Integrating cell-signalling pathways with NF- κB and IKK function

Page created by Daniel Brewer
 
CONTINUE READING
REVIEWS

                               Integrating cell-signalling pathways
                               with NF-κB and IKK function
                               Neil D. Perkins
                               Abstract | Nuclear factor (NF)-κB and inhibitor of NF-κB kinase (IKK) proteins regulate many
                               physiological processes, including the innate- and adaptive-immune responses, cell death
                               and inflammation. Disruption of NF-κB or IKK function contributes to many human diseases,
                               including cancer. However, the NF-κB and IKK pathways do not exist in isolation and there
                               are many mechanisms that integrate their activity with other cell-signalling networks.
                               This crosstalk constitutes a decision-making process that determines the consequences
                               of NF-κB and IKK activation and, ultimately, cell fate.

                              Nuclear factor (NF)-κB transcription factors can both        inflammatory diseases and cancers. The development of
                              induce and repress gene expression by binding to             NF-κB and IKK inhibitors for the treatment of inflamma-
                              discrete DNA sequences, known as κB elements, in             tory diseases and cancers, together with the observation
                              promoters and enhancers1. In mammalian cells, there          that many drugs currently in clinical use have significant
                              are five NF-κB family members, RelA (p65), RelB, c-Rel,      effects on NF-κB activity, means that understanding the
                              p50/p105 (NF-κB1) and p52/p100 (NF-κB2) (FIG. 1), and        pathways that regulate NF-κB function will have many
                              different NF-κB complexes are formed from their homo-        diagnostic and prognostic applications6–9.
                              and heterodimers. In most cell types, NF-κB complexes
                              are retained in the cytoplasm by a family of inhibitory      NF-κ κB complexes
                              proteins known as inhibitors of NF-κB (IκBs) (FIG. 1).       All NF-κB family members contain an N-terminal
                              Activation of NF-κB typically involves the phosphory-        domain of approximately 300 amino acids called the
                              lation of IκB by the IκB kinase (IKK) complex, which         Rel-homology domain (RHD), which mediates DNA
                              results in IκB degradation. This releases NF-κB and          binding and dimerization. Furthermore, Rel subfamily
                              allows it to translocate freely to the nucleus1 (BOX 1).     members, RelA, RelB and c-Rel, all contain unrelated
                                  The genes regulated by NF-κB include those con-          C-terminal transcriptional activation domains (FIG. 1).
                              trolling programmed cell death (apoptosis), cell adhe-       Despite obvious structural similarities and their ability to
                              sion, proliferation, the innate- and adaptive-immune         bind related DNA sequences, genetic studies have shown
                              responses, inflammation, the cellular-stress response        that all NF-κB subunits have distinct and non-overlap-
                              and tissue remodelling1–5. However, the expression of        ping functions3,10. In most unstimulated, non-diseased
                              these genes is tightly coordinated with the activity of      mammalian cells, Rel subunits are found predominantly
                              many other signalling and transcription-factor pathways.     in the cytoplasm bound to a member of the IκB family
                              Therefore, the outcome of NF-κB activation depends on        of inhibitory proteins1. In mammalian cells, there are
                              the nature and the cellular context of its induction. For    three principal IκBs, IκBα, IκBβ and IκBε, which func-
                              example, it has become apparent that NF-κB activity can      tion in part by masking a conserved nuclear localization
                              be regulated by both oncogenes and tumour suppressors,       sequence (NLS) that is found in the RHD of the NF-κB
                              resulting in either stimulation or inhibition of apoptosis   subunits. For IκBα, this NLS masking is only partially
                              and proliferation6. Alternatively, NF-κB regulation of the   effective and NF-κB–IκBα complexes shuttle into the
College of Life Sciences,
                              Jun N-terminal kinase (JNK)-signalling pathway can have      nucleus even in the absence of cellular stimulation1.
Division of Gene Regulation
and Expression, James Black   opposing results that depend on the cell type (discussed     However, IκBα also contains a nuclear export sequence
Centre, Dow Street,           below). This review discusses the mechanisms that link       (NES), which causes the rapid export of such complexes
University of Dundee,         the NF-κB pathway to other cell-signalling pathways          back to the cytoplasm1.
Dundee, DD1 5EH,              and asks why integration of NF-κB function is required           Both p50 and p52 are synthesized as longer precursor
Scotland, UK.
e-mail:
                              for the specific regulation of its target genes. Defects     proteins, p105 and p100, that contain ankyrin-repeat
n.d.perkins@dundee.ac.uk      in these mechanisms can lead to inappropriate NF-κB          motifs in their C termini, similar to those found in IκB
doi:10.1038/nrm2083           or IKK activity and can contribute to the pathology of       proteins (FIG. 1). p100 and p105 also function as IκB-like

NATURE REVIEWS | MOLECULAR CELL BIOLOGY                                                                              VOLUME 8 | JANUARY 2007 | 49
                                                      © 2007 Nature Publishing Group
REVIEWS

a The NF-κB family                                                                                                               and p52 can function as nuclear transcription factors.
                                                                                                                                 Both p50 and p52 homodimers can interact with
RelA    1                      RHD                                          TA2         TA1      551
(p65)
                                                                                                                                 another IκB family member, BCL-3, which, unlike the
                19                                      301     428                 521                                          other IκBs, is nuclear and functions as a transcriptional
                                                                                                                                 co-activator1.
RelB    1       LZ                    RHD                                     TAD                557
                                                                                                                                     Homodimers composed of the p50 and p52 NF-κB
                                                                410
                                                                                                                                 subunits evade regulation by IκBs and, consequently, are
c-Rel   1                      RHD                                                  TA1        TA2      587                      often found constitutively in the nucleus. However, when
            8                                       290                           422 497                                        p50 and p52 form heterodimers with the Rel subunits,
                                                                                        518                                      these subunits are also subject to cytoplasmic retention
p105    1                      RHD                                                 ANK                    DD               969
(p50)                                                                                                                            by IκBs1. The exception to this is the p52–RelB complex,
                43                                        363         543                            751 818 893                 which has a low affinity for IκBα and can therefore evade
p100    1                      RHD                                                ANK                   DD           900         this mechanism of regulation11.
(p52)           38                                        338     487                               699   851
                                                                                                                                     There are several distinct NF-κB-activation path-
                                                                                                      776                        ways. The most frequently observed is the canonical,
b The IκB family                                                                                                                 or classical, pathway (BOX 1) , which is induced in
                               ANK                                                                                               response to various inflammatory stimuli, including the
IκBα    1                                                PEST     317                                                            pro-inflammatory cytokines tumour necrosis factor-α
                          73                      256                                                                            (TNFα) and interleukin-1 (IL-1), engagement of the
                                                    264
IκBβ    1                                                     PEST      361
                                                                                                                                 T-cell receptor (TCR) or exposure to bacterial products
                                                                                                                                 such as lipopolysaccharide (LPS)1. This pathway is typi-
                      57                                298
                                                          307                                                                    fied by the rapid phosphorylation of IκBα at Ser32 and
IκBε    1                                                                                     500                                Ser36 and subsequent ubiquitin-induced degradation by
                                                  258                                   473                                      the 26S proteasome1. In many cell types, IκBβ and IκBε
                                                                                                                                 are also subject to phosphorylation and degradation, but
BCL-3 1                                                                                 446                                      with slower kinetics1,12.
                                126                               351                                                                In the canonical pathway, IκB phosphorylation is due
                                                                                                                                 to IKK-complex activation1. The IKK complex consists
c The IKK family                                                                                                                 of three core subunits, the catalytic subunits IKKα and
                                                                                                                                 IKKβ (also known as IKK1 and IKK2) and several copies
NEMO 1                          CC1                     CC2       LZ         ZF     419
(IKKγ)                                                                                                                           of a regulatory subunit called the NF-κB essential modi-
                     63                     193     258          319 397                                                         fier (NEMO, also known as IKKγ)1. Genetic experiments
                                                              296 346    417
                                                                                                                                 have shown that IKKβ is the predominant IκB kinase in
IKKα    1                  Kinase domain                                LZ                HLH          NBD     745               the canonical pathway2,4.
                15                                      301            455 483          599 638 738 743                              A subset of NF-κB-induction stimuli, such as
                                                                                                                                 stimulation of the CD40 and lymphotoxin-β receptors,
IKKβ    1                  Kinase domain                                LZ                 HLH         NBD 756                   B-cell-activating factor of the TNF family (BAFF), LPS
                15                                  300                458 486            603 642 737 742                        and latent membrane protein-1 (LMP1) of Epstein–
Figure 1 | The mammalian members of the NF-κB, IκB and IKK families.                                                             Barr virus, activate the non-canonical, or alternative,
a | In mammalian cells, there are five nuclear factor (NF)-κB family members, RelA (p65),                                        pathway2,13. Here, activation of IKKα by the NF-κB-
RelB, c-Rel, p50/p105 (NF-κB1) and p52/p100 (NF-κB2). p50 and p52 (not shown) are                                                inducing kinase (NIK) results in the formation of
derived from the longer precursor proteins p105 and p100, respectively. All NF-κB family                                         p52 from p100, as a consequence of phosphorylation-
members contain an N-terminal Rel-homology domain (RHD) that mediates DNA
                                                                                                                                 induced, ubiquitin-dependent processing of p100 by the
binding and dimerization and contains the nuclear-localization domain. The Rel
subfamily, RelA, RelB and c-Rel, contain unrelated C-terminal transcriptional activation                                         26S proteasome2,12. p52–RelB heterodimers, which are
domains (TADs). TA1 and TA2 are subdomains of the RelA transactivation domain.                                                   frequently activated as a consequence of non-canonical
b | The inhibitor of NF-κB (IκB) family consists of IκBα, IκBβ, IκBε and BCL-3. Like p105                                        pathway activation, have a higher affinity for distinct κB
and p100, the IκB proteins contain ankyrin-repeat motifs (ANK) in their C termini. c | The                                       elements and might therefore regulate a distinct subset
three core subunits of the IκB kinase (IKK) complex are shown: the catalytic subunits                                            of NF-κB target genes (BOX 1). It has been suggested
IKKα and IKKβ and the regulatory subunit called the NF-κB essential modifier (NEMO,                                              that the non-canonical pathway is regulated by IKKα
also known as IKKγ). The principal structural motifs of each protein are shown, together                                         homodimers that function independently of the larger
with amino-acid numbers corresponding to the human proteins, although some                                                       IKK complex2. The non-canonical pathway does not
definitions of where a domain begins and ends might differ between publications.                                                 lead to the formation of p50 from p105 and, in many
CC, coiled-coil; DD, region with homology to a death domain; HLH, helix–loop–helix;
                                                                                                                                 cells, high levels of p50 are created in the absence of
LZ, RelB-transactivation-domain containing a putative leucine-zipper-like motif;
NBD, NEMO-binding domain; PEST, domain rich in proline (P), glutamate (E), serine (S)                                            cell stimulation by a co-translational mechanism. In this
and threonine (T); ZF, zinc-finger domain.                                                                                       co-translational mechanism, p50 is generated during
                                                                                                                                 mRNA translation as a consequence of proteolysis of
                                                                                                                                 the nascent p105 polypeptide by the 26S proteasome14,
                                            proteins and retain their NF-κB-subunit dimeric                                      although other mechanisms of p50 generation have
                                            partners in the cytoplasm, inhibiting their activity1.                               also been proposed15,16. However, IKKβ-inducible and
                                            Processing of p100 and p105 can occur through several                                ubiquitin-dependent total degradation of p105, without
                                            mechanisms (discussed below) and is required before p50                              p50 generation, can also occur15.

50 | JANUARY 2007 | VOLUME 8                                                                                                                          www.nature.com/reviews/molcellbio
                                                                                  © 2007 Nature Publishing Group
REVIEWS

                                   Box 1 | Pathways leading to the activation of NF-κB
                                                                                              Atypical pathway                                             Non-canonical pathway
                                                                                              (IKK independent)                                            (LPS, CD40 and lymphotoxin
                                                                                    Hypoxia, H2O2            UV, HER2/Neu                                  receptors, LMP1)
                                                                                      Tyr kinase                        CK2                                               NIK

                                                                                                               TA elA

                                                                                                                                                     lA
                                                                                                       P

                                                                                                                 D

                                                                                                                                                   Re
                                                                                                                 R
                                                                                             (Tyr42)
                                                              Canonical                                                                                            IKKα IKKβ
                                                                                                  p50     RelA
                                                              pathway                                                                 RelA
                                                              (TNFα, IL-1, LPS)                   RHD IκBα RHD                p50
                                                                                                                                  IκBα
                                                                    NEMO

                                                                                                                                                                  Re
                                                                                                                                    P P

                                                                                                                                                                    lB
                                                                  IKKα IKKβ                       Degradation or           Degradation                                          P
                                                                                                  dissociation of IκB      of IκB

                                                                                                                                                                     p1 NK
                                                                                                                                                                                    P

                                                                                                                                                                       00
                                                                                                                                                                       A
                                                                                    (Ser32)                                                                      p100               (Ser866)
                                                                                                  P                                                              RHD RelB           (Ser870)

                                                                                                          lA
                                                    Activation of                             P

                                                                                                        Re
                                                    IKK complex                   (Ser36)
                                                                                                  RelA                                               Ubiquitylation
                                                                                     p50                                                             and proteasomal
                                                                             Ub               IκBα
                                                                                                                                                     processing of p100
                                    Atypical pathway                NEMO                    Ubiquitylation
                                    (genotoxic stress,                                      and proteasomal

                                                                                                                                                                  Re AD
                                                                       ATM

                                                                                                                              lA

                                                                                                                                                                    lB
                                                                                                                                                                    T
                                                                                            degradation of IκB

                                                                                                                            Re
                                    IKK dependent)
                                         NEMO
                                                                                                                  p50 RelA                                       p52 RelB
                                                                                        Cytoplasm                                                                    RHD

                                                                                        Nucleus                         Kinases,
                                                                                                                        acetylases,
                                                  SUMO                       Ub                                         phosphatases
                                          NEMO                      NEMO
                                           ATM                         ATM

                                                                                                                                       Zn-finger TFs

                                                                                                                                            bZIP

                                                                                                                                                   bZIP
                                                                                                                           HMG-I
                                                         Co-activator             Co-repressor
                                                                                              P
                                                              P                           P                                        P
                                                                   P                    P

                                                                                                                                                                  Re
                                                         lA

                                                                                                  lA

                                                                                                                                       lA

                                                                                                                                                                    lB
                                                    Re

                                                                                              Re

                                                                                                                                   Re

                                                    Ac
                                           p50 RelA                               p50 RelA                               p50 RelA                                p52 RelB
                                                    Ac                                                                                      TF

                                      Transcriptional                        Transcriptional                      Promoter targeting                       Distinct κB elements
                                      activation                             repression                           and selectivity

                                   The canonical pathway is induced by tumour necrosis factor-α (TNFα), interleukin-1 (IL-1) and many other stimuli, and is
                                   dependent on activation of IKKβ. This activation results in the phosphorylation (P) of IκBα at Ser32 and Ser36, leading to its
                                   ubiquitylation (Ub) and subsequent degradation by the 26S proteasome. Release of the NF-κB complex allows it to relocate to
                                   the nucleus. Under some circumstances, the NF-κB–IκBα complex shuttles between the cytoplasm and the nucleus (not
                                   shown). IKK-dependent activation of NF-κB can occur following genotoxic stress. Here, NF-κB essential modifier (NEMO)
                                   localizes to the nucleus, where it is sumoylated and then ubiquitylated, in a process that is dependent on the ataxia
Co-activators and                  telangiectasia mutated (ATM) checkpoint kinase. NEMO relocates back to the cytoplasm together with ATM, where activation
co-repressors                      of IKKβ occurs. IKK-independent atypical pathways of NF-κB activation have also been described, which include casein
Transcriptional regulatory         kinase-II (CK2) and tyrosine-kinase-dependent pathways. The non-canonical pathway results in the activation of IKKα by the
proteins that are typically        NF-κB-inducing kinase (NIK), followed by phosphorylation of the p100 NF-κB subunit by IKKα. This results in proteasome-
recruited to promoters and         dependent processing of p100 to p52, which can lead to the activation of p52–RelB heterodimers that target distinct
enhancers by interacting with      κB elements. Phosphorylation of NF-κB subunits by nuclear kinases, and modification of these subunits by acetylases and
DNA-bound transcription            phosphatases, can result in transcriptional activation and repression as well as promoter-specific effects. Moreover,
factors. Co-activators stimulate
                                   cooperative interactions with heterologous transcription factors can target NF-κB complexes to specific promoters, resulting
transcription whereas
co-repressors do the opposite.
                                   in the selective activation of gene expression following cellular exposure to distinct stimuli. Ac, acetylation; bZIP, leucine-
Co-activators are frequently       zipper-containing transcription factor; HMG-I, high-mobility-group protein-I; IκB, inhibitor of κB; IKK, IκB kinase;
histone acetyltransferases         LMP1, latent membrane protein-1; LPS, lipopolysaccharide; NF-κB, nuclear factor-κB; RHD, Rel-homology domain;
whereas co-repressors are          TAD, transcriptional activation domain; TF, transcription factor; UV, ultraviolet; Zn-finger TF, zinc-finger-containing
often histone deacetylases.        transcription factor. This figure is modified with permission from REF. 6 © (2006) Macmillan Publishers Ltd.

NATURE REVIEWS | MOLECULAR CELL BIOLOGY                                                                                                             VOLUME 8 | JANUARY 2007 | 51
                                                                  © 2007 Nature Publishing Group
REVIEWS

                                    IKK-complex activation                                          IKK-independent pathways have the potential to
                                    Numerous NF-κB-activating stimuli, which frequently             induce differentially modified forms of NF-κB subunits
                                    engage distinct cell-surface receptors and/or cytoplasmic       with distinct functions.
                                    signalling pathways, converge on the IKK complex
                                    and induce the canonical pathway1,12,17–19. The key to          NF-κκ B-independent effects of the IKKs
                                    activation of the IKK complex is the regulatory subunit         IKK activation does more than merely induce IκB deg-
                                    NEMO. Activation of the canonical pathway results               radation and the release of NF-κB. IKK subunits also
                                    in complex, Lys63-linked ubiquitylation of signal-              programme the cellular response to the initial activating
                                    ling molecules, ultimately resulting in Lys63-linked            stimulus (FIGS 2,3). In part, this can occur through post-
                                    ubiquitylation of NEMO (discussed in more detail in             translational modification of NF-κB subunits, such as
                                    REFS 12,18–20). In contrast to Lys48-linked ubiquityl-          RelA, which helps to define which genes are induced
                                    ation, which results in protein degradation by the 26S          or repressed by the NF-κB complexes6,12. However,
                                    proteasome, Lys63-linked ubiquitylation has a role in           several NF-κB- and IκB-independent targets of IKKα
                                    signalling by facilitating interactions with proteins that      and IKKβ are now being identified, which reveals that
                                    contain ubiquitin-binding domains12,18–20. Ubiquitylation       activation of the IKKs could result in more widespread
                                    of NEMO allows the recruitment of kinases — such                effects on cell signalling than previously realized.
                                    as transforming growth factor-β (TGFβ)-activated                Many of these NF-κB-independent functions of IKKs
                                    kinase-1 (TAK1), which phosphorylates IKKβ in its               have only been recently discovered, and these putative
                                    activation loop at Ser177 and Ser181 — to the IKK               roles for IKKs therefore require further confirmation.
                                    complex 12,18–20. However, a mouse knockout of the              Nonetheless, there is a clear trend towards recognizing
                                    E2 ubiquitin-conjugating enzyme UBC13, which selec-             a more promiscuous role for IKKs, and it is likely that
                                    tively stimulates Lys63-linked ubiquitylation, has cast         many more substrates have yet to be discovered.
                                    doubt on whether ubiquitylation of NEMO is required
                                    for NF-κB activation under all circumstances. Instead,          The consequences of IKKβ activation. Activation of
                                    this study indicated that NEMO ubiquitylation might be          IKKβ stimulates anti-apoptotic, pro-inflammatory
                                    required for the activation of mitogen-activated protein        and proliferative pathways 24,25 (FIG. 2) , although
                                    (MAP)-kinase signalling21 (see below).                          pro-apoptotic functions have also been described26,27.
                                        Other mechanisms of canonical pathway activation                 Consistent with its anti-apoptotic role, IKKβ has
                                    — for example, by genotoxic stimuli, such as ionizing           been shown to phosphorylate and inhibit FOXO3a, a
                                    radiation, or by some chemotherapeutic drugs — also             transcription factor and tumour suppressor that can
                                    result in NEMO-dependent IKKβ activation22. Here, in            induce either apoptosis or cell-cycle arrest28. However,
                                    some contexts, NEMO first translocates to the nucleus,          such effects might be context dependent as, conversely,
                                    where it is sumoylated and then phosphorylated by               IKKβ has also been found to activate FOXO3a in
                                    the checkpoint kinase ataxia telangiectasia mutated             regressing mammary glands, leading to upregulation
                                    (ATM)23 (BOX 1). NEMO sumoylation is then replaced              of the death-receptor ligand TWEAK and induction of
                                    by mono-ubiquitylation, leading to the nuclear export           apoptosis27. Whether these latter effects involve direct
Checkpoint kinases                  of NEMO as a complex with ATM. The NEMO–ATM                     phosphorylation of FOXO3a was not investigated.
The checkpoint kinases ATM          complex subsequently activates the IKK complex in a                  IKKβ can also phosphorylate 14-3-3β when this
and ATR are differentially          process that requires the IKK-associated protein ELKS23         protein is complexed with tristetraprolin (TPP), an
activated in response to            (a protein that is rich in glutamate (E), leucine (L), lysine   AU-rich element (ARE) binding protein that regulates
distinct forms of genotoxic
stress. Generally, ATR and ATM
                                    (K) and serine (S)). Whether this pathway is activated          mRNA stability 29. IKKβ phosphorylation inhibits
activate the downstream             in response to all genotoxic stresses, in all cell types, is    TPP–14-3-3β ARE binding and might therefore pro-
checkpoint kinases CHK1 and         currently unclear. Nonetheless, this serves as an illustra-     mote the stability of cytokine, chemokine and growth
CHK2, respectively. Together        tion of how IKK activity and, consequently, induction           factor mRNAs, which all contain the ARE motif 29.
these kinases help orchestrate
                                    of NF-κB DNA binding, can be integrated with parallel                Pro-inflammatory cytokines, such as TNFα and IL-1,
the cellular response to DNA
damage.                             signalling pathways.                                            can lead to insulin resistance and the development of
                                        Atypical, IKK-independent pathways of NF-κB                 type-2 diabetes. This effect can, in part, be attributed
14-3-3 proteins                     induction also provide mechanisms to integrate par-             to inhibition of insulin signalling that results from
A large eukaryotic class of         allel signalling pathways with NF-κB activity (BOX 1).          IKKβ phosphorylation of insulin-receptor-substrate-1
proteins that are involved in
cell division, apoptosis, signal
                                    For example, some stimuli, such as hypoxia and                  (IRS1)30–32.
transduction, transmitter           reoxygenation, hydrogen-peroxide stimulation and                     IKKβ can also participate in a negative feedback
release, receptor function,         treatment of cells with nerve growth factor (NGF)               loop that downregulates the signalling pathways that
gene expression and enzyme          or the tyrosine-phosphatase inhibitor pervanadate               lead to its activation. The TCR induces IKKβ through
activation. They function by
                                    result in the phosphorylation of IκBα at Tyr42, which           activation of the Carma1–BCL10–MALT1 (CBM) com-
binding to various different,
specific target proteins, usually   results in either its degradation or dissociation from          plex. IKKβ is required for the induction of the CBM
in response to phosphorylation      NF-κB6,12. Treatment with ultraviolet (UV) light, or            complex through an ill-defined mechanism. However,
of these targets.                   expression of the HER2 (also known as erbB-2 or Neu)            it is also necessary for the attenuation of CBM-complex
                                    oncogene in breast cancer cells, can result in IκBα             signalling. This attenuation effect results from IKKβ
Type-2 diabetes
Diabetes that results from
                                    phosphorylation by casein kinase-II (CK2) at sites              directly phosphorylating BCL10, which disrupts its
insulin resistance or from          in its C-terminal PEST domain12. As activated IKKα              association with MALT1, thereby inactivating the CBM
reduced production of insulin.      and IKKβ also phosphorylate RelA (see below), these             complex33.

52 | JANUARY 2007 | VOLUME 8                                                                                             www.nature.com/reviews/molcellbio
                                                             © 2007 Nature Publishing Group
REVIEWS

                                                                                                                                                   Cell proliferation
                                 Disruption of BCL10–MALT1
                                 interaction, inhibition of                                                                             MAP kinase activation
                                 T-cell signalling and impaired
                                 activation of NF-κB
                                                                                                                            TPL2/
                                                                                                                            COT
                                                                                 P                          (Ser927)
                                                           Carma1                                                      P
                                                                         BCL10                                             P (Ser932)

                                                                                                               p1 NK
                                                            MALT1

                                                                                                                 05
                                                                                                                 A
                                                                                                      p50 p105
                                                                                                      RHD RHD
                                                                                                               TPL2/
                                                                                                               COT
                                 Inhibition of                  P                                                                       (Ser536)
                                 insulin                                                                                                                     Nuclear
                                                        IRS1                                   IKKβ                                         P                localization
                                 signalling
                                                                                                                      (Ser32) P                              of RelA,
                                                                                                                                                             stimulation of

                                                                                                                                          TA elA
                                                                                                                    (Ser36) P

                                                                                                                                            D
                                                                                                                                            R
                                                                                                                                                             transactivation
                                                                                                                                                             functions
                                                                                                                                        RelA
                                                                                                                           p50           RHD
                                                                         P                                                       IκBα
                                                                  DOK1
                                                                                               P                  P                            Canonical pathway
                                       Downregulation of MAP                             14-3-3β           FOXO3a                              activation
                                       kinase signalling                               TPP

                                                               Inhibition of mRNA binding,                 Inactivation of FOXO3a,             Innate immune response,
                                                               possible stabilization of cytokine,         inhibition of apoptosis             inflammation, cell survival
                                                               chemokine and growth factor                                                     and cell adhesion
                                                               mRNAs

                                                                                                           Cell proliferation
                                                                         Cell proliferation

                                Figure 2 | The consequences of IKKβ activation. Activation of IKKβ stimulates anti-apoptotic, pro-inflammatory and
                                proliferative pathways. As well as activating the canonical NF-κB-signalling pathway through phosphorylation (P) of the
                                IκB proteins, IKKβ phosphorylates several other substrates, including NF-κB subunits. For example, phosphorylation of
                                RelA at Ser536 can result in nuclear localization and stimulation of its transactivation functions by promoting interactions
                                with co-activator proteins. In some unactivated cells, the p105 NF-κB subunit is found in a complex with the mitogen-
                                activated protein (MAP)/extracellular signal-regulated kinase (ERK) (MEK) kinase TPL2 (also known as COT). IKKβ
                                phosphorylates p105 resulting in its degradation, which releases TPL2 resulting in activation of the pro-proliferative MAP
                                kinase-signalling pathway. Another way in which IKKβ might exert its anti-apoptotic effects is through phosphorylation
                                and inhibition of the tumour suppressor FOXO3a. By contrast, phosphorylation of the adaptor protein DOK1 is thought to
                                contribute to its ability to inhibit MAP kinase signalling and cell proliferation. IKKβ phosphorylates and inhibits the 14-3-3β
                                protein when complexed with tristetraprolin (TPP), an AU-rich element (ARE) binding protein that regulates mRNA
                                stability. IKKβ phosphorylation inhibits TPP–14-3-3β ARE binding and might therefore stabilize cytokine, chemokine and
                                growth factor transcripts. IKKβ can also inhibit insulin signalling by targeting insulin-receptor substrate-1 (IRS1).
                                Phosphorylation of BCL10 as part of the T-cell receptor (TCR) Carma1–BCL10–MALT1 complex disrupts the BCL10–MALT1
                                interaction, therefore attenuating T-cell signalling. As TCR signalling activates the NF-κB pathway, BCL10 phosphorylation
                                by IKKβ serves as a negative feedback loop. ANK, ankyrin-repeat motif; IκB, inhibitor of κB; IKK, IκB kinase; NF-κB, nuclear
                                factor-κB; RHD, Rel-homology domain; TAD, transcriptional activation domain.

                                   There are also links between IKKβ activity and the                 the cellular FLICE-like inhibitory protein (c-FLIP), the
                                pro-proliferative MAP kinase pathway. In unstimulated                 coatomer-β subunit protein COPB2, JNK-interacting
                                cells, such as macrophages and fibroblasts, the MAP/                  leucine-zipper protein (JLP) and A20-binding inhibitor
                                extracellular signal-regulated kinase (ERK) (MEK)                     of NF-κB (ABIN)36–39. So, IKKβ-induced p105 proteolysis
                                kinase TPL2 (also known as COT) is found in a complex                 might conceivably activate various NF-κB-independent
                                with a subset of the p105 NF-κB subunit. IKKβ activity                pathways. IKKβ can also phosphorylate DOK1, an adap-
                                can induce proteolysis of p105, which in turn releases                tor protein that downregulates MAP kinase activation,
bHLH transcription factor       the bound TPL2, resulting in MAP kinase-pathway acti-                 providing another possible link for the regulation of this
A transcription factor          vation34,35. Whether this pathway provides the basis for              pathway 40.
containing a DNA-binding and    activation of MAP kinase signalling by Lys63 modified                    A clear implication of these results is that the pro-
dimerization domain
characterized by a region of
                                NEMO21 is not currently clear. p105 has been reported to              inflammatory, anti-apoptotic, pro-proliferative and
basic amino acids followed by   associate with several other non-NF-κB proteins, such as              tumour-promoting characteristics that have been
a helix–loop–helix motif.       the basic helix–loop–helix (bHLH) transcription factor LYL1,          ascribed to IKKβ2,4,8 might in part result from these

NATURE REVIEWS | MOLECULAR CELL BIOLOGY                                                                                                 VOLUME 8 | JANUARY 2007 | 53
                                                            © 2007 Nature Publishing Group
REVIEWS

                                                                                                                                                    Degradation of
                                                                                                                                      P             RelA
                                           Regulates chromatin                                                             (Ser536)

                                                                                                                                  TA elA
                                           remodelling, activates

                                                                                                                                    D
                                                                                                                                    R
                                           transcription                                                                                            Resolution of
                                                                                                                                                    inflammation
                                                                            P (H3 Ser10)                               p50 RelA
                                                                                                                       RHD RHD

                                                                                                                                          Re
                                                                                                                                                                     Proteolytic

                                                                                                                                            lB
                                                                                    CBP                                                                 (Ser866)
                                   Induction of                                                                                                     P                processing
                                                                    P                      IKKα

                                                                                                                                           p1 NK
                                   INFα                                                                                                                 P            of p100 to p52

                                                                                                                                             00
                                                                                                                                             A
                                   expression              IRF7                                                                       p100              (Ser870)
                                                                                                                                      RHD
                                                                                                                                             RelB
                                                                                                    IKKα
                                                                                                                                                           Non-canonical pathway
                                                                                                                                                           activation

                                                                        P                                                                    P
                                                                                                                                                         Adaptive immune response,
                                                               Cyclin D1                                                                                 cell proliferation
                                                                                                                                      SMRT

                                    Degradation of cyclin D1                                    P                     P               Derepression of NF-κB-
                                                                              P           ERα                  β-catenin              dependent transactivation
                                                                              SRC3

                                                                                  Induction of cyclin D1 expression

                                                                                          Cell proliferation

                                  Figure 3 | The consequences of IKKα activation. In addition to activation of the non-canonical signalling pathway
                                  through phosphorylation of the p100 NF-κB subunit, IKKα is now thought to phosphorylate a number of other substrates.
                                  For example, phosphorylation of RelA at Ser536 in macrophages can stimulate its degradation and thus promote
                                  resolution of inflammation. IKKα also phosphorylates proteins that are not components of the NF-κB and IKK pathway.
                                  Through associating with cyclic-AMP responsive element binding (CREB)-binding protein (CBP), IKKα functions as a
                                  histone H3 serine 10 (H3 Ser10) kinase and thereby regulates chromatin remodelling. IKKα also regulates cyclin D1
                                  expression independently of NF-κB by phosphorylating oestrogen receptor-α (ERα), its co-activator SRC3 as well as
                                  β-catenin and cyclin D1 itself. IKKα also affects transcription through phosphorylation of the SMRT co-repressor, which
Cyclin D1                         leads to derepression of NF-κB transactivation, and interferon regulatory factor-7 (IRF7), which can induce the expression
A cyclin that forms
                                  of interferon-α (IFNα).Not shown are the kinase-independent effects of IKKα on keratinocyte differentiation and tooth
heterodimers with the cyclin-
dependent kinases CDK4 or
                                  development. ANK, ankyrin-repeat motif; IKK, inhibitor of NF-κB kinase; NF-κB, nuclear factor-κB; RHD, Rel homology
CDK6, thereby regulating          domain; TAD, transcriptional activation domain.
transition through the
G1 phase. Cyclin D1 also has
CDK-independent functions
and can function as a regulator   NF-κB-independent effects. Furthermore, it should                            factor (TCF) family of transcription factors, which bind
of transcription.                 be appreciated that although the development of IKKβ                         directly to the cyclin D1 promoter45–47. Interestingly,
                                  inhibitors has numerous potential therapeutic application                    β-catenin has also been described as an inhibitor of
β-catenin
A dual-function protein that      for both inflammatory diseases and cancer7–9, their                          NF-κB transcriptional activity48,49. Furthermore, it has
has a role in the cytoplasm       effects will not be totally mediated through the inhibition                  been reported that IKKα can directly phosphorylate the
regulating cell adhesion and a    of NF-κB function.                                                           cyclin D1 protein at Thr286 which, in direct contrast to
role in the nucleus as a                                                                                       its more established effects on the cyclin D1 promoter,
transcriptional co-activator
that mediates the Wnt-signal-
                                  The consequences of IKKα activation. IKKα can have a                         seems to induce cyclin D1 degradation50.
transduction pathway.             pro-proliferative function by inducing p52 activity and                          IKKα has also been found to regulate oestrogen-
Frequently mutated in cancer,     by consequently inducing the cyclin D1 promoter41,42.                        induced cell-cycle progression by indirectly inducing
β-catenin can function as an      Indeed, IKKα also seems to be able to regulate cyclin D1                     the expression, acetylation and target-gene expression
oncogene.
                                  expression through numerous NF-κB-independent                                of the E2F1 transcription factor, indicating a potentially
E2F1                              mechanisms (FIG. 3). These include the phosphorylation                       significant role for IKKα in breast cancer51. Other
A member of the E2F family of     and activation of the oestrogen receptor-α (ERα) trans-                      NF-κB-independent transcriptional effects of IKKα
transcription factors, which,     cription factor, together with its co-activator protein                      include the phosphorylation of the SMRT co-repressor
together with the                 SRC3, in breast cancer cells43,44. Moreover, IKKα can                        while it is bound to NF-κB complexes on the promoter52.
retinoblastoma tumour
suppressor, regulate genes that
                                  stimulate the cyclin D1 promoter through the phospho-                        This does not seem to disrupt the SMRT–NF-κB inter-
control cell-cycle progression    rylation and stabilization of the β-catenin oncoprotein, a                   action, but instead results in release of chromatin bound
and DNA synthesis.                transcriptional co-activator for members of the T-cell                       histone deacetylase-3 (HDAC3), leading to derepression

54 | JANUARY 2007 | VOLUME 8                                                                                                                 www.nature.com/reviews/molcellbio
                                                               © 2007 Nature Publishing Group
REVIEWS

                                 of NF-κB-dependent transcription52. IKKα can also             although a more dynamic role as a regulator of protein
                                 phosphorylate and activate interferon regulatory              function is also possible. For example, a nuclear role for
                                 factor-7 (IRF7), which is required for interferon-α           IκBα as a promoter-bound transcriptional repressor of
                                 (IFNα) production53.                                          the Notch-target gene hes1, which encodes a transcrip-
                                     In addition, IKKα regulates chromatin structure and       tional repressor and regulator of cell differentiation, has
                                 facilitates gene expression by functioning as a histone       been described68. One implication of these findings is
                                 H3 serine 10 (H3 Ser10) kinase54,55. In part, this function   that, similar to the phenotypes of IKK-knockout mice,
                                 is facilitated by IKKα interacting with the transcrip-        the phenotypes of model systems that rely solely on the
                                 tional co-activator cyclic-AMP responsive element             expression of a non-degradable IκBα ‘super-repressor’
                                 binding (CREB)-binding protein55 (CBP). Under some            protein might not result exclusively from inhibition of
                                 circumstances, the predominant H3 Ser10 kinases are           NF-κB function.
                                 mitogen- and stress-activated protein kinase-1 (MSK1)
                                 and MSK2, which are induced by p38 MAP kinase                 Modulation of NF-κ      κB subunits
                                 activation56,57. H3 Ser10 phosphorylation is crucial for      NF-κB subunits are subject to many modifications and
                                 the activation of a subset of NF-κB-dependent genes58.        regulatory interactions that define their transcriptional
                                 As p38 MAP kinase is induced by many of the same              activity and target-gene specificity. These interactions
                                 inflammatory stimuli and cell stresses that also induce       form a crucial interface with other signalling pathways
                                 NF-κB, this provides a mechanism through which this           in the cell; modifications of NF-κB subunits by IKK-
                                 signalling pathway can determine the consequences of          independent routes can determine their ability to interact
                                 NF-κB activation.                                             with co-activators or co-repressors, whereas cooperative
                                     IKKα also has kinase-independent roles in the             DNA binding with heterologous transcription factors
                                 regulation of keratinocyte differentiation59 and tooth        can determine their ability to selectively target promo-
                                 development60. Taken together, it can be concluded that       ters and enhancers (BOX 1). Indeed, these regulatory
                                 IKKα regulation of these diverse pathways will result         mechanisms provide explanations for cell-type- and
                                 in profound effects on the expression of many genes,          stimulus-specific effects of NF-κB, such as the ability to
                                 independently of any direct regulation of NF-κB.              either inhibit or facilitate apoptosis6,12,25. These pathways
                                                                                               also provide important routes through which oncogenes
                                 IKK-independent functions of NEMO. There is also              and tumour suppressors can modulate NF-κB activity 6.
                                 evidence that NEMO has IKK-independent functions.
                                 For example, the ability of NEMO to translocate to the        Post-translational modifications. The true extent of
                                 nucleus and to associate with ATM following DNA               NF-κB-subunit modifications is currently unknown,
                                 damage23 clearly implies that NEMO can function inde-         with only the RelA protein having been studied in any
                                 pendently of IKKα or IKKβ. Indeed, nuclear-localized          detail12. However, from these studies, it is clear that there
                                 NEMO has been shown to compete with both RelA and             are many subunit modifications with diverse functional
                                 IKKα for binding to the N-terminal domain of CBP61.           consequences12. Both IKKα and IKKβ are RelA kinases.
                                 This interaction has the potential to regulate many other     However, many other signalling pathways also use
                                 transcription factors that use CBP. For example, NEMO         modification of RelA and the other NF-κB subunits to
                                 stimulates the interaction of CBP with hypoxia-inducible      control NF-κB-pathway function. For example, RelA is
                                 factor-2α (HIF2α), independently of the IKK subunits,         phosphorylated at: Ser276 by the catalytic subunit of
                                 and also directly interacts with HIF2α, thereby enhancing     protein kinase A (PKAc), MSK1 and MSK2; at Ser311
                                 its transcriptional activity62.                               by the atypical PKCζ; at Ser468 by IKKβ, IKKε and
                                                                                               glycogen-synthase kinase-3β (GSK3β); at Ser529 by CK2;
                                  κB proteins
                                 Iκ                                                            and at Ser536 by IKKβ, IKKα, IKKε, NF-κB activating
                                 Whether the IκB proteins themselves have NF-κB-               kinase (NAK, also known as TANK-binding kinase-1
                                 independent functions is less clear, although there have      (TBK1)) and RSK1 (also known as p90 ribosomal
                                 been interesting, albeit largely unconfirmed, reports         protein S6 kinase (p90S6K))12. These can all generally be
                                 indicating that this might be the case. For example, pro-     described as stimulatory modifications that enhance the
                                 teomic analysis of IκB-associated proteins indicated that,    transcriptional activity of RelA and its ability to interact
                                 in addition to interactions with NF-κB subunits, IKKs         with co-activators, such as p300 and CBP69,70. RelA is
                                 and components of the ubiquitin-proteasome machin-            also acetylated at several sites by p300 and CBP12,69–71.
                                 ery, IκBα also associates with the replication proteins       By contrast, phosphorylation at Thr505 by the CHK1
                                 minichromosome maintenance-5 (MCM5) and MCM7                  checkpoint kinase, which can be activated by ATM- and
                                 (REF. 36). IκBβ and IκBε were found to associate with         Rad3-related (ATR) kinase in response to ARF tumour
                                 several uncharacterized proteins in this study. IκBα was      suppressor induction or DNA damage following cisplatin
                                 also reported to interact with cyclin-dependent kinase-4      treatment, inhibits RelA transactivation and results in
CBP and p300                     (CDK4)63 together with the p53 tumour suppressor64–66,        its increased association with HDAC1 (REFS 72–74).
CBP and p300 are highly          and can inhibit HIV-1 Rev protein function independ-          This results in the repression of BCL-xL expression, an
homologous transcriptional       ently of NF-κB67. The significance of these reports is not    anti-apoptotic gene that is typically induced by NF-κB
co-activator proteins with
histone acetyltransferase
                                 currently clear. It is possible that IκB degradation might    in response to inflammatory stimuli, and sensitization to
domains. Both interact with      release sequestered proteins in a manner that is ana-         apoptosis. RelA-dependent repression of anti-apoptotic
various transcription factors.   logous to p105-mediated activation of TPL2 (see above),       gene expression is also sometimes observed following

NATURE REVIEWS | MOLECULAR CELL BIOLOGY                                                                                   VOLUME 8 | JANUARY 2007 | 55
                                                         © 2007 Nature Publishing Group
REVIEWS

                                   NF-κB activation by UV-light exposure and some chemo-        for the exquisite virus-specific expression of IFNβ: other
                                   therapeutic drugs, although the mechanisms involved          inducers of NF-κB fail to activate the full complement
                                   are probably diverse and do not all involve Thr505           of IFNβ enhancer-binding proteins and so fail to induce
                                   phosphorylation74–77. The effects of the many subunit        its formation. The principles of enhanceosome forma-
                                   modifications are also probably promoter specific78 and      tion, including the integration of NF-κB activity with
                                   allow heterologous signalling pathways to specifically       other signalling pathways and the specificity this brings,
                                   modulate the NF-κB pathway.                                  are found at many other promoters, although the
                                       RelA Ser536 phosphorylation by IKKβ provides a           components might differ88.
                                   mechanism to integrate NF-κB with the phosphoinositide           NF-κB can interact with many types of transcription fac-
                                   3-kinase (PI3K) and PKB/AKT-signalling pathway, which        tor, including bZIP transcription factors such as C/EBPβ,
                                   is induced by growth factors, cytokines and oncogenes        members of the Jun, ATF, CREB and Fos family and zinc-
                                   such as Ras79. Similar to IKKβ activation, this pathway      finger-containing proteins such as specificity protein-1
                                   promotes cell survival, and several reports indicate that    (Sp1) and early growth response-1 (EGR1)86,87,91,92 (BOX 1).
                                   IKKβ-dependent RelA Ser536 phosphorylation requires          Also, recruitment of NF-κB to promoters might not
                                   PI3K/AKT activity80–82. Interestingly, in vitro, AKT is      always require a κB element. Recruitment of the RelA–
                                   required for the IKKβ-mediated phosphorylation of            HDAC4 complex to the Kruppel-like factor-2 (KLF2)
                                   RelA, but not of IκBα81. Underlining its potential role      promoter by the MADS box factor myocyte-enhancer
                                   as an integration point, IKKβ-dependent phosphoryla-         factor-2 (MEF2), independently of RelA DNA bind-
                                   tion of RelA at Ser536 that follows TCR stimulation is       ing, has recently been proposed93. Furthermore, the
                                   PI3K/AKT-independent but requires TPL2, PKCθ and             p52 NF-κB subunit can be recruited by p53 to multiple
                                   NIK kinases83. The complexity of these signalling path-      p53-regulated promoters, independently of the DNA-
                                   ways is further illustrated by the diverse consequences of   binding activity of p52 (REF. 94). There might well be many
                                   Ser536 phosphorylation which, in addition to enhancing       other mechanisms of crosstalk between transcription
                                   RelA transactivation, can also induce its translocation      factors. For example, the non-canonical NF-κB pathway
                                   to the nucleus, independently of IκB degradation, and        can be induced by the signal transducer and activator of
                                   enhance its proteolytic degradation84,85.                    transcription-3 (STAT3), which binds DNA in a complex
                                                                                                with p52 (REF. 95). By contrast, active STAT1, induced by
                                   Crosstalk with heterologous transcription factors.           IFNα expression, has been reported to interact with RelA
                                   Cooperative interactions with DNA-bound transcrip-           and inhibit its DNA binding and nuclear localization96.
                                   tion factors provide another important mechanism to              Recent data indicate that cooperative DNA binding
                                   integrate NF-κB function with other signalling pathways.     might not always be the mechanism of the synergistic
bZIP-motif-containing
                                   The position and orientation of many (if not most) NF-κB     transcriptional effects that are observed between NF-κB
transcription factor               binding sites in promoters and enhancers is not random,      and heterologous transcription factors87,97. Rather, these
A transcription factor that        and both the context and the sequence of the κB element      might derive from cooperative recruitment of co-activa-
contains a DNA-binding and         can determine the function of NF-κB subunits. Promoter       tor proteins, as also occurs at the IFNβ enhanceosome98,
dimerization domain
                                   structure can therefore facilitate the cooperative bind-     or from the ability of different transcription factors to
characterized by a region of
basic amino acids followed by      ing of NF-κB subunits with heterologous transcription        affect different steps in the transcriptional activation
a leucine-zipper motif.            factors, thereby enhancing the recruitment of one or both    process. For example, synergism between RelA and
                                   to weak or nucleosome-associated binding sites86,87.         the constitutive and widely expressed DNA-binding
Jun, ATF, CREB and Fos                 One of the best characterized examples of such effects   protein Sp1 might derive in part from the ability of Sp1
family
A transcription factor family,
                                   is provided by the IFNβ enhancer. IFNβ expression is         to stimulate the initiation of transcription, whereas RelA
members of which all contain       induced upon viral infection and is NF-κB depend-            enhances subsequent re-initiation steps99. Similarly, trans-
related bZIP domains and form      ent88. However, other non-viral inducers of NF-κB fail       criptional synergy between the Drosophila melanogaster
homo- and heterodimers that        to induce IFNβ expression. This is due to the specific       NF-κB homologue Dorsal and the bHLH transcription
bind related DNA sequences.
                                   architecture of the IFNβ enhancer, in which the κB           factor Twist, which is essential for the expression of Snail,
Collectively, dimers of Jun and
Fos family members are often       motif is part of a composite element that contains           a protein required for the specification of mesoderm, is
referred to as the AP1             DNA-binding sites for a heterodimer of the bZIP-motif-       thought to occur primarily through their ability to make
transcription factor.              containing transcription factors Jun and ATF2, members       separate contacts with distinct, rate-limiting compo-
                                   of the IRF family and the high-mobility-group protein        nents of the RNA polymerase II (Pol II) transcriptional
Kruppel-like factor
A protein that is homologous
                                   HMG-I89. Mutation or altering the spacing or orienta-        machinery100. Whatever the mechanism, the many inter-
to the Kruppel transcription       tion of the transcription-factor binding sites in the        actions between NF-κB and other transcription factors
factor, originally characterized   enhancer prevents cooperative DNA binding and the            provide a key mechanism to both integrate NF-κB sig-
in Drosophila melanogaster.        formation of a higher-order transcription-factor–DNA         nalling with other cellular processes and define the exact
Kruppel-like factors possess
                                   complex, known as the enhanceosome90. Furthermore,           nature of the NF-κB response to specific cell stimuli.
zinc-finger-containing DNA-
binding domains.                   the IFNβ κB element differs from those in many other
                                   promoters, as it contains a central core with the sequence   Feedback loops. Activation of NF-κB results in repro-
MADS box                           AAATT that allows binding of HMG-I to the minor              gramming of the cell’s gene-expression patterns to
The MADS box is a conserved        groove89. HMG-I also binds other sites in the enhancer       respond to a specific stimulus or changed environment.
DNA-binding motif originally
identified in the MCM1, AG,
                                   and facilitates cooperative DNA binding of other tran-       Many NF-κB target genes generate feedback to influ-
DEFA and SRF proteins, from        scription factors as well as enhanceosome formation89.       ence the function of NF-κB and also to profoundly affect
which it derives its name.         Formation of the enhanceosome structure is required          other signalling pathways.

56 | JANUARY 2007 | VOLUME 8                                                                                           www.nature.com/reviews/molcellbio
                                                           © 2007 Nature Publishing Group
REVIEWS

                                                          TNFα                                      B cells107. These, in turn, through cooperative effects
                                                                              NEMO/IKKβ
                                                                                                    similar to those described above, are then required for
                                                                         MnSOD
                                                                                                    the induction of a second wave of NF-κB target genes.
                                                                                                    The duration of the NF-κB response is prolonged
                                                          ROS
                                                                          FHC                       following LPS stimulation relative to that caused by other
                                                                                          NF-κB     stimuli, such as TNFα. This is a result of IRF3-dependent
                                                                        GADD45β         UV light    TNFα synthesis leading to a positive feedback loop that
                                                         MKK7                                       restimulates IKK activity108,109. Interestingly, RelA has
                                                                          XIAP                      been shown to function as a transcriptional co-activator
                                                                                                    for IRF3 following treatment with LPS, but not other
                                                          JNK                    PKCδ
                                                                                                    stimuli110.
                                                                                                        Together, mechanisms such as these can control
                                    Proliferation                                                   the timing and functional consequences of NF-κB
                                    (epidermal cells)                                               activation, allowing its cell-type and stimulus-specific
                                                                                                    functions to be revealed.

                                                                                                    Integration with other signalling pathways
                                                                                                    The integration of the NF-κB pathway with some important
                                                        Apoptosis                                   cell-signalling pathways has been analysed in detail. The
                                    Figure 4 | Crosstalk between the NF-κB and JNK-                 complex relationships that exist provide examples of the
                                    signalling pathways. Exposure of cells to tumour                intricate crosstalk that can occur between fundamental
                                    necrosis factor-α (TNFα) can result in the generation of
                                                                                                    cell-signalling pathways. Importantly, these studies also
                                    reactive oxygen species (ROS), leading to activation of
                                    mitogen-activated-protein kinase kinase-7 (MKK7) and            show that feedback can occur in both directions and that
                                    Jun N-terminal kinase (JNK). If left unopposed, JNK             the functional consequences of this can vary, depending
                                    activation will lead to apoptosis, except in some cell types,   on the context.
                                    such as epidermal cells, in which it can stimulate cell
                                    proliferation. However, TNFα signalling also activates IKKβ     Crosstalk with the JNK-signalling pathway. Many cell
                                    and the canonical NF-κB pathway. NF-κB induces the              stresses and stimuli that induce the NF-κB pathway,
                                    expression of antioxidizing enzymes such as manga-              such as TNFα, can also activate JNK signalling111,112,
                                    nese-superoxide dismutase (MnSOD) and ferritin heavy            which has many consequences, including pro-apoptotic
                                    chain (FHC), which counteract ROS generation. Also,             effects 111,112. However, NF-κB activation by TNFα
                                    other proteins activated by NF-κB, such as GADD45β and
                                                                                                    prevents apoptosis through the induction of several
                                    XIAP, can oppose JNK activation by inhibiting other
                                    components of the JNK-signalling pathway. By contrast,          anti-apoptotic genes25 and by the suppression of the
                                    following ultraviolet (UV)-light exposure, NF-κB induces        JNK pathway111,112 (FIG. 4). Activation of JNK by TNFα
                                    the expression of protein kinase Cδ (PKCδ), which               requires the generation of reactive oxygen species (ROS),
                                    activates JNK. The point at which XIAP affects the JNK          a process that can be counteracted by NF-κB through
                                    pathway is not currently known. IKK, inhibitor of NF-κB         the induction of genes that encode antioxidizing
                                    kinase; NEMO, NF-κB essential modifier; NF-κB, nuclear          enzymes such as manganese-superoxide dismutase
                                    factor-κB.                                                      (MnSOD) and ferritin heavy chain (FHC)111–113. In addi-
                                                                                                    tion, the products of other activated NF-κB target genes,
                                                                                                    such as GADD45β (also known as MYD118), which is an
                                       With the exception of RelA, all the NF-κB subunits           inhibitor of the JNK upstream kinase mitogen-activated-
                                    and IκBα contain κB sites in their promoters1,5. The            protein kinase kinase-7 (MKK7, also known as JNKK2)
                                    positive regulation of NF-κB subunits can result in the         and the anti-apoptotic protein XIAP, also suppress
IRF3
                                    composition of the NF-κB complex changing over time,            JNK activation (the effect of XIAP occurs through an
A transcription factor that
regulates genes that contain        whereas induction of IκBα provides a crucial negative           undefined mechanism)111,112. In many cell types, sup-
the interferon-sensitive            feedback loop that helps terminate the NF-κB response.          pression of JNK-induced apoptosis can contribute to the
response element (ISRE)             Other NF-κB target genes, such as the CYLD tumour               tumour-promoting activities of NF-κB111–113. However,
sequence. IRF3 activity can be      suppressor and A20, both of which encode deubiquityl-           in epidermal cells, JNK stimulates cell proliferation and
induced through binding of LPS
to Toll-like receptor-4, which
                                    ating enzymes, can also inhibit and limit the NF-κB             therefore, in these cells, the inhibition of JNK by NF-κB
also activates NF-κB.               response by targeting signalling molecules, such as             has a tumour-suppressing function6,114. By contrast, after
                                    TRAF proteins, which are required for IKK-complex               UV stimulation, RelA directly induces the expression of
E3 ubiquitin ligase                 activation101–106. Interestingly, CYLD was also recently        PKCδ, which in turn activates JNK77.
A protein that directly interacts
                                    shown to deubiquitylate the p50 and p52 co-activator
with target proteins and
catalyses their ubiquitylation.     BCL-3, leading to inhibition of both cyclin D1 expression       Crosstalk with p53. The p53 tumour suppressor and
Lys48-linked ubiquitylation         and proliferation in keratinocytes106.                          transcription factor is one of the first lines of defence
can lead to proteasomal                Activation of other genes also determines NF-κB-target-      against the effects of genotoxic damage or oncogene
degradation whereas Lys63-          gene specificity. For example, members of the activator         activation, and it typically induces apoptosis or cell-
linked ubiquitylation can
facilitate protein interactions
                                    protein-1 (AP1) and ATF families of transcription factors,      cycle arrest in response to these stimuli115. The amount
and activation of signalling        such as JunB, JunD, B-ATF and ATF4, are rapidly induced         of p53 protein in cells is regulated by HDM2 (known as
pathways.                           by NF-κB following LPS stimulation of precursor                 MDM2 in mice) which functions as an E3 ubiquitin ligase

NATURE REVIEWS | MOLECULAR CELL BIOLOGY                                                                                       VOLUME 8 | JANUARY 2007 | 57
                                                                 © 2007 Nature Publishing Group
REVIEWS

                                                                   ATR         CHK1

                                                                                           p300/CBP
                                                                                       Competition for
                                                                                       binding
                                                 Upregulation of
                                                 HDM2 promoter                              RSK1

                                                                                                         Ser536 P       P Thr505
                               ARF       HDM2                            p53                                    NF-κB
                                                                                      Induction of p53
                                                                                      expression

                                        BCL-3

                                                                                                                   lA
                                                                                                                 Re
                                                                                                           p50 RelA
                                                  p53

                                                                                                Genotoxic                          Inflammatory stimuli,
                                                                                                stress, ARF induction              genotoxic stress

                                            Induction of                         Repression of             Induction of            Induction of
                                            pro-apoptotic genes:                 anti-apoptotic genes:     pro-apoptotic           anti-apoptotic genes:
                                            Bax, PUMA, DR5                       BCL-xL, XIAP, A20         genes:                  BCL-xL, XIAP, IAP1, IAP2,
                                                                                                           Fas, FasL, DR5          A20

                                                                                          Apoptosis
                         Figure 5 | Integration of the p53 pathway with the function of the RelA (p65) NF-κB subunit. Crosstalk between
                         the tumour suppressor p53 and NF-κB pathways regulates apoptosis. In response to inflammatory stimuli, NF-κB
                         induces anti-apoptotic genes that antagonize the pro-apoptotic function of p53. Moreover, NF-κB, or the IκB family
                         member BCL-3, can induce the expression of HDM2 (known as MDM2 in mice) and reduce p53 protein levels.
                         Additional antagonism comes from competition between p53 and RelA for binding to shared co-activator proteins
                         such as p300 and cyclic-AMP responsive element binding (CREB)-binding protein (CBP). By contrast, there are also
                         cooperative pathways between p53 and NF-κB. Some inducers of NF-κB result in repression of anti-apoptotic genes
                         and induction of pro-apoptotic genes. Furthermore, induction of the ARF tumour suppressor can activate the CHK1
                         checkpoint kinase, which phosphorylates (P) RelA at Thr505, leading to repressed expression of the anti-apoptotic
                         NF-κB gene target BCL-xL. p53 can induce RelA nuclear localization and DNA binding through activation of RSK1
                         (also known as p90 ribosomal protein S6 kinase (p90S6K)). NF-κB has also been reported to induce the expression of
                         p53. Which of these pathways dominates will probably depend on the cell type and the nature of the inducing stimulus
                         for both p53 and NF-κB. Not shown are the regulatory mechanisms linking p53 to the p52 NF-κB subunit. ATR, ataxia
                         telangiectasia mutated (ATM)- and Rad3-related; IκB, inhibitor of κB; NF-κB, nuclear factor-κB.

                         and induces p53 proteolysis115. Expression of HDM2                    In contrast to these antagonistic effects, there is also
                         and the consequent downregulation of p53 can be                    significant cooperative crosstalk between the NF-κB
                         induced by IKK and by NF-κB activation116, or by the               and p53 pathways (FIG. 5). For example, p53 can induce
                         IκB-like BCL-3 co-activator117 (FIG. 5). The RelA NF-κB            RSK1 activity, which phosphorylates RelA at Ser536,
                         subunit has also been shown to antagonize p53 trans-               resulting in the nuclear localization of NF-κB 84.
                         activation through sequestration of the p300 and CBP               Moreover, in some circumstances RelA and p53
                         co-activators118. These effects on p53, together with the          cooperatively induce apoptosis119–121. This can occur
                         other functions of NF-κB, such as induction of anti-               through the coordinated induction of pro-apoptotic
                         apoptotic genes, all contribute to the tumour-promoting            target genes such as death receptor-5 (DR5), which
                         effects of NF-κB.                                                  contains both p53 and NF-κB response elements122.

58 | JANUARY 2007 | VOLUME 8                                                                                          www.nature.com/reviews/molcellbio
                                                 © 2007 Nature Publishing Group
REVIEWS

a Induction of IκBα expression                          b Co-activator competition                       There is also crosstalk between p53 and the p52
     Ligand                                                                                          NF-κB subunit. Activation of p53 can inhibit cyclin D1
                             IκBα                                                                    expression by inducing the association of the p52 NF-κB
                                        p50      RelA                                     p300/CBP   subunit with co-repressor complexes42. Moreover, in some
                                                                                                     cell lines, p52 is directly recruited to p53-regulated pro-
                                                                                                     moters, where it regulates co-activator and co-repressor
                                        NF-κB                                                        recruitment, leading to either repression or activation
                                                                                                     of p53 target genes such as p21WAF1, DR5 and PUMA94.
     Iκ Bα                                                    κB element                             Through this pathway, p52 has the potential to regulate
    Nucleus                         Cytoplasm
                                                                                                     p53-dependent cell-cycle arrest and apoptosis.
c Sequestration of NF-κB                                d Disruption of co-activator-
                                                          complex formation                          Crosstalk with nuclear receptors. Nuclear receptors (NRs)
                                                                                                     are transcription factors that are typically regulated by
                                                                                   IRF3
                                                                                                     the binding of specific ligands, such as glucocorticoids,
                                                                                                     oestrogen, thyroid hormone and retinoic acid124–126.
                                                                                                     Many NRs possess anti-inflammatory functions that are
                                                                                                     mediated largely through the inhibition of NF-κB and
                                                                                                     AP1 transcriptional activity124. What is revealing about
                                                                                                     studies of NRs, and what is especially relevant when
       IRF3      IRF3                                                                                considering the other pathways engaged in crosstalk by
                                                                                                     NF-κB, is the diversity of mechanisms through which
          IRSE                                                     κB element
                                                                                                     NRs can inhibit NF-κB function124 (FIG. 6).
e Inhibition of RNA polymerase II                       f Recruitment of HDACs                           Most mechanisms of NF-κB repression seem to
   hyperphosphorylation                                                                              involve direct interactions between NF-κB and NRs, and
                                 P-TEFb                                                              these are referred to as transrepression124–126. However,
                                                              HDAC2                                  there are also indirect mechanisms, including induction
                                    P                                                                of IκBα expression127,128 and competition for co-activators
                                                                                                     such as p300 and CBP129.
                        Pol II                                                                           Direct interaction with NRs can result in seques-
                                                                                                     tration of NF-κB, which inhibits IRF3-dependent
                                                                                                     promoters at which RelA functions as a transcriptional
          κB element                                                  κB element                     co-activator110,130. Conversely, IRF3 can also function
                                                                                                     as a co-activator for some NF-κB-regulated genes and
Figure 6 | Crosstalk between the NF-κB and nuclear-receptor pathways. Multiple                       here, binding of RelA to the glucocorticoid receptor
mechanisms contribute to the ability of nuclear receptors (NRs) to repress the NF-κB
                                                                                                     prevents IRF3 recruitment, leading to transcriptional
pathway and thereby function in an anti-inflammatory manner. Different mechanisms
seem to selectively regulate distinct subsets of NF-κB-regulated genes. Some                         repression130. Interestingly, other NRs do not disrupt the
pathways of NR-mediated repression are indirect and involve: (a) induction of IκBα                   interaction between IRF3 and RelA, but rather repress
expression; or (b) competition for co-activator proteins such as cyclic-AMP responsive               distinct subsets of NF-κB-regulated genes, indicating
element binding (CREB)-binding protein (CBP) and p300. However, most mechanisms                      that there are other mechanisms of repression124,130.
of NF-κB repression involve direct interactions with NRs and are referred to as                      Direct recruitment of NRs by RelA to NF-κB-regulated
transrepression. (c ) Direct interaction with NRs can result in sequestration of NF-κB,              promoters can have various other effects that can lead
which inhibits interferon regulatory factor-3 (IRF3)-dependent promoters, for which                  to repression of transcription. These include the recruit-
RelA functions as a transcriptional co-activator. (d) Conversely, interactions of NRs                ment of HDACs (or a failure to clear co-repressors from
with RelA can prevent IRF3 from functioning as a co-activator at some NF-κB-                         the repressed basal state of the gene) and the inhibition
regulated promoters. RelA-dependent recruitment of NRs to promoters can lead to
                                                                                                     of Pol II phosphorylation131,132. These diverse mecha-
repression of transcription by other mechanisms, including: (e) inhibition of
RNA polymerase II (Pol II) phosphorylation (P) by P-TEFb; (f) recruitment of histone                 nisms seem to affect specific subsets of NF-κB-regulated
deacetylases (HDACs); or a failure to clear co-repressors from the repressed basal state             genes, providing opportunities for selective and context-
of the gene (not shown). IκB, inhibitor of κB; IRSE, interferon-sensitive response                   dependent regulation of NF-κB function.
element; NF-κB, nuclear factor-κB; P-TEFb, positive transcription elongation factor b.
This figure is modified with permission from REF. 124 © (2006) Elsevier.                             Conclusions
                                                                                                     These examples illustrate the complexity of the NF-κB
                                                                                                     pathway, but only scratch the surface of this complicated
                                    Alternatively, in contrast to the results described              area of biology. The different modifications of the NF-κB
                                    above, NF-κB activity can also increase p53 expres-              subunits, and how they control the selectivity of NF-κB-
                                    sion and stability, which results at least in part from          target-gene regulation, has only begun to be investigated.
                                    the direct regulation of the p53 gene by NF-κB120,121.           Furthermore, in many contexts, the contribution of
                                    In addition, the ARF tumour suppressor, which binds              NF-κB interactions with other transcriptional regulators
                                    to and inhibits HDM2, thereby activating p53, can                to NF-κB subunit function and target-gene specificity has
                                    induce ATR and CHK1 checkpoint kinase activity73,123             not been investigated in depth, and the significance of
                                    leading to RelA Thr505 phosphorylation, BCL-xL                   IKK-independent mechanisms of NF-κB activation has
                                    repression and sensitization to apoptosis73.                     not yet been clearly defined. Therefore, although much

NATURE REVIEWS | MOLECULAR CELL BIOLOGY                                                                                        VOLUME 8 | JANUARY 2007 | 59
                                                             © 2007 Nature Publishing Group
You can also read