex virus (HSV) infections. ical acceptor site (K856) is ubiquitinated, which leads to the
T, B, and NK cells from patients responded poorly to stimuli limited processing of p100 to p52 in the proteasome (Sun,
through TCR, BCR, and Fc receptors (Niemela et al., 2015). 2012). So it is no surprise that all the mutations found so far
The immunodeficiency observed in the patients led to the dis- (p.R853*, p.R853Afs*29, p.K855Sfs*7, p.D865G, p.D865Vfs*17,
covery of CASP8 as a facilitatory molecule in the NF-kB p.A867V, p.A867Cfs*19) affect the NRD domain and either sub-
pathway. During antigen receptor signaling, CASP8 is indispens- stitute amino acids adjacent to S866, S870, and K856 or remove
able for full NF-kB induction and gene transcription by linking the the C terminus that harbors these residues. These mutations
IKK complex to IkBa in preparation for the phosphotransfer abrogate the processing of p100 to p52 before or after CD40-
reaction. Consistently, CASP8-deficient patient cells showed CD40L signaling (Chen et al., 2013; Lee et al., 2014). The
severely impaired NF-kB nuclear translocation in responses to D865G mutation has a dominant effect over the wild-type
immune stimuli, however, normal responses to CD40L stimula- protein. It interferes with the CP by blocking TNF-a-induced
tion (Su et al., 2005). Two more patients found recently NF-kB nuclear translocation (Lee et al., 2014). Unprocessed
confirmed the immunodeficiency but also broadened the pheno- p100 can function as a CP inhibitor by excessive dimerization
type with late-onset multi-organ lymphocytic infiltration with with p65 (Basak et al., 2007), which might explain how NFKB2
granulomas (Niemela et al., 2015). Why does CASP8 LOF gives mutations cause effects similar to NFKB1 deficiency in terms
a live human phenotype, whereas in mice it is embryonically le- of immune abnormalities. However, individuals carrying
thal key? This is likely due to the highly homologous Caspase- NFKB2 N terminus mutations acting in a GOF manner present

50 Cell 168, January 12, 2017


with a more severe CID phenotype (H. Kuehn and S. Rose- 2011; Herman et al., 2012; Pérez de Diego et al., 2010; Sancho-
nzweig, personal communication). Further investigation of these Shimizu et al., 2011). TLR3 recognizes dsDNA in a broad range of
mutations may elucidate the dominant inheritance of the disease tissues and cell types. When activated by dsDNA, TLR3 is phos-
as well as the pathogenesis of the adrenal disorder. phorylated and forms dimers that bind to TRIF to trigger the
RELB. Autosomal recessive RELB deficiency has been discov- downstream signals. The TLR3-TRIF complex recruits TRAF3,
ered in three patients from the same consanguineous family. The TBK1, and IKKƐ, which activates IRF3. The TLR3-TRIF complex
patients presented as CID from infancy, with recurrent infections, also recruits RIP1, TAB2, and TAK1 and then phosphorylates IKK
severe autoimmune skin diseases, and failure to thrive (Sharfe to activate the CP. In fact, the TLR3-TRIF complex supplies a
et al., 2015). The homozygous, LOF mutations led to absence common node for several signaling pathways, which together
of RELB expression but normal levels of other NF-kB/Rel pro- mediate essential antiviral responses in the brain (Lafaille et al.,
teins. Interestingly, the patient’s T cells exhibited increased p65 2012). Patient cells defective of the TLR3-TRIF-TRAF3-TBK1
nuclear translocation and hyperactivation in response to TCR pathway show impaired activation of both IGF3 and NF-kB and
stimulation, whereas B cells showed decreased c-Rel nuclear decreased production of both IFN and IL-6. Both IRF3 and
translocation after CD40 stimulation (Sharfe et al., 2015). This im- NF-kB seem to contribute greatly to the antiviral responses.
plies a cell-specific homeostatic balance between the CP and On one hand, patients with IRF3 mutations share the susceptibil-
AP. Moreover, the T and B cell defects in RELB-deficient patients ity to HSV encephalitis (Andersen et al., 2015). On the other hand,
were not observed in relb knockout mice, which had inflamma- NEMO deficiency also creates susceptibility to HSV encephalitis
tory and hematopoietic abnormalities (Gerondakis et al., 2006; (Audry et al., 2011).
Sun, 2012). TRAF6 and the EDA Signaling. Mutations in EDA (ectodyspla-
TRAFs and Specific NF-kB Pathways. The TRAF (Tumor necro- sin A), EDAR (ectodysplasin A receptor), EDARADD (EDAR-
sis factor receptor-associated factor) family proteins connect re- associated death domain), and TRAF6 (Tumor necrosis factor
ceptors to both the CP and AP, as well as multiple other path- receptor-associated factor 6) all manifest EDA/HED with a
ways (Hayden and Ghosh, 2008). There are seven TRAFs, clinical phenotype similar to patients with hypomorphic NEMO
which link various receptors to downstream pathways to induce mutations, although they lack immunodeficiency (Headon
specific signals. The six typical TRAF members (TRAF1–6) share et al., 2001; Kere et al., 1996; Wisniewski and Trzeciak, 2012a,
similar functional domains, including a C-terminal TRAF domain 2012b). EDA, which belongs to the tumor necrosis factor family,
that mediates homodimerization and interactions with other pro- is expressed on the ectoderm interfollicular cells. EDA activates
teins, a zinc finger domain, and a RING finger domain with EDAR on the surface of follicular cells in mesenchyme, which, in
E3 ubiquitinase ligase activities (except for TRAF1). TRAFs 2, turn, binds to EDARADD and TRAF6 to induce downstream
3, 5, and 6 have E3 ubiquitinase ligase activity. signaling. The EDAR-EDARADD-TRAF6 complex recruits
TRAFs are well-studied signaling adapters for the TNF recep- TAB1, TAB2, and TAK1, which activate the CP by IKK phosphor-
tors (TNFRs). They either directly bind the TRAF-interacting motif ylation. The fact that EDA-EDAR-EDARADD-TRAF6-NEMO mu-
in the cytoplasmic domain of the TNFRs or bind indirectly via tations all present as EDA/HED revealing a prominent function in
other adaptor proteins like TRADD (TNFR1-associated death ectoderm development (Trzeciak and Koczorowski, 2016).
domain protein), which tethers them to specific TNFRs with cyto- Although TRAF6 has been implicated in multiple NF-kB path-
plasmic death domains. Upon binding to the receptors, TRAFs ways, sometimes synergistic with TRAF3, the disease due to
recruit and activate downstream signaling molecules such as TRAF6 mutations shows no resemblance to syndromes due to
TAK1 and IKK. TRAFs also have ubiquitinase activity. This func- other NF-kB components including TRAF3. The exclusive
tion is particularly important in the AP to control the level of NIK. dependence of EDA signaling on TRAF6, together with the exclu-
In cells prior to activation signals, TRAF induces ubiquitination of sive dependence of TLR3 signaling on TRAF3, demonstrates
NIK by forming a complex with the E3 ligase cIAP. Activation that different receptors ignite specific responses through the
leads to the degradation of TRAFs, which fosters NIK accumula- common NF-kB signaling complexes. This specificity is still
tion and AP activation. poorly understood at the molecular level.
The two TRAFs associated with monogenic diseases illus- Other Monogenic Diseases Associated with NF-kB Pathways.
trate the biological functions of the entire pathway. Mutations Immunodeficiency and Dysregulation. Mutations in MYD88
affecting components in the same pathway produce similar phe- (myeloid differentiation primary response 88) and IRAK4 (inter-
notypes in patients. Mutations in the TLR3-TRIF-TRAF3-TBK1 leukin-1 receptor-associated kinase 4) cause clinically indis-
pathway cause HSV encephalitis, whereas those in the EDA- tinguishable phenotypes, characterized by life-threatening,
EDAR-EDARADD-TRAF6 pathway lead to EDA (also known as recurrent pyogenic bacterial infections, especially by invasive
Hypohidrotic ectodermal dysplasia [HED]) (Trzeciak and Koczor- pneumococci (Picard et al., 2003; von Bernuth et al., 2008). Pa-
owski, 2016), which resembles hypomorphic NEMO mutations. tient cells exhibit failures in multiple TLRs (including TLR2, TLR3,
TRAF3 and the TLR3 Signaling. Mutations in TRAF3 (Tumor TLR4, TLR5, TLR9) and two IL-1Rs (IL-1R and IL-18R). The acti-
necrosis factor receptor-associated factor 3), TRIF/TICAM1 vation of NF-kB and JNK pathways downstream of these signals
(toll-like receptor adaptor molecule 1), TLR3 (Toll-like receptor were absent, demonstrated by the absence of IkB degradation
3), and TBK1 (TANK binding kinase 1) cause patients to be highly and JNK phosphorylation, and this led to decreased IL-6 and
susceptible to HSV (Herpes simplex virus) encephalitis, despite IFN-b. This fits with cellular data showing that MYD88 and IRAKs
robust health otherwise. This is an astonishingly specific presen- mediate signaling from almost all TLRs (except for TLR3) and
tation considering the broad function of the pathway (Guo et al., interleukin-1 receptor and related receptors (IL-1Rs). Patient

Cell 168, January 12, 2017 51


cells showed a broad range of defects in response to stimulation cell B cell lymphoma (DLBCL) (Young et al., 2015). DLBCL is
of these receptors. This includes poor responsiveness to the most prevalent lymphoid cancer and the ABC subtype is
poly(I:C), which is thought to be recognized by TLR3. Mouse highly aggressive and resists chemotherapy. Constitutive
knockout models showed a broad vulnerability to essentially all NF-kB activation is ‘‘conditionally essential’’ meaning that the
pathogens tested (common bacteria, virus, parasites, and fungi). transformation phenotype requires it and without it, ABL lym-
However, the restricted susceptibility to pyogenic bacteria phoma cells die (Bidère et al., 2009). Nontransformed B cells
indicates a more specialized function in humans. have no such requirement. This is one of the key concepts.
Developmental Disorders. TAK1/MAP3K7 (mitogen-activated When the conditions of transformation are wired into lymphoma
protein kinase kinase kinase 7) is a member of the serine/threo- cells of this subtype, certain pathways become turned on and
nine protein kinase family. TAK1 unites with TRAF6, TAB1, and the cell becomes dependent, even addicted, to this adrenaline
TAB2 to induce NF-kB signaling. It also activates JNK in re- rush of molecular stimulatory pathways for growth and survival.
sponses to TGF-b signaling. Mutations in TAK1 have been clini- NF-kB, which is normally quiescent and dependent on very spe-
cally associated with frontometaphyseal dysplasia (FMD), a pro- cific molecular signals, is turned on in an unrelenting drive toward
gressive skeletal dysplasia affecting the long bones and skull a self-enforcing ABC lymphoma gene expression pattern. Key to
(Basart et al., 2015; Wade et al., 2016). uncontrolled lymphoma growth is an anti-apoptotic transcrip-
TAB2 (TAK1-binding protein 2) is also a part of the TRAF6- tional program, with boosted expression of apoptosis inhibitors
TAB1/2-TAK1 complex, which activates NF-kB and JNK such as Bcl-2. This underpins the resistance of these malig-
signaling in responses to TGF-b. This explains why TAB2 defi- nancies to chemotherapeutic agents, which provoke apoptosis
ciency has been clinically associated with congenital heart de- to have their toxic effect. Constitutive deregulation of NF-kB pre-
fects including unusual valve dysplasia and Tetralogy of Fallot dictably alters other regulators, such as Bcl-6, PRDM-1, IRF-4
rather than a prominent immune phenotype (Weiss et al., 2015). and others, to give the specific B cell phenotype.
RIPK4/RIP4 (receptor interacting serine/threonine kinase 4) is ABC cells lines may be paradigmatic of other forms of hemato-
a serine/threonine protein kinase that interacts with protein ki- poietic cancers in that experimental elimination of a whole range
nase C-delta. In the balance of inflammation versus nonapop- of NF-kB signaling wires including IKKb, CARMA1, BCL10,
totic cell death that characterizes the RIP family of kinases, MALT1 (or its caspase activity), or LUBAC components induce
RIP4 tips toward NF-kB signaling for inflammation especially in programmed death of the cancer cell. The same has been
skin cells. Mutations in RIPK4 are associated with Popliteal pte- observed when removing components of the BCR signal gener-
rygium syndrome (Bartsocas-Papas type). It is considered to be ation apparatus including CD79A, CD79B, Btk, Syk, Blnk,
a close resembling of IKKa deficiency, which we discussed in PLCg2, and PKCb. Hence, the life of an ABC lymphoma cell de-
section 2 (Kalay et al., 2012). pends on inducing and maintaining active NF-kB. A principal
TRAPPC9/NIBP (trafficking protein particle complex, subunit culprit behind unrelenting NF-kB activation is BCR clustering
9/NIK and IKBKB-binding protein) was originally identified as a reminiscent of the antigen-engaged receptor. The putative trig-
NIK and IKKb interacting protein, which enhances the cyto- gering antigen has not been identified, but plausible candidates
kine-induced NF-kB. The mutations in TRAPPC9 cause mental are endogenous autoantigens (Young et al., 2015). In addition,
retardation (autosomal recessive type 13) (Mir et al., 2009; Mo- enhanced TLR signaling due to mutations in MyD88 and IRAK1
chida et al., 2009). However, the disease is more likely to be further aggravate uncontrolled NF-kB in ABC-DLBCL. Finally,
associated with the function of TRAPPC9 in the TRAPP complex mutations inactivating A20 and potentially other down-modula-
(trafficking protein particle complex), which regulates the vesicle tors add even more fuel to the NF-kB conflagration. Hence,
trafficking of ER-Golgi, intra-Golgi, and endosome-Golgi traf- entire pathways used in normal immune function become recon-
ficking (Hu et al., 2005; Zong et al., 2011). figured in the service of malignancy. Also, the mutations that
activate these pathways and induce NF-kB are selective in
Cancer generating ABC-DLBCL; other variants, such as the ‘‘germinal
Somatic mutations, ones that arise in individual cells as a person center B cell’’ (GCB) form of DLBCL does not show these char-
ages, in genes affecting NF-kB have roles as ‘‘drivers’’ of the acteristic mutations and does not depend on NF-kB (Pasqua-
oncogenic process in specific malignancies. These mutations lucci and Dalla-Favera, 2015). Germline mutations that promote
have two principle effects: stimulation of proliferative pathways NF-kB activation may also create an inherited or de novo predis-
and inhibition of apoptosis, in malignant cells together with position to the development of ABC lymphoma. The key onco-
supporting stroma and blood vessels. There are also corollary genic role of NF-kB is evident in other types of lymphoma, espe-
effects by NF-kB on inflammation and immunity that promote cially Burkitt’s lymphoma, caused in part by EBV, which induces
favorable niches for cancer cells to take root and expand. NF-kB through its LMP-1 function (Neoptolemos et al., 2010).
NF-kB involvement in the pathogenesis of a wide variety of Finally, genes encoding RHD NF-kB proteins or their key
tumors, anti-tumor immunity, and cancer therapies is reported signaling proteins are common at breakpoints of chromosomal
in thousands of publications, so we will focus only on the princi- translocations in lymphomas and leukemias (Nishikori, 2005).
ple malignancy of lymphoid cells, lymphoma, to illustrate a few
important concepts. NF-kB is deranged in lymphomas by NF-kB as a Therapeutical Target
somatic mutations as well as by oncogenic viruses such as Given its key role in immunity, cancer, and other aspects of hu-
Epstein-Barr virus. One instructive example is the role of man physiology, the concept prevails that NF-kB should be a
NF-kB in the ‘‘activated B cell’’ (ABC) subtype of diffuse large valuable pharmaceutical target. In fact, drugs already marketed,

52 Cell 168, January 12, 2017


such as raloxifene, apparently exert therapeutic effects by inhib- This is a testament to how evolution has used these proteins in
iting NF-kB (Olivier et al., 2006). The widespread involvement in different ways in the two species. In many cases, we do not
signaling means that many drug targets exist in the relevant know precisely what these differences are: they may be quanti-
pathway components and a single successful drug may have tative or qualitative. Because NF-kB is involved in infection con-
utility in multiple diseases. The centrality of NF-kB in innate trol and the pathogens impinging on the lives of humans and
and adaptive immune receptor signaling suggests that NF-kB mice are so different, evolution would appear to have adapted
inhibitory drugs could be widely efficacious against autoimmune to the precise requirements of the two species. If similar work
and lymphoproliferative disorders. However, these crossover were possible with more species, an increasing richness of
effects may also be a harbinger of unexpected adverse side ef- specificity would presumably emerge.
fects. Inhibition of NF-kB with test therapeutics has been found
to cause unexpected, even counterintuitive, effects such as neu- AUTHOR CONTRIBUTIONS
trophilia, fever, and abnormal IL-1 release. We will illustrate this
paradox with drug development in cancer. Since NF-kB inhibits Q.Z., M.J.L., and D.B. were involved in conceptualizing, researching, and
apoptosis and this is obligatory for cell survival, for example, in writing this review.
ABC-DLBCL lymphoma, then blocking NF-kB should be an
effective treatment (Young et al., 2015). Furthermore, upregu- ACKNOWLEDGMENTS
lated NF-kB promotes cell proliferation, metastasis, metabolic
We apologize to the many hundreds of colleagues over the past 30 years
changes, and other abnormalities that favor the expansion and whose work has created this rich tapestry of science but could not be cited
spread of malignancy (Kim et al., 2006; Perkins, 2012). Thus, due to space limitations. We thank Sankar Ghosh for critically reading the
strategies to suppress NF-kB have been clinically tested, most manuscript. This work was supported by the Intramural Research Program
prominently proteasome blockers and IKK inhibitors. Bortezimib of the NIAID, NIH.
(Velcade) is approved for multiple myeloma, a plasma cell malig-
nancy, whose aggressiveness depends in part on NF-kB (Kim REFERENCES
et al., 2006). Also, thalidomide and other putative IKK inhibitors
have been successful in myeloma. These agents, however, Andersen, L.L., Mørk, N., Reinert, L.S., Kofod-Olsen, E., Narita, R., Jørgensen,
S.E., Skipper, K.A., Höning, K., Gad, H.H., Østergaard, L., et al. (2015). Func-
have been thwarted by side effects potentially due to NF-kB
tional IRF3 deficiency in a patient with herpes simplex encephalitis. J. Exp.
suppression including nephrotoxicity, neuropathy, and also the Med. 212, 1371–1379.
relapse of more aggressive forms of malignancy (Mina et al.,
Audry, M., Ciancanelli, M., Yang, K., Cobat, A., Chang, H.H., Sancho-Shimizu,
2016). Finally, as the pivotal role of the immune system in fighting V., Lorenzo, L., Niehues, T., Reichenbach, J., Li, X.X., et al. (2011). NEMO is a
malignancy has come into focus with new discoveries in immu- key component of NF-kappaB- and IRF-3-dependent TLR3-mediated immu-
notherapy, NF-kB inhibition is cast into a new light (Couzin- nity to herpes simplex virus. J. Allergy Clin. Immunol. 128, 610–617.
Frankel, 2013). Potent NF-kB inhibitors may emasculate T cells Baltimore, D. (2011). NF-kB is 25. Nat. Immunol. 12, 683–685.
that antigenically recognize and kill tumor cells, thereby wors- Basak, S., Kim, H., Kearns, J.D., Tergaonkar, V., O’Dea, E., Werner, S.L.,
ening disease. Thus, the broad role of NF-kB in cellular regula- Benedict, C.A., Ware, C.F., Ghosh, G., Verma, I.M., and Hoffmann, A.
tion makes its druggability complicated. (2007). A fourth IkappaB protein within the NF-kappaB signaling module.
Cell 128, 369–381.
Conclusions and Future Perspectives Basart, H., van de Kar, A., Adès, L., Cho, T.J., Carter, E., Maas, S.M., Wilson,
In the 30 years that we have known about the seminal roles L.C., van der Horst, C.M., Wade, E.M., Robertson, S.P., and Hennekam, R.C.
(2015). Frontometaphyseal dysplasia and keloid formation without FLNA
NF-kBs, much has been learned about their biochemical proper-
mutations. Am. J. Med. Genet. A. 167, 1215–1222.
ties, their responses to signaling events and their physiological
Ben-Neriah, Y. (2002). Regulatory functions of ubiquitination in the immune
roles. Given the enormous number of genes activated by NF-kBs system. Nat. Immunol. 3, 20–26.
and the diverse modes of signaling that impinge upon these tran-
Bidère, N., Ngo, V.N., Lee, J., Collins, C., Zheng, L., Wan, F., Davis, R.E., Lenz,
scription factors, there remains much to learn. Most of the basic G., Anderson, D.E., Arnoult, D., et al. (2009). Casein kinase 1alpha governs an-
work on NF-kB was done in mice and their cells. However, by tigen-receptor-induced NF-kappaB activation and human lymphoma cell
taking advantage of the striking phenotypes of human mutants survival. Nature 458, 92–96.
in NF-kBs and their related proteins, we can now generate Biggs, P.J., Wooster, R., Ford, D., Chapman, P., Mangion, J., Quirk, Y.,
data on the roles of NF-kB in human biology and compare those Easton, D.F., Burn, J., and Stratton, M.R. (1995). Familial cylindromatosis
to mouse data. As this review shows, that has proved very useful. (turban tumour syndrome) gene localised to chromosome 16q12-q13:
The many investigators seeking human mutations have now Evidence for its role as a tumour suppressor gene. Nat. Genet. 11, 441–443.

found defects in almost all of the core molecules in the NF-kB Bignell, G.R., Warren, W., Seal, S., Takahashi, M., Rapley, E., Barfoot, R.,
Green, H., Brown, C., Biggs, P.J., Lakhani, S.R., et al. (2000). Identification
pathways. This provides a rich set of tools to study signaling
of the familial cylindromatosis tumour-suppressor gene. Nat. Genet. 25,
and transcription in humans by proteins related to infection 160–165.
and inflammation. Studies in humans that correlate biochemical
Blonska, M., and Lin, X. (2011). NF-kB signaling pathways regulated by
alterations with physiological defects are difficult to perform but CARMA family of scaffold proteins. Cell Res. 21, 55–70.
these ‘‘experiments of nature’’ offer the opportunity to make
Boisson, B., Laplantine, E., Prando, C., Giliani, S., Israelsson, E., Xu, Z., Ab-
these connections. hyankar, A., Israël, L., Trevejo-Nunez, G., Bogunovic, D., et al. (2012). Immu-
It is striking how often the effects in humans differ from those in nodeficiency, autoinflammation and amylopectinosis in humans with inherited
mice when similar or even identical mutations are compared. HOIL-1 and LUBAC deficiency. Nat. Immunol. 13, 1178–1186.

Cell 168, January 12, 2017 53


Boisson, B., Laplantine, E., Dobbs, K., Cobat, A., Tarantino, N., Hazen, M., Li- Egawa, T., Albrecht, B., Favier, B., Sunshine, M.J., Mirchandani, K., O’Brien,
dov, H.G., Hopkins, G., Du, L., Belkadi, A., et al. (2015). Human HOIP and W., Thome, M., and Littman, D.R. (2003). Requirement for CARMA1 in antigen
LUBAC deficiency underlies autoinflammation, immunodeficiency, amylopec- receptor-induced NF-kappa B activation and lymphocyte proliferation. Curr.
tinosis, and lymphangiectasia. J. Exp. Med. 212, 939–951. Biol. 13, 1252–1258.
Boztug, H., Hirschmugl, T., Holter, W., Lakatos, K., Kager, L., Trapin, D., Pickl, Elgueta, R., Benson, M.J., de Vries, V.C., Wasiuk, A., Guo, Y., and Noelle, R.J.
W., Förster-Waldl, E., and Boztug, K. (2016). NF-kB1 haploinsufficiency (2009). Molecular mechanism and function of CD40/CD40L engagement in the
causing immunodeficiency and EBV-driven lymphoproliferation. J. Clin. Immu- immune system. Immunol. Rev. 229, 152–172.
nol. 36, 533–540. Farkas, K., Deák, B.K., Sánchez, L.C., Martı́nez, A.M., Corell, J.J., Botella,
Brightbill, H.D., Jackman, J.K., Suto, E., Kennedy, H., Jones, C., 3rd, Chala- A.M., Benito, G.M., López, R.R., Vanecek, T., Kazakov, D.V., et al. (2016).
sani, S., Lin, Z., Tam, L., Roose-Girma, M., Balazs, M., et al. (2015). Conditional The CYLD p.R758X worldwide recurrent nonsense mutation detected in
deletion of NF-kB-inducing kinase (NIK) in adult mice disrupts mature B cell patients with multiple familial trichoepithelioma type 1, Brooke-Spiegler syn-
survival and activation. J. Immunol. 195, 953–964. drome and familial cylindromatosis represents a mutational hotspot in the
gene. BMC Genet. 17, 36.
Buchbinder, D., Stinson, J.R., Nugent, D.J., Heurtier, L., Suarez, F., Sukumar,
G., Dalgard, C.L., Masson, C., Parisot, M., Zhang, Y., et al. (2015). Mild B-cell Filipe-Santos, O., Bustamante, J., Haverkamp, M.H., Vinolo, E., Ku, C.L., Puel,
lymphocytosis in patients with a CARD11 C49Y mutation. J. Allergy Clin. Im- A., Frucht, D.M., Christel, K., von Bernuth, H., Jouanguy, E., et al. (2006).
munol. 136, 819–821. X-linked susceptibility to mycobacteria is caused by mutations in NEMO im-
pairing CD40-dependent IL-12 production. J. Exp. Med. 203, 1745–1759.
Charbit-Henrion, F., Jeverica, A.K., Begue, B., Markelj, G., Parlato, M., Avcin,
Fliegauf, M., Bryant, V.L., Frede, N., Slade, C., Woon, S.T., Lehnert, K., Winzer,
S.L., Callebaut, I., Bras, M., Parisot, M., Jazbec, J., et al. (2016). Deficiency in
S., Bulashevska, A., Scerri, T., Leung, E., et al. (2015). Haploinsufficiency of the
mucosa associated lymphoid tissue lymphoma translocation 1 (MALT1): A
NF-kB1 Subunit p50 in Common Variable Immunodeficiency. Am. J. Hum.
novel cause of Ipex-like syndrome. J. Pediatr. Gastroenterol. Nutr. Published
Genet. 97, 389–403.
online: June 2, 2016.
Fuchs, S., Rensing-Ehl, A., Pannicke, U., Lorenz, M.R., Fisch, P., Jeelall, Y.,
Chen, K., Coonrod, E.M., Kumánovics, A., Franks, Z.F., Durtschi, J.D., Mar-
Rohr, J., Speckmann, C., Vraetz, T., Farmand, S., et al. (2015). Omenn syn-
graf, R.L., Wu, W., Heikal, N.M., Augustine, N.H., Ridge, P.G., et al. (2013).
drome associated with a functional reversion due to a somatic second-site
Germline mutations in NFKB2 implicate the noncanonical NF-kB pathway in
mutation in CARD11 deficiency. Blood 126, 1658–1669.
the pathogenesis of common variable immunodeficiency. Am. J. Hum. Genet.
93, 812–824. Fuchs-Telem, D., Sarig, O., van Steensel, M.A., Isakov, O., Israeli, S., Nous-
beck, J., Richard, K., Winnepenninckx, V., Vernooij, M., Shomron, N., et al.
Chun, H.J., Zheng, L., Ahmad, M., Wang, J., Speirs, C.K., Siegel, R.M., Dale,
(2012). Familial pityriasis rubra pilaris is caused by mutations in CARD14.
J.K., Puck, J., Davis, J., Hall, C.G., et al. (2002). Pleiotropic defects in lympho-
Am. J. Hum. Genet. 91, 163–170.
cyte activation caused by caspase-8 mutations lead to human immunodefi-
ciency. Nature 419, 395–399. Gavino, C., Hamel, N., Zeng, J.B., Legault, C., Guiot, M.C., Chankowsky, J.,
Lejtenyi, D., Lemire, M., Alarie, I., Dufresne, S., et al. (2016). Impaired
Corda, G., and Sala, A. (2016). Cutaneous cylindroma: It’s all about MYB. RASGRF1/ERK-mediated GM-CSF response characterizes CARD9 defi-
J. Pathol. 239, 391–393. ciency in French-Canadians. J. Allergy Clin. Immunol. 137, 1178–1188.
Courtois, G., Smahi, A., Reichenbach, J., Döffinger, R., Cancrini, C., Bonnet, Gerondakis, S., and Siebenlist, U. (2010). Roles of the NF-kappaB pathway in
M., Puel, A., Chable-Bessia, C., Yamaoka, S., Feinberg, J., et al. (2003). A hy- lymphocyte development and function. Cold Spring Harb. Perspect. Biol. 2,
permorphic IkappaBalpha mutation is associated with autosomal dominant a000182.
anhidrotic ectodermal dysplasia and T cell immunodeficiency. J. Clin. Invest.
Gerondakis, S., Grumont, R., Gugasyan, R., Wong, L., Isomura, I., Ho, W., and
112, 1108–1115.
Banerjee, A. (2006). Unravelling the complexities of the NF-kappaB signalling
Courtois, G., Pescatore, A., Gautheron, J., Fusco, F., Ursini, M.V., and Sen- pathway using mouse knockout and transgenic models. Oncogene 25,
egas, A. (2016). NF-KB-Related Genetic Diseases (Springer Berlin Heidelberg). 6781–6799.
Couzin-Frankel, J. (2013). Breakthrough of the year 2013. Cancer immuno- Glocker, E.O., Hennigs, A., Nabavi, M., Schäffer, A.A., Woellner, C., Salzer, U.,
therapy. Science 342, 1432–1433. Pfeifer, D., Veelken, H., Warnatz, K., Tahami, F., et al. (2009). A homozygous
Damgaard, R.B., Walker, J.A., Marco-Casanova, P., Morgan, N.V., Titheradge, CARD9 mutation in a family with susceptibility to fungal infections. N. Engl.
H.L., Elliott, P.R., McHale, D., Maher, E.R., McKenzie, A.N., and Komander, D. J. Med. 361, 1727–1735.
(2016). The deubiquitinase OTULIN is an essential negative regulator of inflam- Greil, J., Rausch, T., Giese, T., Bandapalli, O.R., Daniel, V., Bekeredjian-Ding,
mation and autoimmunity. Cell 166, 1215–1230. I., Stutz, A.M., Drees, C., Roth, S., Ruland, J., et al. (2013). Whole-exome
sequencing links caspase recruitment domain 11 (CARD11) inactivation to se-
Dierlamm, J., Baens, M., Wlodarska, I., Stefanova-Ouzounova, M., Hernan-
vere combined immunodeficiency. J. Allergy Clin. Immunol. 131, 1376–1383.
dez, J.M., Hossfeld, D.K., De Wolf-Peeters, C., Hagemeijer, A., Van den
Berghe, H., and Marynen, P. (1999). The apoptosis inhibitor gene API2 and a Guo, Y., Audry, M., Ciancanelli, M., Alsina, L., Azevedo, J., Herman, M., An-
novel 18q gene, MLT, are recurrently rearranged in the t(11;18)(q21;q21) guiano, E., Sancho-Shimizu, V., Lorenzo, L., Pauwels, E., et al. (2011). Herpes
associated with mucosa-associated lymphoid tissue lymphomas. Blood 93, simplex virus encephalitis in a patient with complete TLR3 deficiency: TLR3 is
3601–3609. otherwise redundant in protective immunity. J. Exp. Med. 208, 2083–2098.

Döffinger, R., Smahi, A., Bessia, C., Geissmann, F., Feinberg, J., Durandy, A., Hachmann, J., and Salvesen, G.S. (2016). The Paracaspase MALT1. Biochimie
Bodemer, C., Kenwrick, S., Dupuis-Girod, S., Blanche, S., et al. (2001). 122, 324–338.
X-linked anhidrotic ectodermal dysplasia with immunodeficiency is caused Hara, H., Wada, T., Bakal, C., Kozieradzki, I., Suzuki, S., Suzuki, N., Nghiem,
by impaired NF-kappaB signaling. Nat. Genet. 27, 277–285. M., Griffiths, E.K., Krawczyk, C., Bauer, B., et al. (2003). The MAGUK family
protein CARD11 is essential for lymphocyte activation. Immunity 18, 763–775.
Dondelinger, Y., Darding, M., Bertrand, M.J., and Walczak, H. (2016).
Poly-ubiquitination in TNFR1-mediated necroptosis. Cell. Mol. Life Sci. 73, Harden, J.L., Krueger, J.G., and Bowcock, A.M. (2015). The immunogenetics
2165–2176. of psoriasis: A comprehensive review. J. Autoimmun. 64, 66–73.

Duong, B.H., Onizawa, M., Oses-Prieto, J.A., Advincula, R., Burlingame, A., Hayden, M.S., and Ghosh, S. (2008). Shared principles in NF-kappaB
Malynn, B.A., and Ma, A. (2015). A20 restricts ubiquitination of pro-inter- signaling. Cell 132, 344–362.
leukin-1b protein complexes and suppresses NLRP3 inflammasome activity. Hayden, M.S., and Ghosh, S. (2012). NF-kB, the first quarter-century: Remark-
Immunity 42, 55–67. able progress and outstanding questions. Genes Dev. 26, 203–234.

54 Cell 168, January 12, 2017


Headon, D.J., Emmal, S.A., Ferguson, B.M., Tucker, A.S., Justice, M.J., (2013). Deep dermatophytosis and inherited CARD9 deficiency. N. Engl. J.
Sharpe, P.T., Zonana, J., and Overbeek, P.A. (2001). Gene defect in ecto- Med. 369, 1704–1714.
dermal dysplasia implicates a death domain adapter in development. Nature Lanternier, F., Mahdaviani, S.A., Barbati, E., Chaussade, H., Koumar, Y., Levy,
414, 913–916. R., Denis, B., Brunel, A.S., Martin, S., Loop, M., et al. (2015). Inherited CARD9
Herman, M., Ciancanelli, M., Ou, Y.H., Lorenzo, L., Klaudel-Dreszler, M., Pau- deficiency in otherwise healthy children and adults with Candida species-
wels, E., Sancho-Shimizu, V., Pérez de Diego, R., Abhyankar, A., Israelsson, induced meningoencephalitis, colitis, or both. J. Allergy Clin. Immunol. 135,
E., et al. (2012). Heterozygous TBK1 mutations impair TLR3 immunity and un- 1558–1568.
derlie herpes simplex encephalitis of childhood. J. Exp. Med. 209, 1567–1582. Lee, C.E., Fulcher, D.A., Whittle, B., Chand, R., Fewings, N., Field, M., An-
Hu, W.H., Pendergast, J.S., Mo, X.M., Brambilla, R., Bracchi-Ricard, V., Li, F., drews, D., Goodnow, C.C., and Cook, M.C. (2014). Autosomal-dominant
Walters, W.M., Blits, B., He, L., Schaal, S.M., and Bethea, J.R. (2005). NIBP, a B-cell deficiency with alopecia due to a mutation in NFKB2 that results in non-
novel NIK and IKK(beta)-binding protein that enhances NF-(kappa)B activa- processable p100. Blood 124, 2964–2972.
tion. J. Biol. Chem. 280, 29233–29241. Lemmers, B., Salmena, L., Bidère, N., Su, H., Matysiak-Zablocki, E., Mura-
Hubeau, M., Ngadjeua, F., Puel, A., Israel, L., Feinberg, J., Chrabieh, M., Be- kami, K., Ohashi, P.S., Jurisicova, A., Lenardo, M., Hakem, R., and Hakem,
lani, K., Bodemer, C., Fabre, I., Plebani, A., et al. (2011). New mechanism of A. (2007). Essential role for caspase-8 in Toll-like receptors and NFkappaB
X-linked anhidrotic ectodermal dysplasia with immunodeficiency: Impairment signaling. J. Biol. Chem. 282, 7416–7423.
of ubiquitin binding despite normal folding of NEMO protein. Blood 118, Lenardo, M., Pierce, J.W., and Baltimore, D. (1987). Protein-binding sites in Ig
926–935. gene enhancers determine transcriptional activity and inducibility. Science
Iwai, K. (2014). Diverse roles of the ubiquitin system in NF-kB activation. Bio- 236, 1573–1577.
chim. Biophys. Acta 1843, 129–136. Lenardo, M.J., Fan, C.M., Maniatis, T., and Baltimore, D. (1989). The involve-
Jabara, H.H., Ohsumi, T., Chou, J., Massaad, M.J., Benson, H., Megarbane, ment of NF-kappa B in beta-interferon gene regulation reveals its role as widely
A., Chouery, E., Mikhael, R., Gorka, O., Gewies, A., et al. (2013). A homozygous inducible mediator of signal transduction. Cell 57, 287–294.
mucosa-associated lymphoid tissue 1 (MALT1) mutation in a family with com- Lenz, G., Davis, R.E., Ngo, V.N., Lam, L., George, T.C., Wright, G.W., Dave,
bined immunodeficiency. J. Allergy Clin. Immunol. 132, 151–158. S.S., Zhao, H., Xu, W., Rosenwald, A., et al. (2008). Oncogenic CARD11 muta-
Jordan, C.T., Cao, L., Roberson, E.D., Duan, S., Helms, C.A., Nair, R.P., Duffin, tions in human diffuse large B cell lymphoma. Science 319, 1676–1679.
K.C., Stuart, P.E., Goldgar, D., Hayashi, G., et al. (2012). Rare and common Liu, F., Xia, Y., Parker, A.S., and Verma, I.M. (2012). IKK biology. Immunol. Rev.
variants in CARD14, encoding an epidermal regulator of NF-kappaB, in psori- 246, 239–253.
asis. Am. J. Hum. Genet. 90, 796–808. Lopez-Granados, E., Keenan, J.E., Kinney, M.C., Leo, H., Jain, N., Ma, C.A.,
Juilland, M., and Thome, M. (2016). Role of the CARMA1/BCL10/MALT1 com- Quinones, R., Gelfand, E.W., and Jain, A. (2008). A novel mutation in
plex in lymphoid malignancies. Curr. Opin. Hematol. 23, 402–409. NFKBIA/IKBA results in a degradation-resistant N-truncated protein and is
Kalay, E., Sezgin, O., Chellappa, V., Mutlu, M., Morsy, H., Kayserili, H., Kreiger, associated with ectodermal dysplasia with immunodeficiency. Hum. Mutat.
E., Cansu, A., Toraman, B., Abdalla, E.M., et al. (2012). Mutations in RIPK4 29, 861–868.
cause the autosomal-recessive form of popliteal pterygium syndrome. Am. Lougaris, V., Tabellini, G., Vitali, M., Baronio, M., Patrizi, O., Tampella, G.,
J. Hum. Genet. 90, 76–85. Biasini, A., Moratto, D., Parolini, S., and Plebani, A. (2015). Defective natural
Karin, M., and Lin, A. (2002). NF-kappaB at the crossroads of life and death. killer-cell cytotoxic activity in NFKB2-mutated CVID-like disease. J. Allergy
Nat. Immunol. 3, 221–227. Clin. Immunol. 135, 1641–1643.

Katakam, A.K., Brightbill, H., Franci, C., Kung, C., Nunez, V., Jones, C., 3rd, Ma, A., and Malynn, B.A. (2012). A20: Linking a complex regulator of ubiquity-
Peng, I., Jeet, S., Wu, L.C., Mellman, I., et al. (2015). Dendritic cells require lation to immunity and human disease. Nat. Rev. Immunol. 12, 774–785.
NIK for CD40-dependent cross-priming of CD8+ T cells. Proc. Natl. Acad. Ma, X., Becker Buscaglia, L.E., Barker, J.R., and Li, Y. (2011). MicroRNAs in
Sci. USA 112, 14664–14669. NF-kappaB signaling. J. Mol. Cell Biol. 3, 159–166.
Kere, J., Srivastava, A.K., Montonen, O., Zonana, J., Thomas, N., Ferguson, MacDuff, D.A., Reese, T.A., Kimmey, J.M., Weiss, L.A., Song, C., Zhang, X.,
B., Munoz, F., Morgan, D., Clarke, A., Baybayan, P., et al. (1996). X-linked an- Kambal, A., Duan, E., Carrero, J.A., Boisson, B., et al. (2015). Phenotypic
hidrotic (hypohidrotic) ectodermal dysplasia is caused by mutation in a novel complementation of genetic immunodeficiency by chronic herpesvirus infec-
transmembrane protein. Nat. Genet. 13, 409–416. tion. eLife 4, 4.
Keusekotten, K., Elliott, P.R., Glockner, L., Fiil, B.K., Damgaard, R.B., Kulathu, Mackay, F., and Cancro, M.P. (2006). Travelling with the BAFF/BLyS family:
Y., Wauer, T., Hospenthal, M.K., Gyrd-Hansen, M., Krappmann, D., et al. Are we there yet? Semin. Immunol. 18, 261–262.
(2013). OTULIN antagonizes LUBAC signaling by specifically hydrolyzing Massoumi, R., Chmielarska, K., Hennecke, K., Pfeifer, A., and Fässler, R.
Met1-linked polyubiquitin. Cell 153, 1312–1326. (2006). Cyld inhibits tumor cell proliferation by blocking Bcl-3-dependent
Kim, H.J., Hawke, N., and Baldwin, A.S. (2006). NF-kappaB and IKK as thera- NF-kappaB signaling. Cell 125, 665–677.
peutic targets in cancer. Cell Death Differ. 13, 738–747. Mathis, B.J., Lai, Y., Qu, C., Janicki, J.S., and Cui, T. (2015). CYLD-mediated
Kovalenko, A., Chable-Bessia, C., Cantarella, G., Israël, A., Wallach, D., and signaling and diseases. Curr. Drug Targets 16, 284–294.
Courtois, G. (2003). The tumour suppressor CYLD negatively regulates McDonald, D.R., Mooster, J.L., Reddy, M., Bawle, E., Secord, E., and Geha,
NF-kappaB signalling by deubiquitination. Nature 424, 801–805. R.S. (2007). Heterozygous N-terminal deletion of IkappaBalpha results in func-
Lafaille, F.G., Pessach, I.M., Zhang, S.Y., Ciancanelli, M.J., Herman, M., Ab- tional nuclear factor kappaB haploinsufficiency, ectodermal dysplasia, and im-
hyankar, A., Ying, S.W., Keros, S., Goldstein, P.A., Mostoslavsky, G., et al. mune deficiency. J. Allergy Clin. Immunol. 120, 900–907.
(2012). Impaired intrinsic immunity to HSV-1 in human iPSC-derived TLR3- Meylan, E., and Tschopp, J. (2005). The RIP kinases: Crucial integrators of
deficient CNS cells. Nature 491, 769–773. cellular stress. Trends Biochem. Sci. 30, 151–159.
Lahtela, J., Nousiainen, H.O., Stefanovic, V., Tallila, J., Viskari, H., Karikoski, Mina, R., Cerrato, C., Bernardini, A., Aghemo, E., and Palumbo, A. (2016). New
R., Gentile, M., Saloranta, C., Varilo, T., Salonen, R., and Kestilä, M. (2010). pharmacotherapy options for multiple myeloma. Expert Opin. Pharmacother.
Mutant CHUK and severe fetal encasement malformation. N. Engl. J. Med. 17, 181–192.
363, 1631–1637. Mir, A., Kaufman, L., Noor, A., Motazacker, M.M., Jamil, T., Azam, M., Kahrizi,
Lanternier, F., Pathan, S., Vincent, Q.B., Liu, L., Cypowyj, S., Prando, C., Mi- K., Rafiq, M.A., Weksberg, R., Nasr, T., et al. (2009). Identification of mutations
gaud, M., Taibi, L., Ammar-Khodja, A., Boudghene Stambouli, O., et al. in TRAPPC9, which encodes the NIK- and IKK-beta-binding protein, in

Cell 168, January 12, 2017 55


nonsyndromic autosomal-recessive mental retardation. Am. J. Hum. Genet. Price, S., Shaw, P.A., Seitz, A., Joshi, G., Davis, J., Niemela, J.E., Perkins, K.,
85, 909–915. Hornung, R.L., Folio, L., Rosenberg, P.S., et al. (2014). Natural history of auto-
Mochida, G.H., Mahajnah, M., Hill, A.D., Basel-Vanagaite, L., Gleason, D., Hill, immune lymphoproliferative syndrome associated with FAS gene mutations.
R.S., Bodell, A., Crosier, M., Straussberg, R., and Walsh, C.A. (2009). A trun- Blood 123, 1989–1999.
cating mutation of TRAPPC9 is associated with autosomal-recessive intellec- Rowe, A.M., Murray, S.E., Raué, H.P., Koguchi, Y., Slifka, M.K., and Parker,
tual disability and postnatal microcephaly. Am. J. Hum. Genet. 85, 897–902. D.C. (2013). A cell-intrinsic requirement for NF-kB-inducing kinase in CD4
Mooster, J.L., Le Bras, S., Massaad, M.J., Jabara, H., Yoon, J., Galand, C., and CD8 T cell memory. J. Immunol. 191, 3663–3672.
Heesters, B.A., Burton, O.T., Mattoo, H., Manis, J., and Geha, R.S. (2015). Ruefli-Brasse, A.A., French, D.M., and Dixit, V.M. (2003). Regulation of
Defective lymphoid organogenesis underlies the immune deficiency caused NF-kappaB-dependent lymphocyte activation and development by paracas-
by a heterozygous S32I mutation in IkBa. J. Exp. Med. 212, 185–202. pase. Science 302, 1581–1584.
Mousallem, T., Yang, J., Urban, T.J., Wang, H., Adeli, M., Parrott, R.E., Salmena, L., Lemmers, B., Hakem, A., Matysiak-Zablocki, E., Murakami, K.,
Roberts, J.L., Goldstein, D.B., Buckley, R.H., and Zhong, X.P. (2014). Au, P.Y., Berry, D.M., Tamblyn, L., Shehabeldin, A., Migon, E., et al. (2003).
A nonsense mutation in IKBKB causes combined immunodeficiency. Blood Essential role for caspase 8 in T-cell homeostasis and T-cell-mediated immu-
124, 2046–2050. nity. Genes Dev. 17, 883–895.
Neoptolemos, J.P., Stocken, D.D., Bassi, C., Ghaneh, P., Cunningham, D.,
Sancho-Shimizu, V., Pérez de Diego, R., Lorenzo, L., Halwani, R., Alangari, A.,
Goldstein, D., Padbury, R., Moore, M.J., Gallinger, S., Mariette, C., et al.; Eu-
Israelsson, E., Fabrega, S., Cardon, A., Maluenda, J., Tatematsu, M., et al.
ropean Study Group for Pancreatic Cancer (2010). Adjuvant chemotherapy
(2011). Herpes simplex encephalitis in children with autosomal recessive
with fluorouracil plus folinic acid vs gemcitabine following pancreatic cancer
and dominant TRIF deficiency. J. Clin. Invest. 121, 4889–4902.
resection: A randomized controlled trial. JAMA 304, 1073–1081.
Sasaki, K., and Iwai, K. (2015). Roles of linear ubiquitinylation, a crucial regu-
Ngo, V.N., Davis, R.E., Lamy, L., Yu, X., Zhao, H., Lenz, G., Lam, L.T., Dave, S.,
lator of NF-kB and cell death, in the immune system. Immunol. Rev. 266,
Yang, L., Powell, J., and Staudt, L.M. (2006). A loss-of-function RNA interfer-
175–189.
ence screen for molecular targets in cancer. Nature 441, 106–110.
Niemela, J., Kuehn, H.S., Kelly, C., Zhang, M., Davies, J., Melendez, J., Dreil- Schimke, L.F., Rieber, N., Rylaarsdam, S., Cabral-Marques, O., Hubbard, N.,
ing, J., Kleiner, D., Calvo, K., Oliveira, J.B., and Rosenzweig, S.D. (2015). Cas- Puel, A., Kallmann, L., Sombke, S.A., Notheis, G., Schwarz, H.P., et al. (2013).
pase-8 deficiency presenting as late-onset multi-organ lymphocytic infiltration A novel gain-of-function IKBA mutation underlies ectodermal dysplasia with
with granulomas in two adult siblings. J. Clin. Immunol. 35, 348–355. immunodeficiency and polyendocrinopathy. J. Clin. Immunol. 33, 1088–1099.

Nishikori, M. (2005). Classical and alternative NF-kB activation pathways and Schmidt-Supprian, M., Bloch, W., Courtois, G., Addicks, K., Israël, A., Rajew-
their roles in lymphoid malignancies. J. Clin. Exp. Hematop. 45, 9. sky, K., and Pasparakis, M. (2000). NEMO/IKK gamma-deficient mice model
incontinentia pigmenti. Mol. Cell 5, 981–992.
Noma, H., Eshima, K., Satoh, M., and Iwabuchi, K. (2015). Differential depen-
dence on nuclear factor-kB-inducing kinase among natural killer T-cell subsets Sen, R., and Baltimore, D. (1986). Multiple nuclear factors interact with the
in their development. Immunology 146, 89–99. immunoglobulin enhancer sequences. Cell 46, 705–716.
Ohnishi, H., Kawamoto, N., Seishima, M., Ohara, O., and Fukao, T. (2016). Sha, W.C., Liou, H.C., Tuomanen, E.I., and Baltimore, D. (1995). Targeted
A Japanese family case with juvenile onset Behçet’s disease caused by disruption of the p50 subunit of NF-kappa B leads to multifocal defects in
TNFAIP3 mutation. Allergol. Int. Published online: July 19, 2016. http://dx. immune responses. Cell 80, 321–330.
doi.org/10.1016/j.alit.2016.06.006.
Sharfe, N., Merico, D., Karanxha, A., Macdonald, C., Dadi, H., Ngan, B., Her-
Olivier, S., Close, P., Castermans, E., de Leval, L., Tabruyn, S., Chariot, A., brick, J.A., and Roifman, C.M. (2015). The effects of RelB deficiency on
Malaise, M., Merville, M.P., Bours, V., and Franchimont, N. (2006). Raloxi- lymphocyte development and function. J. Autoimmun. 65, 90–100.
fene-induced myeloma cell apoptosis: A study of nuclear factor-kappaB inhi-
Shigemura, T., Kaneko, N., Kobayashi, N., Kobayashi, K., Takeuchi, Y., Na-
bition and gene expression signature. Mol. Pharmacol. 69, 1615–1623.
kano, N., Masumoto, J., and Agematsu, K. (2016). Novel heterozygous
Pannicke, U., Baumann, B., Fuchs, S., Henneke, P., Rensing-Ehl, A., Rizzi, M., C243Y A20/TNFAIP3 gene mutation is responsible for chronic inflammation
Janda, A., Hese, K., Schlesier, M., Holzmann, K., et al. (2013). Deficiency of in autosomal-dominant Behçet’s disease. RMD Open 2, e000223.
innate and acquired immunity caused by an IKBKB mutation. N. Engl. J.
Med. 369, 2504–2514. Smahi, A., Courtois, G., Vabres, P., Yamaoka, S., Heuertz, S., Munnich, A.,
Israël, A., Heiss, N.S., Klauck, S.M., Kioschis, P., et al.; The International Incon-
Pasparakis, M., Courtois, G., Hafner, M., Schmidt-Supprian, M., Nenci, A.,
tinentia Pigmenti (IP) Consortium (2000). Genomic rearrangement in NEMO
Toksoy, A., Krampert, M., Goebeler, M., Gillitzer, R., Israel, A., et al. (2002).
impairs NF-kappaB activation and is a cause of incontinentia pigmenti. Nature
TNF-mediated inflammatory skin disease in mice with epidermis-specific dele-
405, 466–472.
tion of IKK2. Nature 417, 861–866.
Smale, S.T. (2012). Dimer-specific regulatory mechanisms within the NF-kB
Pasqualucci, L., and Dalla-Favera, R. (2015). The genetic landscape of diffuse
family of transcription factors. Immunol. Rev. 246, 193–204.
large B-cell lymphoma. Semin. Hematol. 52, 67–76.
Peltzer, N., Rieser, E., Taraborrelli, L., Draber, P., Darding, M., Pernaute, B., Snow, A.L., Xiao, W., Stinson, J.R., Lu, W., Chaigne-Delalande, B., Zheng, L.,
Shimizu, Y., Sarr, A., Draberova, H., Montinaro, A., et al. (2014). HOIP defi- Pittaluga, S., Matthews, H.F., Schmitz, R., Jhavar, S., et al. (2012). Congenital
ciency causes embryonic lethality by aberrant TNFR1-mediated endothelial B cell lymphocytosis explained by novel germline CARD11 mutations. J. Exp.
cell death. Cell Rep. 9, 153–165. Med. 209, 2247–2261.

Pérez de Diego, R., Sancho-Shimizu, V., Lorenzo, L., Puel, A., Plancoulaine, S., Stepensky, P., Keller, B., Buchta, M., Kienzler, A.K., Elpeleg, O., Somech, R.,
Picard, C., Herman, M., Cardon, A., Durandy, A., Bustamante, J., et al. (2010). Cohen, S., Shachar, I., Miosge, L.A., Schlesier, M., et al. (2013). Deficiency of
Human TRAF3 adaptor molecule deficiency leads to impaired Toll-like recep- caspase recruitment domain family, member 11 (CARD11), causes profound
tor 3 response and susceptibility to herpes simplex encephalitis. Immunity 33, combined immunodeficiency in human subjects. J. Allergy Clin. Immunol.
400–411. 131, 477–485.
Perkins, N.D. (2012). The diverse and complex roles of NF-kB subunits in can- Su, H.C., and Lenardo, M.J. (2008). Genetic defects of apoptosis and primary
cer. Nat. Rev. Cancer 12, 121–132. immunodeficiency. Immunol. Allergy Clin. North Am. 28, 329–351.
Picard, C., Puel, A., Bonnet, M., Ku, C.L., Bustamante, J., Yang, K., Soudais, Su, H., Bidère, N., Zheng, L., Cubre, A., Sakai, K., Dale, J., Salmena, L., Ha-
C., Dupuis, S., Feinberg, J., Fieschi, C., et al. (2003). Pyogenic bacterial infec- kem, R., Straus, S., and Lenardo, M. (2005). Requirement for caspase-8 in
tions in humans with IRAK-4 deficiency. Science 299, 2076–2079. NF-kappaB activation by antigen receptor. Science 307, 1465–1468.

56 Cell 168, January 12, 2017


Sugiura, K. (2014). The genetic background of generalized pustular psoriasis: Wieacker, P., Zimmer, J., and Ropers, H.H. (1985). X inactivation patterns in
IL36RN mutations and CARD14 gain-of-function variants. J. Dermatol. Sci. 74, two syndromes with probable X-linked dominant, male lethal inheritance.
187–192. Clin. Genet. 28, 238–242.
Sun, S.C. (2011). Non-canonical NF-kB signaling pathway. Cell Res. 21,  u, F., Santos-Valente, E., Garncarz, W., Bilic, I.,
Willmann, K.L., Klaver, S., Dog
71–85. Mace, E., Salzer, E., Conde, C.D., Sic, H., et al. (2014). Biallelic loss-of-function
Sun, S.C. (2012). The noncanonical NF-kB pathway. Immunol. Rev. 246, mutation in NIK causes a primary immunodeficiency with multifaceted aber-
125–140. rant lymphoid immunity. Nat. Commun. 5, 5360.
Tanaka, M., Fuentes, M.E., Yamaguchi, K., Durnin, M.H., Dalrymple, S.A., Wisniewski, S.A., and Trzeciak, W.H. (2012a). A new mutation resulting in the
Hardy, K.L., and Goeddel, D.V. (1999). Embryonic lethality, liver degeneration, truncation of the TRAF6-interacting domain of XEDAR: A possible novel cause
and impaired NF-kappa B activation in IKK-beta-deficient mice. Immunity 10, of hypohidrotic ectodermal dysplasia. J. Med. Genet. 49, 499–501.
421–429. Wisniewski, S.A., and Trzeciak, W.H. (2012b). A rare heterozygous TRAF6
Tokunaga, F. (2013). Linear ubiquitination-mediated NF-kB regulation and its variant is associated with hypohidrotic ectodermal dysplasia. Br. J. Dermatol.
related disorders. J. Biochem. 154, 313–323. 166, 1353–1356.
Torres, J.M., Martinez-Barricarte, R., Garcı́a-Gómez, S., Mazariegos, M.S., Xue, L., Morris, S.W., Orihuela, C., Tuomanen, E., Cui, X., Wen, R., and Wang,
Itan, Y., Boisson, B., Rholvarez, R., Jiménez-Reinoso, A., del Pino, L., Rodrı́- D. (2003). Defective development and function of Bcl10-deficient follicular,
guez-Pena, R., et al. (2014). Inherited BCL10 deficiency impairs hematopoietic marginal zone and B1 B cells. Nat. Immunol. 4, 857–865.
and nonhematopoietic immunity. J. Clin. Invest. 124, 5239–5248. Yin, L., Wu, L., Wesche, H., Arthur, C.D., White, J.M., Goeddel, D.V., and
Trzeciak, W.H., and Koczorowski, R. (2016). Molecular basis of hypohidrotic Schreiber, R.D. (2001). Defective lymphotoxin-beta receptor-induced
ectodermal dysplasia: An update. J. Appl. Genet. 57, 51–61. NF-kappaB transcriptional activity in NIK-deficient mice. Science 291,
Tsoi, L.C., Spain, S.L., Knight, J., Ellinghaus, E., Stuart, P.E., Capon, F., Ding, 2162–2165.
J., Li, Y., Tejasvi, T., Gudjonsson, J.E., et al.; Collaborative Association Study Young, R.M., Shaffer, A.L., 3rd, Phelan, J.D., and Staudt, L.M. (2015). B-cell
of Psoriasis (CASP); Genetic Analysis of Psoriasis Consortium; Psoriasis receptor signaling in diffuse large B-cell lymphoma. Semin. Hematol. 52,
Association Genetics Extension; Wellcome Trust Case Control Consortium 2 77–85.
(2012). Identification of 15 new psoriasis susceptibility loci highlights the role Yu, J.W., Hoffman, S., Beal, A.M., Dykon, A., Ringenberg, M.A., Hughes, A.C.,
of innate immunity. Nat. Genet. 44, 1341–1348. Dare, L., Anderson, A.D., Finger, J., Kasparcova, V., et al. (2015). MALT1 Pro-
Vande Walle, L., Van Opdenbosch, N., Jacques, P., Fossoul, A., Verheugen, tease Activity Is Required for Innate and Adaptive Immune Responses. PLoS
E., Vogel, P., Beyaert, R., Elewaut, D., Kanneganti, T.D., van Loo, G., and Lam- ONE 10, e0127083.
kanfi, M. (2014). Negative regulation of the NLRP3 inflammasome by A20 Zhang, J., Stirling, B., Temmerman, S.T., Ma, C.A., Fuss, I.J., Derry, J.M., and
protects against arthritis. Nature 512, 69–73. Jain, A. (2006). Impaired regulation of NF-kappaB and increased susceptibility
von Bernuth, H., Picard, C., Jin, Z., Pankla, R., Xiao, H., Ku, C.L., Chrabieh, M., to colitis-associated tumorigenesis in CYLD-deficient mice. J. Clin. Invest.
Mustapha, I.B., Ghandil, P., Camcioglu, Y., et al. (2008). Pyogenic bacterial in- 116, 3042–3049.
fections in humans with MyD88 deficiency. Science 321, 691–696. Zhou, Q., Wang, H., Schwartz, D.M., Stoffels, M., Park, Y.H., Zhang, Y., Yang,
Wade, E.M., Daniel, P.B., Jenkins, Z.A., McInerney-Leo, A., Leo, P., Morgan, D., Demirkaya, E., Takeuchi, M., Tsai, W.L., et al. (2016). Loss-of-function mu-
T., Addor, M.C., Adès, L.C., Bertola, D., Bohring, A., et al. (2016). Mutations tations in TNFAIP3 leading to A20 haploinsufficiency cause an early-onset
in MAP3K7 that Alter the Activity of the TAK1 Signaling Complex Cause Fron- autoinflammatory disease. Nat. Genet. 48, 67–73.
tometaphyseal Dysplasia. Am. J. Hum. Genet. 99, 392–406. Zong, M., Wu, X.G., Chan, C.W., Choi, M.Y., Chan, H.C., Tanner, J.A., and Yu,
Weiss, K., Applegate, C., Wang, T., and Batista, D.A. (2015). Familial TAB2 mi- S. (2011). The adaptor function of TRAPPC2 in mammalian TRAPPs explains
crodeletion and congenital heart defects including unusual valve dysplasia and TRAPPC2-associated SEDT and TRAPPC9-associated congenital intellectual
tetralogy of fallot. Am. J. Med. Genet. A. 167A, 2702–2706. disability. PLoS ONE 6, e23350.

Cell 168, January 12, 2017 57

Вам также может понравиться