Vous êtes sur la page 1sur 23

Oncogene (2006) 25, 6758–6780

& 2006 Nature Publishing Group All rights reserved 0950-9232/06 $30.00
www.nature.com/onc

REVIEW

NF-jB and the immune response

MS Hayden1, AP West1 and S Ghosh1,2


1
Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA and 2Department of Molecular
Biophysics and Biochemistry, Yale University School of Medicine, New Haven, CT, USA

One of the primary physiological roles of nuclear factor- Considered broadly, mammalian immune responses
kappa B (NF-jB) is in the immune system. In particular, can be divided into innate and adaptive responses. The
NF-jB family members control the transcription of immune response begins with the host recognizing the
cytokines and antimicrobial effectors as well as genes that presence of foreign pathogens, followed by responses at
regulate cellular differentiation, survival and proliferation, the cellular, tissue and organismal levels, that ultimately
thereby regulating various aspects of innate and adaptive lead to the clearance of the pathogen. As such, immune
immune responses. In addition, NF-jB also contributes to responses can be broken down into individual signal
the development and survival of the cells and tissues that transduction events through which changes in the
carry out immune responses in mammals. This review, extracellular environment elicit altered gene expression
therefore, describes the role of the NF-jB pathway in the at the cellular level. In a remarkable number of
development and functioning of the immune system. instances, NF-kB is the transcription factor that
Oncogene (2006) 25, 6758–6780. doi:10.1038/sj.onc.1209943 mediates these transcriptional changes. The gene pro-
ducts characteristic of early events in immune responses
Keywords: NF-kB; T-cell receptor; B-cell receptor; include cytokines and other soluble factors that propa-
inflammation; TLR; hematopoiesis gate and elaborate the initial recognition event. The
activation and modulation of NF-kB is also a common
target of these factors. Thus, in a surprising number of
situations NF-kB mediates the critical changes that are
characteristic of innate and adaptive immune responses.
Introduction In mammals, the NF-kB family is composed of five
related transcription factors: p50, p52, RelA (aka p65),
The discovery and characterization of the nuclear c-Rel and RelB (see Gilmore, 2006). These transcription
factor-kappa B (NF-kB) family of transcription factors factors are related through an N-terminal DNA-
resulted from studies in two major areas of research: binding/dimerization domain, called the Rel homology
immunology and cancer biology. Although the role of domain, through which they can form homodimers
NF-kB in cancer biology is becoming progressively and heterodimers, which bind to a variety of related
better established, historically much of our current target DNA sequences called kB sites to modulate
knowledge of NF-kB resulted from efforts directed at gene expression. RelA, c-Rel and RelB also contain
understanding the regulation and function of the C-terminal transcription activation domains (TADs),
immune response. In keeping with the critical role which enable them to activate target gene expression. In
played by NF-kB in different areas of immunology, contrast, p50 and p52 do not contain C-terminal
numerous excellent reviews have been published cover- transactivation domains; therefore, p50 and p52
ing the role of NF-kB in Toll-like receptor (TLR) and homodimers can repress transcription unless they are
antigen receptor (AgR) signaling, lymphoid organo- bound to a protein containing a TAD, such as Bcl-3.
genesis and hematopoiesis (Mebius, 2003; Bonizzi and Alternatively, p50 and p52 often form heterodimers
Karin, 2004; Hayden and Ghosh, 2004; Lin and Wang, with RelA, c-Rel or RelB and act as transcriptional
2004; Siebenlist et al., 2005; Akira et al., 2006). This activating dimers.
review will, therefore, attempt to provide a more In most cells, NF-kB complexes are inactive, residing
comprehensive, if less detailed, review of the diverse primarily in the cytoplasm in a complex with any of the
functions of NF-kB in immunology, with the goal of family of inhibitory IkB proteins. When the pathway is
illuminating how it is that so much in immunology activated, the IkB protein is degraded and the NF-kB
seems to revolve around this family of transcription complex enters the nucleus to modulate target gene
factors. expression. In almost all cases, the common step in this
activating process is mediated by an IkB kinase (IKK)
complex, which phosphorylates IkB and targets it for
proteasomal degradation (see Scheidereit, 2006). The
Correspondence: Professor S Ghosh, Yale University School of
Medicine, Department of Immunobiology, 300 Cedar Street, New
IKK complex consists of two catalytically active kinases
Haven, CT 06510, USA. (IKKa and IKKb) and a regulatory scaffold protein,
E-mail: sankar.ghosh@yale.edu NEMO. In what is called the canonical (or classical)
NF-jB and immunology
MS Hayden et al
6759
pathway, IKKb and NEMO are required for the its function during hematopoiesis is far more nuanced
activation of complexes such as p50/RelA, p50/c-Rel, than one might expect. In the current review, we limit
etc., whereas IKKa is relatively dispensable. Conversely, our discussion to those instances where the role for
in the non-canonical (or alternative) pathway IKKa NF-kB is illustrative of its broader functions in the
alone controls the activation of complexes that are immune system.
inhibited by the IkB protein p100. These two NF-kB Before delving into specific aspects of NF-kB function
pathways can be activated by overlapping but distinct in hematopoiesis, it is worthwhile to discuss the short-
sets of stimuli, and also target activation/repression of comings of the experimental approaches that are used.
overlapping but distinct sets of target genes. One of the For example, embryonic lethality of RelA knockout
most conserved functions of the NF-kB signaling mice prevents straightforward analysis of the hemato-
pathway is the regulation of the immune system; indeed, poietic events that are relevant to adult animals. In other
NF-kB is even the primary regulator of innate immunity instances, severe defects in lymphoid organogenesis in
in insects such as Drosophila and mosquitoes (see the absence of NF-kB make it difficult to determine
Minakhina and Steward, 2006). This review focuses on whether the observed defects are intrinsic to the
the vast role played by NF-kB in mammalian immunity. hematopoietic lineage or are due to alterations in the
relevant organ, for example, stromal tissues, within
which hematopoietic development occurs. In some
instances, such as for RelA or IKKb knockouts,
Development and formation of the immune system
embryonic lethality can be rescued by deletion of the
tumor necrosis factor-receptor (TNF-R) or tumor
The mammalian immune system consists of a function-
necrosis factor-alpha (TNFa), which permits analysis
ally linked group of anatomically disparate tissues and
of hematopoiesis in these mice, but potentially distorts
cell types. The dispersed cellular components of the
certain aspects of the hematopoietic pathway. Similar
immune system that arise from the bone marrow receive concerns apply to adoptive transfer experiments that
much of the attention in immunology and the study of
can be influenced by the cytokine milieu to which
NF-kB has likewise focused on its role in leukocytes.
transferred cells are exposed. In each case, therefore, one
However, lymphoid organs that facilitate coordination must ask whether the defect exhibited by a cell lacking
and dissemination of immune responses carried out by
some component of the NF-kB pathway is relevant to
immune cells are also key sites of NF-kB function.
the course of normal hematopoiesis or simply to the
Therefore, whereas this section is largely concerned with
experimental system being employed. Finally, there are
the role of NF-kB in hematopoiesis, the role of NF-kB
numerous instances where one NF-kB family member
in lymphoid organogenesis is also discussed briefly.
can complement the function of another member, or
alternatively, where the absence of one family member
NF-kB and hematopoiesis impedes the function or expression of other family
Most cells of the immune system are subject to rapid members. Nevertheless, despite these limitations, genetic
turnover. This process requires the regulation of the analysis has unequivocally illustrated a key role for
competing forces of cell proliferation and cell death – NF-kB in the development and survival of hemato-
processes heavily influenced by NF-kB-regulated genes. poietic cells.
Bone marrow-derived hematopoietic cells in particular
are subject to high levels of turnover and consequently
are particularly sensitive to changes in rates of apoptosis NF-kB in development of innate immune cells
or proliferation. Likewise, during immune responses DC development is largely dependent on canonical
immune cells selectively undergo rapid expansion that NF-kB complexes, although a particular subset appears
must be resolved by targeted cell death. Although the to require only the non-canonical RelB containing
role of NF-kB in development and homeostasis of NF-kB complexes. RelB is known to facilitate the de-
hematopoietic cells has focused largely on B-cell and velopment of DCs (Burkly et al., 1995; Weih et al.,
T-cell maturation, it is likely that as our understanding 1995); specifically development of CD8a, but not
increases of the pathways responsible for the develop- CD8a þ , DCs (Wu et al., 1998). Conversely, double
ment of natural killer (NK) cells, dendritic cells (DCs), knockout studies have shown that canonical p50/RelA
macrophages, etc. our appreciation for the role of NF-kB complexes are required for the development of both
in the biology of these cell types will also expand. CD8a þ and CD8a DCs, but not other myeloid and
Hematopoietic components of the immune system lymphoid lineages, most likely by mediating the
include cells of the lymphoid, myeloid and granulocytic response of DCs to TNFa (Ouaaz et al., 2002; Abe
lineages. These lineages give rise to T cells, B cells, et al., 2003). Survival of DCs in the periphery following
monocytes, macrophages, DCs (both myeloid and activation tends to be short, but can be prolonged upon
lymphoid), NK cells, basophils, eosinophils, neutrophils CD40L expression on T cells. CD40L activates both the
and mast cells (Figure 1). Many cells of the body can canonical and non-canonical NF-kB pathways and
contribute to immune responses; however, these bone hence DCs deficient in both p50 and c-Rel, or DCs
marrow-derived cells are the core constituents of both overexpressing a mutant super-repressor form of IkBa,
the innate and adaptive immune responses. Although demonstrate significantly decreased survival (Ouaaz
NF-kB generally plays a prosurvival role in these cells, et al., 2002; Kriehuber et al., 2005).
Oncogene
NF-jB and immunology
MS Hayden et al
6760

Pluripotent HSC

Myeloid Stem Cell Lymphoid Stem Cell

Platelets

Basophil Granulocyte-Monocyte
Erythrocytes Progenitor Progenitor T cell B cell NK cell

Basophil Mast Cell Eosinophil

Monocyte Neutrophil Dendritic Cells

Macrophage

Figure 1 Schematic of NF-kB in hematopoiesis. Red arrows indicate stages in which NF-kB activation is thought to contribute
negatively and green arrows indicate a positive function in the development of the indicated lineages. Curved arrows indicate examples
in which NF-kB contributes to the survival of the mature cell population, either in the resting state or during immune responses. Gray
arrows indicate developmental events for which NF-kB plays no role or for which the role of NF-kB has not being clearly
demonstrated. See the text for details.

IkBa knockout mice display robust granulocytosis pronounced defect in NK cells and lymphoid lineages
(Beg et al., 1995), and suggest an antiapoptotic role (Samson et al., 2004). Therefore, elevated levels of
for NF-kB during granulocyte development. Likewise, NF-kB activity in these cells appears to exert a proapop-
NF-kB has an antiapoptotic role in mature granulocytes. totic effect. Thus, it appears that the role of NF-kB in
For example, neutrophils, which undergo daily turnover granulopoiesis is selective and cell-type-specific.
and rapidly apoptose in vitro, exhibit accelerated Furthermore, as described for lymphocyte development
apoptosis as well as sensitization to proapoptotic stimuli (below), the requirement for individual NF-kB subunits
following NF-kB inhibition. Unlike lymphocytes, which is not uniform at different developmental stages.
are relatively long-lived in the absence of activation,
protection from apoptosis in neutrophils is more
important during the inflammatory response than in NF-kB in development of B and T cells
homeostasis. Indeed, many TLR ligands increase As in cells of the innate immune system, NF-kB is vital
neutrophil survival in vitro, likely due to NF-kB- for the development and function of adaptive immune
mediated expression of antiapoptotic genes (Francois cells (Siebenlist et al., 2005). Although lymphocytes may
et al., 2005). Although neutrophils are capable of exhibit great longevity in the periphery, their selection in
activating NF-kB in response to many pro-inflamma- the bone marrow and thymus is characterized by a high
tory stimuli (McDonald, 2004), they lack p52 and RelB rate of apoptosis. As a consequence, the antiapoptotic
(McDonald et al., 1997), the very subunits that are properties of NF-kB play a key role in lymphopoiesis.
crucial for the maintenance of long-lived lymphocytes. The centrality of its antiapoptotic function is supported
Thus in the case of neutrophils, NF-kB fulfills its in part by the demonstration that most of the require-
predicted role as a prosurvival and proinflammatory ments for NF-kB during T-cell development can be
factor. overcome by transgenic expression of the prototypical
The general granulocytosis observed in IkBa knock- antiapoptotic factor Bcl-2 (Sentman et al., 1991). The
out mice suggested that an antiapoptotic role could be necessity of NF-kB for lymphopoiesis is strikingly
broadly assigned to NF-kB in this lineage. However, illustrated in human genetic diseases wherein the gene
chimeras generated with cells from ikba/ikbe/mice encoding NEMO is inactivated by mutation (see
(i.e., lacking IkBa and IkBe) instead display a modest Courtois and Gilmore, 2006). Because the NEMO gene
defect in both myelopoiesis and granulopoiesis of is located on the X-chromosome, it is usually subject to
transferred cells (Goudeau et al., 2003), and a random inactivation in individual cells in females.
Oncogene
NF-jB and immunology
MS Hayden et al
6761
However, in female patients who are heterozygous for a
Periphery
mutant version of NEMO all peripheral lymphocytes
Common Bone Marrow Thymus
possess an intact NEMO gene, rather than the 50%
predicted by random inactivation, suggesting that in the Lymphoid
Precursor
absence of NEMO-dependent NF-kB signaling, B and T
cells fail to develop. Pro-B cell DN
The effects of NEMO inactivation in both mice and Thymocyte
humans solidify the role of NF-kB in lymphopoiesis,
even though the details by which NF-kB functions in
this process remain obscure. NF-kB plays diverse roles Pre-B cell
in lymphocyte development that can be grouped
according to when in development it functions – that
is, before, during or after pre-AgR signaling. Although Immature DP
no single NF-kB subunit knockout mouse has as severe B cell Thymocyte
of a phenotype as NEMO deficiencies with regard to the
generation of mature lymphocytes, double knockouts
demonstrate that the antiapoptotic function of NF-kB is
important in the maturation and survival of lympho-
cytes. For example, loss of both of the canonical NF-kB Transitional SP
family members p50 and RelA, or both RelA and c-Rel, B cell Thymocyte
halts development early in lymphopoiesis, before Spleen
expression of the pre-AgRs (Horwitz et al., 1997;
Grossmann et al., 1999), suggesting that NF-kB is Naive B cell Naive T cell
involved in the expression of antiapoptotic factors
required for early lymphoid cell survival in response to Figure 2 NF-kB function during lymphopoiesis. NF-kB plays a
proapoptotic stimuli (Figure 2). In fact, early CD34 þ prosurvival role in common lymphoid precursor cells that give rise
bone marrow cells can activate NF-kB in response to to B- and T-cell lineages. B-cell development occurs in the bone
TNFa, and in these cells NF-kB acts as a prosurvival marrow, where NF-kB protects pre-B cells from proapoptotic
stimuli including TNFa. Signaling to NF-kB through the pre-B-cell
factor (Pyatt et al., 1999). receptor mediates survival of pre-B-cells that then undergo light
Evidence suggests that expression of the pre-AgR chain recombination to produce a functional B-cell receptor.
leads to survival signals that depend, at least in part, on Expression of BCR leads to NF-kB-dependent differentiation into
NF-kB. For example, pre-T-cell receptor (pre-TCR) immature B cells. High levels of BCR signaling, that is, through
recognition of self-antigen, results in negative selection through the
expression in double negative (DN; CD8CD4) thy- loss of NF-kB activity. Transitional B cells exit the bone marrow
mocytes coincides with high levels of NF-kB activity, and migrate to the spleen where they mature and differentiate, a
and NF-kB activity at this stage is necessary for DN process that also requires NF-kB. T-cell development occurs
survival and maturation (Figure 2). Therefore, enforced following migration of precursor cells into the thymus. Stimulation
IKKb activation eliminates the requirement for TCR of NF-kB through pre-TCRa provides a prosurvival signal
allowing recombination of the TCRa chain and maturation to
recombination, whereas inhibition of NF-kB by expres- the DP stage. Optimal signaling through the TCRa/b complex
sion of an IkBa super-repressor decreases DN thymo- induces NF-kB-dependent survival pathways, whereas a failure
cyte maturation and survival (Voll et al., 2000). to signal or high-level signaling results in death by neglect or
Signaling through the pre-B-cell receptor (pre-BCR) negative selection, respectively. NF-kB activity is required for the
maintenance of long-lived B and T cells.
also likely induces antiapoptotic signals through NF-kB.
Consequently, the reduced pre-B-cell population seen
upon expression of the IkBa super-repressor in bone
marrow cells can be rescued by overexpression of the self-peptide:MHC with an affinity that falls within a
antiapoptotic NF-kB target gene Bcl-XL (Feng et al., defined range are positively selected to become single-
2004; Jimi et al., 2005). However, whereas evidence positive T cells. Somewhat counterintuitively, it appears
points toward NF-kB-mediated production of antia- that NF-kB functions in both positive and negative
poptotic factors it remains unclear how NF-kB is selection of thymocytes. During negative selection,
activated downstream of the pre-AgR. NF-kB facilitates the induction of apoptosis following
Selection of DP (double positive; CD4 þ CD8 þ ) high-affinity TCR ligation (Hettmann et al., 1999; Mora
thymocytes depends on the ability of their TCR to et al., 2001b), perhaps by facilitating expression of
recognize peptide:MHC (major histocompatibility com- proapoptotic genes and the consequent sensitization to
plex) complexes. Thymocytes that express a TCR that is proapoptotic signals (French et al., 1996; Kishimoto
unable to bind MHC die in a process termed ‘death by et al., 1998). The role of NF-kB in positive selection of
neglect’, whereas those that bind peptide:MHC are thymocytes is more in keeping with the better-estab-
either positively or negatively selected depending on the lished role of NF-kB as an inducer of antiapoptotic
strength of this interaction. Thymocytes that bind self- genes. Unlike in thymocytes, however, NF-kB functions
peptide:MHC with very high affinity are likely to be as a prosurvival factor during negative selection of
self-reactive, and hence are deleted through negative B cells. Immature B cells display constitutive NF-kB
selection. Thus, only DP thymocytes that recognize activity that is down-regulated following BCR ligation
Oncogene
NF-jB and immunology
MS Hayden et al
6762
(Wu et al., 1996). Decreased NF-kB activity might then of transitional B-cell maturation, which mirrors that
sensitize these cells to proapoptotic signals. Interest- seen in Bcl-XL knockout mice (Motoyama et al., 1995;
ingly, some signaling components required for NF-kB Gross et al., 2001; Schiemann et al., 2001). Thus, only
activation in mature B and T cells can be genetically those knockouts that target both the canonical and non-
disrupted without affecting their development, suggest- canonical NF-kB pathways have an effect that approx-
ing that pathways leading to activation of NF-kB in imates the phenotype seen in BAFF or Bcl-XL
developing B or T cells differ significantly from the deficiency.
pathways engaged following AgR ligation in mature
lymphocytes.
Following positive and negative selection, DP thy-
NF-jB and lymphoid organogenesis
mocytes must make a lineage commitment and become
single positive (SP) thymocytes (CD4 þ CD8 or
In addition to its role in the development of cells that
CD4CD8 þ ), which shortly thereafter emigrate from
directly mediate immune responses, NF-kB also plays
the thymus. Analyses of kB-site luciferase transgenic
an important role in the development and function of
reporter mice have shown that CD8 SP cells have
primary and secondary lymphoid tissues. Primary
significantly higher levels of NF-kB activity than CD4
(central) lymphoid organs include the bone marrow
SP thymocytes (Voll et al., 2000). Conversely, the
antiapoptotic factor Bcl-2 is more highly expressed in and thymus whereas secondary (peripheral) lymphoid
organs include lymph nodes (LNs), Peyer’s patches,
CD4 than CD8 cells, suggesting that CD8 SP thymo-
mucosal-associated lymphoid tissue (MALT) and the
cytes are more dependent on NF-kB for survival.
However, during the course of the development from spleen. Among the primary lymphoid organs, the bone
marrow remains active throughout life, whereas thymic
the DP stage to emigration from the thymus, both DP
activity dwindles with the onset of adulthood. The
and SP lineages require NF-kB. Targeted deletion of
secondary lymphoid tissues are associated with the
floxed-NEMO using cd4-promoter-driven Cre recombi-
maintenance and activation of mature lymphocytes, and
nase expression, or overexpression of kinase dead
provide an environment within which the interaction of
IKKb, results in loss of mature peripheral T cells
lymphocytes and other leukocytes can be carefully
(Schmidt-Supprian et al., 2004). These data strongly
suggest that NF-kB activation is required for late stages orchestrated. Although there is clearly a role for NF-kB
in the development and regulation of bone, this role has
of T-cell development; however, ikkb/,tnfr1/ double
not yet been clearly correlated with effects on hemato-
knockouts, ikkb/ chimeras or cd4-Cre IKKb condi-
tional knockouts are not defective in the production of poiesis (Jimi and Ghosh, 2005). Careful anatomical and
histological examination of NF-kB-deficient mice has
naı̈ve T cells (Senftleben et al., 2001b; Schmidt-Supprian
resulted in NF-kB being assigned an increasingly
et al., 2004), suggesting a requirement for NEMO but
prominent role in lymphoid organogenesis.
not IKKb.
Immature B cells exit the bone marrow, becoming
transitional B cells, and complete development into Secondary lymphoid organs
either follicular or marginal zone B cells. NF-kB- The secondary lymphoid organs have highly charac-
regulated expression of prosurvival factors is important teristic structural features that are crucial to the
to these final steps of B-cell development (Grossmann development and activation of lymphocytes. Analysis
et al., 2000). Interestingly, the activation of NF-kB in of the role of NF-kB in lymphoid organogenesis in
late B-cell maturation is the result of signaling by both knockouts has been complicated by the necessity of
canonical and non-canonical NF-kB pathways. Thus interfering with the TNF response to rescue the lethality
deficiency in NEMO, IKKa or IKKb decreases the associated with NF-kB deficiency. The initial events of
numbers of mature B cells (Kaisho et al., 2001; lymphoid organogenesis involve the association of
Senftleben et al., 2001b; Pasparakis et al., 2002). lymphotoxin (LT)a1b2-expressing hematopoietic cells
Likewise, either p50/p52 or RelA/c-Rel double knock- and vascular cell adhesion molecule-1 (VCAM1)-
out progenitor cells are defective in their ability to expressing stromal cells (for a review see Mebius,
mature beyond the transitional B-cell stage (Franzoso 2003). This interaction initiates a positive feedback-
et al., 1997; Grossmann et al., 1999). A requirement for signaling loop in which NF-kB plays a prominent role
both the canonical and non-canonical NF-kB pathways (Figure 3). Cytokines implicated in this signaling loop –
may explain why deletion of p50 and p52 produces a LTa1b2, RANKL (receptor activator of NF-kB ligand)
more complete block in B-cell development than loss of and TNFa – are known to activate NF-kB. Also,
RelA and c-Rel. Although both canonical and non- mediators of lymphoid organogenesis and homeostasis,
canonical NF-kB pathways are functional during B-cell such as the adhesion molecules ICAM (intercellular
development, recent work (Batten et al., 2000; adhesion molecule), VCAM, PNAd (peripheral node
Schiemann et al., 2001; Claudio et al., 2002) has under- addressin), GlyCAM-1 (glycosylation-dependent cell
scored the importance of BAFF ligation in selectively adhesion molecule) and MadCAM (mucosal addressin
activating the non-canonical NF-kB pathway and the cellular adhesion molecule), cytokines including TNFa,
consequent expression of antiapoptotic Bcl-2 family and organogenic chemokines such as CXCL12 (GRO/
members in transitional B cells (see Mackay et al., 2003). MIP-2), CXCL13 (BLC), CCL19 (ELC) and CCL21
Indeed, BAFF knockout mice exhibit a complete failure (SLC), are regulated by NF-kB.
Oncogene
NF-jB and immunology
MS Hayden et al
6763
normal B-cell follicles, germinal centers (GCs) and
Chemokines Peyer’s patch development (Caamano et al., 1998;
VCAM-1
Franzoso et al., 1998; Paxian et al., 2002); RelB is
Hematopoietic likewise also required for Peyer’s patch development
cell (Yilmaz et al., 2003). Although LN development occurs
α4β1
in RelB knockout mice, the nodes are small at birth and
are resorbed perinatally. In addition to LTbR, knock-
outs of RANK, which likewise signals through the non-
LTα1β2
canonical pathway, also lack peripheral LNs (Dougall
LTβR et al., 1999).
Splenic architecture is crucial for B-cell development
as well as for the initiation and maturation of B-cell
RANK Local stromal responses. The spleen is divided histologically into white
RANKL
cell and red pulp zones. Macrophages in the red pulp are
responsible for destroying erythrocytes that are da-
maged or have reached the end of their lifespan. The
white pulp is populated by splenic lymphocytes and
consists of B-cell follicles and T-cell zones. Splenic
architecture allows for dynamic changes, most notably
in the formation of GCs, during the initiation and
maturation of B-cell responses. Multiple NF-kB knock-
Figure 3 NF-kB function in the early events of lymphoid outs exhibit defects in some aspect of splenic architec-
organogenesis. NF-kB is a vital part of the positive feedback loop ture; however, as for other lymphoid organs, the
between hematopoietic and stromal cells that comprises the early analysis of splenic architecture has been complicated
events of lymphoid organogenesis. LTa1b2-expressing hematopoie-
tic cells induce production of VCAM-1 through the canonical
by defects that occur upon deletion of TNF-R used to
NF-kB pathway and chemokines through the non-canonical rescue the embryonic lethality. Nevertheless, there has
(IKKa-dependent) pathway in LTbR-expressing stromal cells. been considerable progress in deciphering the role of
Stromal expression of chemokines induces the upregulation of NF-kB family members in development and mainte-
integrins (a4b1) on hematopoietic cells resulting in increased nance of splenic architecture. Mice in which RelA has
recruitment of LTa1b2-expressing cells and signaling through
stromal LTbR. RANKL stimulation of NF-kB through TRAF6 been targeted for deletion exhibit aberrant segregation
is also crucial for the upregulation of LTa1b2 in hematopoietic cells. of B- and T-cell areas and defects in one particular
macrophage population, the metallophilic marginal
zone macrophages. In addition, rela//tnfr1/ spleens
have a more pronounced defect in GC generation
Lymphoid organogenesis exhibits distinct requirements following immunization, than do tnrf1/ mice (Alcamo
for both the canonical and non-canonical NF-kB et al., 2002). However, it is worth emphasizing that
pathways. Signaling through TNF-R, LTbR and defects observed in tnfr1/ animals may, in fact, be due
RANK activates canonical RelA-containing complexes to changes in RelA-dependent responses, and it is
and, hence, it is not surprising that rela//tnfr1/ possible that the role of the RelA/canonical NF-kB
double knockout mice lack Peyer’s patches and LNs pathway in the spleen is underappreciated.
and exhibit disorganized spleens (Alcamo et al., 2002). The importance of the non-canonical pathway in the
The requirement for RelA in development of these spleen has been observed in multiple circumstances.
tissues lies with the stromal cells and is likely due to a Mice in which non-canonical pathway components,
combination of effects: regulation of apoptosis (e.g., that RelB, NIK or IKKa, have been inactivated demonstrate
induced by TNF); regulation of expression of orga- severe defects in splenic architecture, similar to that seen
nogenic factors including VCAM and LTa1b2; and en- in ltbr/ spleens. These defects largely reflect deficien-
hancement of the non-canonical p52/RelB pathway cies in splenic stromal cells (Miyawaki et al., 1994;
through the LTbR signaling pathway. Koike et al., 1996). Mice deficient in the non-canonical
Several lines of evidence highlight the importance of pathway fail to segregate B-cell–T-cell zones and FDC
the non-canonical pathway and activation of p52/RelB networks, and they fail to form GCs following
complexes in LN development. Mice with a point immunization. Marginal zone macrophages, which line
mutation in nik (aly/aly mice) lack multiple secondary the border between red and white pulp areas, are also
lymphoid organs (Miyawaki et al., 1994; Koike et al., absent or disorganized in RelB, p52, NIK or IKKa
1996; Shinkura et al., 1999) and share several pheno- knockouts (Franzoso et al., 1998; Weih et al., 2001).
typic similarities with lymphotoxin and IKKa single Some splenic defects are also attributable to effects on
knockout animals (Mebius, 2003; Bonizzi and Karin, hematopoietic cells. For example, the presence of
2004). p52/RelB, which is activated downstream of NIK metallophilic marginal zone macrophages depends on
and IKKa, is thought to be the primary transcriptional p52 (Franzoso et al., 1997). Finally, knockout of the
mediator of several key organogenic factors including atypical IkB family member BCL-3 also leads to
CXCL12, CXCL13, CCL19, CCL21 and MadCAM-1 alterations in lymphoid architecture that are reminiscent
(Yilmaz et al., 2003). The p52 single knockout lacks of those seen in the absence of p52, with which BCL-3
Oncogene
NF-jB and immunology
MS Hayden et al
6764
forms a transcriptionally active complex. BCL-3 knock- have been shown to alter TLR expression under
out mice lack splenic GCs, and although they exhibit inflammatory conditions (Mueller et al., 2006). How-
normal serum antibody levels, they fail to develop ever, sentinel cells of the innate immune system,
antigen-specific humoral responses (Sha et al., 1995; particularly tissue resident DCs and macrophages,
Caamano et al., 1996; Franzoso et al., 1997; Schwarz express a more complete complement of PRRs, and
et al., 1997). thus are likely to bear the largest portion of the burden
In summary, both the canonical and non-canonical in the earliest events of pathogen recognition.
NF-kB pathways are required for the development of
most secondary lymphoid organs. However, the role of Toll-like receptors
the non-canonical pathway, as assessed by examining TLRs are evolutionarily conserved PRRs that recognize
mice deficient for IKKa, p52, NIK or RelB, is especially unique, essential molecules characteristic of various
important both during organogenesis and maintenance classes of microbes (Akira et al., 2006). The function
of splenic architecture. Recent data suggest that the of TLRs as arbitrators of self/non-self discrimination
non-canonical pathway is also important in thymic highlights their central role in innate immunity as well as
development and organization (Burkly et al., 1995; Weih in the initiation of the adaptive immune response. The
et al., 1995; Kajiura et al., 2004; Kinoshita et al., 2006). 11 characterized mammalian TLRs have varied tissue
However, it is important to note that canonical NF-kB distribution and serve as recognition receptors for
pathway function in these events may be underappre- pathogen-associated molecular patterns (PAMPs) pre-
ciated owing to embryonic lethality and complicated by sent on bacteria, viruses, fungi and parasites. Perhaps
the defects introduced by crossing them onto the tnfr/ due to the multimeric nature of the TLR extracellular
background. Nevertheless, our understanding of non- domain (ED), which consists of multiple leucine-rich
canonical pathway function in secondary lymphoid repeats (LRRs), several receptors are capable of
organs is consistent with the ability of RelB-containing recognizing more than one microbial molecule (Figure 4
complexes to regulate genes encoding key organogenic and below). Heterodimerization of some TLRs and the
chemokines and adhesion molecules that direct leuko- use of co-receptors (e.g., CD14 and MD-2) further
cyte trafficking. The functional consequences of defects expand the repertoire of PAMPs recognized. As we shall
in these processes are severe and have direct ramifica- see below, the ability of TLRs to distinguish between
tions for the host’s ability to mount a robust immune pathogen types is translated into appropriate innate and
response. Alterations in lymphoid architecture likewise adaptive responses through the selective activation of
impede the initiation of the adaptive response as well as NF-kB and other inducible transcription factors. Sig-
the fine-tuning of this response through processes such nificant progress has been made over the past few years
as B-cell affinity maturation. in deciphering the relevant signaling pathways that
operate downstream of TLRs in particular.

Role of NF-jB in the innate response TLR signaling to NF-kB


Ligand binding to TLRs is just now beginning to be
Pattern recognition receptors understood at the molecular level. Extracellular LRRs
To activate an appropriate immune response, the host bind to ligand and, either through receptor oligomeriza-
must first recognize the presence of pathogens. This tion and/or induction of a conformational change across
discrimination between self and non-self is an absolute the plasma membrane, induce the recruitment/activa-
requirement for the initiation of effector functions, such tion of adapter proteins through the Toll/IL-1 Receptor
as the secretion of cytokines and antimicrobial peptides, (TIR) domain. These adapters lead to the activation of
carried out by the cells of the innate immune system. A canonical IKKb-dependent complexes, degradation of
number of pattern recognition receptors (PRRs) have IkBa and IkBb, and liberation of, primarily, RelA and
evolved to recognize microbial invaders. These PPRs c-Rel containing NF-kB complexes. TLR signaling to
include TLRs, members of the CATERPILLAR/NOD NF-kB is divided into two pathways: those that are
family of cytoplasmic receptors, scavenger receptors and MyD88 (myeloid differentiation primary response gene
the complement system. Although epithelial cells are 88)-dependent and those that are MyD88-independent
frequently the first to encounter pathogens, they are also (Figure 5). We will base our discussion primarily on
constantly exposed to non-pathogenic microbes. There- signaling events emanating from TLR4, which despite
fore, whereas a variety of TLRs are differentially having the most complex downstream pathways is the
expressed in epidermis, gut, pulmonary, urinary and most thoroughly studied TLR. Clear differences exist in
reproductive epithelium, in many cases it is thought that signaling from other TLRs as noted throughout our
both TLR expression and responsiveness is tightly discussion, and it is likely that further specializations
controlled in these cells. For example, keratinocytes will become apparent as individual TLR signaling
upregulate TLRs expression and responsiveness follow- pathways are investigated more thoroughly.
ing transforming growth factor-alpha (TGFa) exposure
(Miller et al., 2005); renal epithelial cells increase MyD88-dependent signaling to NF-kB. TLR4 signaling
expression of TLR2 and TLR4 in response to IFNg or is relatively unique amongst TLRs in that the effector
TNFa (Wolfs et al., 2002) and intestinal epithelial cells adaptors are one step removed from the receptor. For
Oncogene
NF-jB and immunology
MS Hayden et al
6765

Lipopolysaccharide (LPS)
Lipoteichoic Acid (LTA)
Glycoinositolphospholipids
Diacyl Lipopeptides
Triacyl Lipopeptides Glucuronoxylomannan Flagellin
Yeast Zymosan
Peptidoglycan (PGN) Mannan

TLR-2 TLR-6 TLR-4 TLR-5 TLR-10 TLR-12

TLR-1 TLR-2

Gamma-D-glutamyl-meso- PFTG TLR-11


diaminopimelic acid (iE-DAP) NOD1 UPEC
dsRNA
ssRNA
CpG-DNA
Muramyl Dipeptide (MDP) NOD2 Hemozoin

dsRNA RIG-I TLR-3

dsRNA TLR-9
MDA5 TLR-7 TLR-8

Figure 4 PRRs that signal to NF-kB and their cognate ligands. TLRs 3, 7, 8, 9 and 11 have been reported to exhibit endosomal or
intracellular localization whereas NOD1, NOD2, RIG-I and MDA5 function in the cytoplasm.

example, MyD88 recruitment to the receptor complex pathways), was identified because of its interaction with
depends upon the TIR-domain containing adapter TRAF6. ECSIT binds to TRAF6 and is required for
protein (TIRAP, also known as Mal) (Fitzgerald et al., TLR and interleulin-1 (IL-1) signaling, but not TNF-
2001; Horng et al., 2002; Yamamoto et al., 2002). TLR2 signaling (Kopp et al., 1999; Xiao et al., 2003). Although
also requires TIRAP to bridge MyD88 to the receptor; these studies suggested that ECSIT functions by
however it is believed that other MyD88-utilizing TLRs recruiting and activating the kinase MEKK1 (mitogen
directly recruit MyD88. Recent reports suggest that the activated protein kinase or ERK kinase (MEK) kinase
requirement for these intermediatory adapters is related 1) (Kopp et al., 1999; Xiao et al., 2003), the role of
to localization of the TLRs to certain domains in the MEKK1 in TLR signaling remains unclear (Xia et al.,
plasma membrane (Kagan and Medzhitov, 2006; Rowe 2000; Yujiri et al., 2000). MEKK3-deficient cells,
et al., 2006). The N-terminal domain of MyD88 however, do not transcribe IL-6 following TLR4 or
contains a death domain (DD) that recruits the DD- IL-1R stimulation and exhibit delayed and weakened
containing serine/threonine kinase interleukin-1- NF-kB DNA binding following lipopolysaccharide
associated kinase-4 (IRAK-4). IRAK-4 and IRAK-1 (LPS) stimulation (Huang et al., 2004). ECSIT interacts
form an active complex capable of recruiting the TNF with both TAK1 and MEKK3 (AP West and S Ghosh,
receptor-associated factor TRAF6 (Figure 5a). The link unpublished observations) and it is, therefore, possible
between TRAF6 and the IKK complex remains some- that ECSIT exerts its role in TLR signaling by
what enigmatic, although a few key players are known. modulating the function of TAK1 and/or MEKK3.
The kinase TAK1 (TGFb-activated kinase-1) is required
for NF-kB, as well as AP-1 and extracellular signal-
related kinase (ERK), activation downstream of MyD88 TRIF-dependent signaling to NF-kB
(Sato et al., 2005; Shim et al., 2005). Although it is Somewhat unexpectedly, when exposed to LPS,
widely accepted that ubiquitination is a key switch at MyD88/ cells display partial NF-kB activation, albeit
this crucial step of NF-kB activation, considerable work with slower kinetics than in wild-type cells (Kawai et al.,
at the molecular level remains to be done to understand 1999). When cells are stimulated through TLR3 and
how ubiquitination leads to activation. TLR4, TRIF (TICAM-1), a TIR-domain containing
In addition to TAK1, another protein, termed ECSIT adapter, mediates activation of NF-kB in the absence of
(evolutionarily conserved signaling intermediate in Toll MyD88 (Oshiumi et al., 2003). Furthermore, in the case
Oncogene
NF-jB and immunology
MS Hayden et al
6766

LPS

TLR4

TIRAP b
a TRAM
c dsRNA
MyD88 TRIF

RIGI
IRAK1

IRAK4

IPS-1
IKKi
FADD d
TRAF6 Ub TBK1 NOD2
RIP1

Ub MDP
ECSIT TRAF6 RIP1
TAK1 P

Ub IRF3
NEMO RIP2
P
Ub
NEMO
P
PP
NF- κB

Gene
Transcription

Figure 5 PRR signaling to NF-kB. Signaling through LPS/TLR4 via the MyD88-dependent (b) and TRIF-dependent (a) pathways
converge on IKK activation through TRAFs. Signaling through dsRNA/RIG-I (c) proceeds through ISP1 to IKKi/TBK1 and through
RIP1 to IKK. Signaling from NOD to NF-kB (d) is thought to involve oligomerization of RIP2 and activation of IKK through
induced proximity. See the text for details.

of TLR3, all downstream signaling appears to be TRIF- (TRIF-related adapter molecule) functions upstream of
dependent. In TLR4 signaling, TRIF is required for TRIF in MyD88-independent signaling from TLR4.
late-phase NF-kB and IRF3 responses, but is not TRAM is required for IRF3 activation and for the
required for activation of JNK (Yamamoto et al., delayed phase of NF-kB activation following TLR4
2003). TRIF signaling to NF-kB and IRF3 also appear engagement. TLR4-induced IRAK activation by
to be separately regulated (Figure 5b). Signaling to IRF- MyD88, however, is unaffected by the absence of
3 occurs through two divergent members of the IKK TRAM and TRAM does not function in TLR3 TRIF-
family, IKKi (IKKe) and TBK1 (T2K) (Fitzgerald et al., dependent signaling pathways (Fitzgerald et al., 2003;
2003; Sharma et al., 2003); however, neither kinase is Yamamoto et al., 2003). Therefore, it appears that
required for NF-kB activation by LPS or TNFa TRAM is only needed for TRIF signaling downstream
(Hemmi et al., 2004; McWhirter et al., 2004). Further- of TLR4. Adding further complexity, it has recently
more, reconstitution of trif/ cells with mutant TRIF been suggested that TLR4, but not TLR3, TRIF-
lacking the TRAF-binding domain selectively restores dependent NF-kB activation is largely due to IRF3-
induction of IRF3 but not NF-kB. Increasingly, there- induced TNFa rather than to direct signaling to IKK
fore, it appears that the events leading from TRIF to (Covert et al., 2005; Werner et al., 2005). Although
IKK activation share a common set of intermediates as the applicability of these finding to other cell types is, as
seen in other NF-kB activation pathways. of yet, unclear, these results may be explained by
TRIF interacts with receptor interacting protein differences in the recruitment of TRIF to the
(RIP)1 and RIP3 through their RIP homotypic interac- receptor; that is, by TRAM in the case of TLR4 versus
tion motif (RHIM), and rip1/ embryonic fibroblasts directly to TLR3, resulting in changes in the avail-
have decreased NF-kB activation following TLR3- ability of TRAF binding site or the availability of
poly(I:C) signaling (Meylan et al., 2004). Finally, additional signaling intermediates at distinct subcellular
another TIR-domain containing adapter TRAM localizations.
Oncogene
NF-jB and immunology
MS Hayden et al
6767
Negative regulation of TLR signaling express the full panoply of antiviral TLRs – suggesting
Inflammatory responses are built upon waves of that other PRRs must be at work. In fact, cells do have
cytokine production and positive feedback mechanisms. at their disposal families of cytoplasmic PRRs that are
As a result, tight control must be placed on the initiation capable of activating NF-kB and other transcriptional
and maintenance of these responses. Multiple negative mediators of the innate immune response. Interestingly,
feedback loops have been described that involve many of these PRRs contain caspase activation and
proteins that are induced or activated upon TLR recruitment domains (CARDs) that are required for
signaling. In a number of instances, the target of these activation of NF-kB following ligand binding. Here, we
regulatory mechanisms is the IRAK family of proteins. provide a brief description of two classes of cytoplasmic
For example, IRAK-M (IRAK3) inhibits signaling to PRRs – CARD-containing members of the CATEPIL-
TRAF6 by fixing IRAK-1/4 to the TLR/MyD88 LAR and DExD/H-box helicase families.
signaling complex; irakm/ knockouts exhibit enhanced
signaling to NF-kB (Kobayashi et al., 2002). Tollip, an
CATEPILLER-NODs. Nucleotide oligomerization
adapter protein constitutively associated with IRAK, is
domain proteins (NOD) 1 and 2 are part of a large
phosphorylated and dissociates following IRAK4 acti-
family termed the CATEPILLER family, which is
vation (Burns et al., 2000; Zhang and Ghosh, 2002).
named for CARD, transcription enhancer, R (purine)-
Negative regulation of TLR signaling by Tollip in the
binding, pyrin, lots of leucine repeats (Figure 4). The
intestinal epithelium may prevent inflammatory re- NOD-LRR subfamily is typified by the presence of
sponses to commensal bacteria (Melmed et al., 2003).
LRRs and nucleotide oligomerization domains. NOD1,
However, Tollip-deficient cells demonstrate only minor
NOD2 and IPAF have CARDs and can signal to NF-
defects in the production of NF-kB-regulated cytokines kB (for a review see Inohara and Nunez, 2003). NOD1
(Didierlaurent et al., 2006). Therefore, it is unclear
recognizes a peptidoglycan containing meso-diaminopi-
whether Tollip indeed functions as initially thought.
melic acid (meso-DAP) and induces NF-kB through a
SIGIRR (TIR8), a member of the IL-1R family, binds
canonical pathway that includes activation of IKKb.
to Toll/IL-1 receptors, IRAK and TRAF6 and may also
NOD2 recognizes muramyl dipeptide, a ubiquitous
function by inhibiting the association of IRAK with
component of nearly all bacterial cell walls. Relatively,
TLRs (Thomassen et al., 1999; Wald et al., 2003).
few signaling intermediates downstream of NOD-LRRs
SIGIRR deficiency yields prolonged activation of
are known; however, there is growing evidence that the
NF-kB by Toll/IL-1 stimulation consistent with a
CARD-containing kinase RIP2 (RICK) is required for
regulatory function. Interestingly, SIGIRR is also highly
NF-kB activation. Intriguingly, the ATP-binding cas-
expressed in epithelial cells, suggesting that it too may sette of both NOD1 and NOD2 is needed for signaling
suppress signaling at sites of constitutive microbial
(Tanabe et al., 2004). RIP2 binds to NEMO and
exposure. Finally, suppressor of cytokine signaling-1
therefore is thought to directly mediate activation of
(SOCS-1) has been reported to negatively regulate LPS
the IKK complex by induced proximity (Inohara et al.,
signaling to NF-kB and socs1/ mice exhibit an
2000). In this model, ligand-dependent oligomerization
inflammatory phenotype that is consistent with this
of NOD-LRRs, which is dependent on the ATP-binding
prediction (Kinjyo et al., 2002; Nakagawa et al., 2002).
cassette, leads to a scaffold containing multiple RIP2
SOCS-1 may function by directly targeting TIRAP/Mal,
molecules, which allows trans-autophosphorylation of
and selectively inhibit TLR4 signaling through the
neighboring IKK complexes (Figure 5d).
TIRAP/Mal/MyD88 pathway (Mansell et al., 2006).
In addition to these TLR-specific regulators of
signaling to NF-kB, other proteins function to control Retinoic acid inducible gene I and melanoma
the extent and duration of NF-kB activation. These differentiation-associated gene 5
factors both set thresholds for activation and help to Two members of the DExD/H-box RNA helicase family
prevent uncontrolled, and potentially deleterious, innate stand out because of the presence of N-terminal CARD
immune responses. The broad array of PAMPs recog- domains. Retinoic acid inducible gene I (RIG-I) and
nized by the TLR system affords the host the ability to melanoma differentiation-associated gene 5 (MDA5) are
mount responses against many pathogens. Nevertheless, RNA helicase-containing cytoplasmic proteins. The
for some pathogens, TLRs alone are not sufficient, and RNA helicase domains of RIG-I and MDA5 bind
some physical spaces, most notably the cytosol, are not directly to double-stranded RNA (dsRNA) and induce
effectively monitored by TLRs. production of type I interferons (Kang et al., 2002;
Andrejeva et al., 2004; Yoneyama et al., 2004). Upon
binding to dsRNA, representing either the viral genome
Cytoplasmic PRRs that activate NF-kB or viral replication intermediate, RIG-I and MDA5
PRRs that recognize bacterial PAMPs are expressed at induce the activation of IRF3 and NF-kB. Interestingly,
the plasma membrane or with LRRs projecting into the initiation of these signaling cascade is abrogated by
lumen of vesicles that are topologically related to the point mutations in the Walker-type ATP-binding site,
extracellular space. However, in such a system, intra- suggesting that their ATPase activity is required for
cellular pathogens are uniquely protected from detec- signaling (Yoneyama et al., 2004). The link between
tion. Furthermore, viral infection and the resulting these two proteins and NF-kB remains somewhat
induction of interferon occurs in many cells that do not unclear (Figure 5c); however overexpression of the
Oncogene
NF-jB and immunology
MS Hayden et al
6768
N-terminal CARD domain alone is sufficient to induce thoroughly studied and the most potent TLR ligand
signaling. Recently, a CARD-containing protein, vari- known. Trace amounts of LPS activate the innate
ably named CARDIF, IPS1, MAVS and VISA, has immune system via TLR4, leading to the production of
been implicated downstream of RIG-I; however, the numerous proinflammatory mediators, such as TNFa,
link between this protein and IKKb is unclear (Kawai IL-1 and IL-6. TLR4-mediated responses to LPS require
et al., 2005; Meylan et al., 2005; Seth et al., 2005; Xu CD14 and MD-2. Other bacterial TLR4 ligands include
et al., 2005). It appears, however, that there are Lipid A analogs (Lien et al., 2001) and mycobacterial
similarities to TRIF mediated signaling, in that RIG-I components (Means et al., 1999).
activation of NF-kB requires FADD and RIP-1 TLR5 recognizes flagellin, a protein component of
(Balachandran et al., 2004; Yoneyama et al., 2005). Gram-negative bacterial flagella and virulence factor for
Recently, it was shown that RIG-I and MDA5 multiple human pathogens (Hayashi et al., 2001). In
differentially recognize various groups of RNA viruses light of the fact that it was thought that proteins would
and are thus critical for a robust antiviral response be too mutable to serve as PAMPs, it is notable that
(Kato et al., 2006). TLR5 recognizes a highly conserved, central core
structure of flagellin that is essential for protofilament
assembly (Smith et al., 2003). Interestingly, the TLR5
recognition site is masked in the filamentous flagellar
Pathogen recognition in innate immunity structure, thus indicating that TLR5 recognizes only
monomeric flagellin (Smith et al., 2003). Furthermore,
Bacterial recognition flagellin appears to bind directly to TLR5 at residues
Pathogens recognized by PRRs can be categorized as 386–407, as TLR5 mutants lacking this domain are
bacterial, viral or eukaryotic. In each of these categories, unable to interact with flagellin in biochemical assays
PAMPs have been described that more or less fit with (Mizel et al., 2003). Recent articles have demonstrated
existing hypotheses of how pathogen recognition by the TLR5-independent recognition of cytosolic Salmonella
innate immune system should occur (Janeway, 1989). typhimurium flagellin via Ipaf, a member of the NOD-
Both in terms of accessibility and uniqueness to LRR family (Franchi et al., 2006; Miao et al., 2006).
prokaryotes, the bacterial cell well is a logical source Ipaf-mediated recognition of cytosolic flagellin induces
of PAMPs for TLRs and other PRRs. caspase-1 activation and subsequent IL-1b secretion by
LPS was originally thought to be the ligand for macrophages. TLR11 recognizes a protein PAMP that is
TLR2, but subsequent studies revealed that contaminat- present on uropathogenic Escherichia coli (Zhang et al.,
ing bacterial lipoprotein in LPS preparations is the 2004). Although the identity of this ligand is unknown,
actual ligand (Wetzler, 2003). TLR2 also mediates its ability to stimulate in a TLR11-dependent manner is
responses to several Gram-positive bacterial cell wall destroyed by proteinase treatment.
components as well as Staphylococcus aureus peptido- Conserved differences in bacterial nucleic acid struc-
glycan (Takeuchi et al., 2000). Additional work has tures can also be recognized by the innate immune
shown that TLR2 is involved in the recognition of a system. TLR9 recognizes bacterial DNA containing
wide range of microbial products and generally func- unmethylated CpG motifs, and TLR9-deficient mice are
tions as a heterodimer with either TLR1 or TLR6 not responsive to CpG DNA challenge (Hemmi et al.,
(Ozinsky et al., 2000; Wyllie et al., 2000). The TLR2/ 2000). The low frequency and high rate of methylation
TLR1 heterodimer recognizes a variety of lipoproteins, of CpG motifs prevent recognition of mammalian DNA
including those from mycobacteria and meningococci by TLR9 under physiological circumstances. A recent
(Takeuchi et al., 2002; Wetzler, 2003), whereas the report indicated that the intracellular, endosomal
TLR2/TLR6 complex recognizes mycoplasma lipopro- restriction of TLR9 is critical for discriminating between
teins and peptidoglycan (Takeuchi et al., 2001). Recent self and nonself DNA, as host DNA, unlike microbial
reports have demonstrated that triacylated lipoproteins DNA, does not usually enter the endosomal compart-
from bacteria are preferentially recognized by the ment (Barton et al., 2006).
TLR1/TLR2 complex, whereas diacylated lipoproteins
are recognized by the TLR2/TLR6 complex (Takeuchi
et al., 2002). However, additional TLR2 ligands do not Viral recognition
require TLR1 or TLR6 for signaling, implying that Although viruses are composed entirely of host products
TLR2 recognizes some ligands as a homodimer or they, nevertheless, have unique components that readily
heterodimer with other non-TLR molecules. Such TLR2 serve as PAMPs. Nucleic acids are also key viral
ligands include the Gram-positive cell wall component PAMPs, and are recognized by TLRs 3, 7, 8 and 9, as
lipoteichoic acid; the mycobacterial cell wall component well as by cytoplasmic receptors of the RIG family (as
lipoarabinomannan; atypical LPS produced by Legionella, described above). TLR3 recognizes dsRNA, a common
Leptospira interrogans, Porphyromonas gingivitis viral replicative intermediate (Alexopoulou et al., 2001).
and Bordetella; and porins present in the outer TLR3 signaling results in the activation of NF-kB
membrane of Neisseria (Massari et al., 2003; Wetzler, and IRF3, ultimately leading to the production of anti-
2003). viral molecules, such as type I interferons (IFN-a/b)
The TLR4 ligand LPS, a glycolipid component of the (Alexopoulou et al., 2001). The importance of RIG-I-
outer membrane of Gram-negative bacteria, is the most and MDA5-mediated viral recognition is further
Oncogene
NF-jB and immunology
MS Hayden et al
6769
supported by gene-targeting experiments demonstrating reported to recognize the malarial pigment hemozoin,
that TLR3 and its adaptor TRIF are not required for a byproduct of heme metabolism in infected erythro-
type I IFN production in some virally infected cells, cytes (Coban et al., 2005) whereas TLR11 recognizes a
such as fibroblasts and conventional DCs (Honda et al., profilin-like protein that is conserved in apicomplexan
2003). However, plasmacytoid DCs exclusively utilize parasites including Toxoplasma gondii (Yarovinsky
TLR3/TRIF signaling for type I IFN production in et al., 2005).
response to RNA viruses and poly(I:C) (Kato et al.,
2005).
Although initially found to recognize synthetic anti- Immediate antimicrobial responses
viral compounds, namely imidazoquinolines, the azo-
quinoline R-848 and loxoribine (Hemmi et al., 2002; PRRs initiate a complex series of events following
Jurk et al., 2002), TLR7 and human TLR8 are now exposure to certain microbial components: the first is the
known to recognize guanosine- or uridine-rich single- mounting of immediate antimicrobial responses at the
stranded RNA derived from RNA viruses (Diebold cellular level. This is an effective and evolutionarily
et al., 2004; Heil et al., 2004; Lund et al., 2004). conserved function of PRRs, and one in which NF-kB
Interestingly, mammalian RNA, which contains many has an important role. The liberation of products with
modified nucleosides, is significantly less stimulatory via direct antimicrobial activity occurs early at sites of
TLRs 7 and 8 than bacterial RNA, suggesting that pathogen entry. TLR ligation is at least partly respon-
nucleoside modification allows mammals to distinguish sible for the NF-kB-dependent expression of defensins –
between endogenous and pathogen-derived RNA cationic peptides that exert direct bactericidal activity by
(Kariko et al., 2005). Similar to TLR3, engagement of inducing membrane permeabilization. Small intestinal
these receptors leads to the production of type I IFNs. Paneth cells, for example, release large amounts of
TLR9 recognizes viral CpG sequences and induces the a-defensins into the intestinal lumen following exposure
induction of IFN-a (Takeshita et al., 2001; Lund et al., to a variety of bacteria/bacterial products (Ayabe et al.,
2003; Krug et al., 2004). However, as membrane 2000). The production of antimicrobial nitrogen and
restriction prevents TLRs from sampling the cytosol oxygen species, which are acutely toxic to a variety of
where much of the viral life cycle occurs, cytosolic PRRs microbes, augments the activity of antimicrobial pep-
provide comprehensive innate immune recognition. For tides. Production of nitric oxide is mediated in part by
example, recognition of cytoplasmic dsDNA leading to inducible nitric oxide synthase (iNOS), which is partially
NF-kB activation and type I interferon production has regulated by NF-kB. Consequently, iNOS production
also been reported, although the relevant receptor has results from TLR or NOD-LRR ligation by PAMPs.
not yet been identified (Ishii et al., 2006; Stetson and Much of the early innate response has been demon-
Medzhitov, 2006). This receptor(s) is predicted to be strated to depend on the canonical NF-kB pathway.
important for type I IFN production in response to Thus, rela//tnfr1/ double knockout mice have
viruses and intracellular pathogens, such as Listeria increased susceptibility to bacterial infection (Alcamo
monocytogenes and Shigella flexneri. Finally, there have et al., 2001). Likewise, B cells from p50/ mice do not
been some reports suggesting that certain viral proteins respond efficiently to LPS, emphasizing the importance
function as PAMPs. For example, TLR4 may recognize of p50-containing complexes, that is, p50/RelA, p50/
respiratory syncytial virus (RSV) F protein (Kurt-Jones p50/BCL-3 and p50/c-Rel, in TLR signaling (Sha et al.,
et al., 2000). 1995). As might be expected, TNFR/IKKb double
knockouts show a more pronounced defect in innate
responses owing to the more complete block in
Recognition of other pathogens canonical NF-kB pathways, and succumb to infection
MyD88-deficient cells demonstrate that many fungal more rapidly than rela//tnfr1/ mice (Li et al.,
species are capable of activating TLR pathways, 1999a, b; Senftleben et al., 2001b). Furthermore, MEFs
although the receptors have not always been identified. from nemo/ mice do not exhibit NF-kB activation by
TLR4 has been shown to recognize Aspergillus hyphae LPS or IL-1 (Rudolph et al., 2000). Therefore, activa-
(Mambula et al., 2002), and Cryptococcus neoformans tion of NF-kB responsive genes by the innate immune
capsular polysaccharide (Shoham et al., 2001). TLR2 system depends on NEMO and likely progresses
and TLR6 are required for recognition of yeast through the canonical NF-kB signaling pathway.
zymosan, whereas TLR4 is thought to recognize certain
yeast mannans (for a review see Levitz, 2004). The
identification of parasite PAMPs has been more elusive, Inflammation
and their existence is somewhat controversial. However,
TLR2 heterodimers reportedly recognize various para- There is a staggering amount of literature that correlates
site GPI-anchored proteins and glycoinositolphospholi- NF-kB activation with inflammation in a wide array of
pids from the parasitic protozoa Trypanosoma cruzi diseases and animal models. There are, likewise,
(Campos et al., 2001). Some TLR knockout mice have numerous studies using gene targeting and inhibitors
been shown to have variable defects in their ability to of NF-kB that have established that NF-kB plays a
defend against various parasites (for a review see causative role in inflammatory processes. We have
Gazzinelli et al., 2004). Recently, TLR9 has been already discussed the role of NF-kB in the survival of
Oncogene
NF-jB and immunology
MS Hayden et al
6770
leukocytes, and how this role is particularly important responses, in most cases signaling converges on the
during the responses that include inflammation. Here, activation of NF-kB and AP-1. NF-kB activation in
we briefly discuss a few of the additional ways in which response to TNF signaling induces expression of
NF-kB regulates inflammation. Inflammation begins antiapoptotic genes such as cIAP1/2 and Bcl-XL (see
with epithelial or stromal cells of the infected tissue or Dutta et al., 2006).
tissue resident hematopoietic cells such as mast cells or TNF family receptors lack intrinsic enzymatic acti-
DCs recognizing an inflammatory stimulus and propa- vity. Instead, signaling is achieved by recruitment of
gating proinflammatory signals. These signals lead to intracellular adapter molecules that associate with the
the recruitment and activation of effector cells, initially cytoplasmic tail of the TNF-R in a signal-dependent
neutrophils and later macrophages and other leuko- manner (Figure 6a). The recruitment of TNF-R1 to
cytes, resulting in the tissue changes characteristic of membrane microdomains, referred to as lipid rafts, with
inflammation – rubor, calor, dolor and tumor (redness, subsequent assembly of the signaling complex, is
heat, pain and swelling, respectively). necessary for signaling to NF-kB and prevention of
As for the immediate antimicrobial products dis- apoptosis (Hueber, 2003; Legler et al., 2003). Ligation of
cussed above, NF-kB is responsible for the transcription TNF-R1 by trimeric TNFa causes aggregation of the
of the genes encoding many proinflammatory cytokines receptor allowing binding of the TNF-R-associated
and chemokines. One important early target of these death domain protein (TRADD). TRADD subse-
effectors is the vascular endothelium. Changes in quently recruits adapter molecules including TRAF2
vascular endothelial cells both recruit circulating leuko-
cytes and provide them with a means of exiting the
vasculature into the infected tissue. NF-kB regulates the
expression of adhesion molecules, both on leukocytes
a b
and endothelial cells, which allow the extravasation of
leukocytes from the circulation to the site of infection
(Eck et al., 1993). Indeed, RelA-deficient mice display a
severe defect in the recruitment of circulating leukocytes
to sites of inflammation (Alcamo et al., 2001). RIP1 Ub
Recruited neutrophils are the key mediators of local FADD
RIP1 Ub
TRAF2/5/6
TRADD
inflammation and NF-kB is important for the survival TRAF2/5
of these cells, which must function in relatively toxic
TRAF3
conditions (Ward et al., 1999). NF-kB is important for TAK1 NIK
the production of the enzymes that generate prostaglan- MEKK3
dins and reactive oxygen species (e.g., iNOS and Cox,
Ub
both NF-kB target genes) and may, furthermore, be P NEMO
P

involved in the signaling induced by prostaglandins


(Poligone and Baldwin, 2001; Catley et al., 2003).
NF-kB has also been implicated in the response to P P
leukotrienes, which like prostaglandins are short-lived NF- κB p100 RelB
paracrine effectors, although it is unclear whether this
represents a direct signaling event. Finally, matrix
metalloproteinases (MMPs) also are crucial mediators
of local inflammation and leukocyte chemotaxis; and
their expression is also regulated by NF-kB (Vincenti
et al., 1998; Vincenti and Brinckerhoff, 2002; Lai et al., Gene
p52 RelB Transcription
2003).
The pathway from pathogen recognition to proin-
flammatory cytokine production demonstrates a parti-
cular reliance on NF-kB. The immediate targets of
NF-kB-dependent proinflammatory cytokines, such as Figure 6 TNF receptor superfamily signaling to NF-kB. Activa-
tion of the canonical NF-kB pathway downstream of TNF-RI is
TNFa, tend to be receptors that, in turn, activate NF-kB. initiated by trimerization through ligand binding and recruitment
Therefore, NF-kB is crucial to the propagation and of FADD, TRADD, RIP1 and TRAF2/5 to the receptor
elaboration of cytokine responses. TNFa is particularly (a). TAK1 and MEKK3 are subsequently recruited to the receptor
important for both local and systemic inflammation, complex through RIP1, and with TRAF2/5, mediate the activation
and it is a potent and well-studied inducer of NF-kB. of IKK. The phosphorylation and degradation of classical IkBs
require IKKb and NEMO. The non-canonical pathway is mediated
by IKKa through NIK (b). In the resting state, NIK is inactivated/
degraded through an interaction with TRAF3. Upon stimulation,
TNF-R superfamily signaling TRAF3 is inactivated/degraded resulting in the accumulation of
The TNF-R superfamily is remarkably diverse with NIK, activation of IKKa, phosphorylation of p100 and liberation
of p100-inhibited NF-kB complexes. Simultaneous activation of
more than two-dozen receptors and nearly as many the canonical NF-kB pathway through either TRAF2, 5, or 6
ligands that are variably expressed throughout the also commonly occurs downstream of receptors that activate the
body. However, despite the physiological diversity of non-canonical pathway. See the text for additional details.

Oncogene
NF-jB and immunology
MS Hayden et al
6771
(Hsu et al., 1996); however, TRAF2 and TRAF5 appear LPS stimulation, in combination with p50 dimers has
to play redundant roles in TNF signaling to NF-kB. been shown to have a role in the inhibition of repeated
TRAF2 or TRAF5-deficient mice have intact TNF LPS responses in macrophages, a phenomenon also
activation of NF-kB, whereas TRAF2/5 double knock- referred to as LPS tolerance (Wessells et al., 2004).
out cells have substantially reduced TNF-induced IKK Furthermore by selectively affecting chromatin remo-
activation (Yeh et al., 1997; Nakano et al., 1999; Tada deling, BCL-3 mediates repression of proinflammatory
et al., 2001). TRAFs may either recruit the IKK genes, but also facilitates expression of the anti-
complex directly (Devin et al., 2001) or indirectly inflammatory gene IL-10. NF-kB p50 also appears to
through the serine/threonine kinase, RIP1. RIP1 can negatively regulate IFNg production and proliferation
also interact independently with TRADD and is an by NK cells (Tato et al., 2006).
essential adapter for TNF-induced NF-kB activation In addition to these and other negative feedback
and protection from apoptosis (Hsu et al., 1996; Ting pathways, it was recently found that inhibition of
et al., 1996; Kelliher et al., 1998; Devin et al., 2000). NF-kB during the resolution phase can prolong the
Upon ubiquitination, RIP1 can bind directly to NEMO inflammatory process and prevent proper tissue repair
and recruit IKK independent of TRAF2 (Zhang et al., (Lawrence et al., 2001). It was subsequently found that
2000). Signaling downstream of RIP1 requires TAK1 IKKa-deficient mice display increased inflammatory
for the activation of IKK (Sato et al., 2000; Shim et al., responses in models of local and systemic inflammation
2005). Whether TAK1 directly activates IKK or this (Lawrence et al., 2005). Macrophages, in particular,
process proceeds through an intermediary such as show increased production of proinflammatory chemo-
MEKK3 is not yet clear (Takaesu et al., 2003; Li kines and cytokines in the absence of IKKa (Lawrence
et al., 2006). However, it does not appear that TAK1 is et al., 2005; Li et al., 2005). It was suggested from these
involved in the activation of the non-canonical NF-kB studies that IKKa negatively regulates proinflammatory
pathway (Shim et al., 2005). gene expression, perhaps through mediating degrada-
The non-canonical NF-kB pathway is unique in that tion of RelA and c-Rel following macrophage activation
it is independent of IKKb and NEMO and instead by LPS.
requires IKKa which is phosphorylated by NF-kB
inducing kinase (NIK) (Xiao et al., 2001; Senftleben
et al., 2001a; Claudio et al., 2002; Dejardin et al., 2002;
see Scheidereit, 2006). The key question concerning Initiation of adaptive responses
signaling by these stimuli is how they are channeled to
NIK and IKKa, even though their receptor signaling Although innate responses alone can bring about potent
domains resemble those of other TNF family members antimicrobial activities, alerting and activating the
(Figure 6b). Because RANKL, BAFF and CD40L may adaptive immune system remains a crucial step for
also activate the canonical pathway through TRAF2/6, robust and durable immune responses. This process is
it would appear that the intracellular signaling domain largely mediated by activation and maturation of
of these receptors possess additional sequence motifs antigen-presenting cells (APCs), which can, in turn
that allow their signaling to NIK. This function is instruct T and B cells to carry out the adaptive response.
mediated by TRAF3, which interacts with these DC maturation mediated by pathogen recognition is
receptors. TRAF3 negatively regulates NIK, and under- crucial for the initiation of the adaptive immune
goes signal-dependent degradation resulting in the response. To activate naı̈ve T cells, DCs must undergo
activation of the non-canonical pathway (Liao et al., multiple changes. First, DCs must gain the ability to
2004). interact with T cells by changing their chemokine
receptor expression and migrating into lymphoid tissues.
Second, DCs must alter their antigen processing
Resolution of inflammation may also involve NF-kB machinery to favor the presentation of pathogen
Resolution of inflammation and subsequent tissue repair epitopes on MHC. Third, APCs must upregulate the
is a crucial event, and its failure is a common source of expression of costimulatory molecules B7.1/B7.2 (or
pathology. It is believed that reversal of inflammation is CD80/CD86), which are regulated by NF-kB and ligate
an active process that is as complex as the inflammatory CD28 providing the second signal necessary to induce
response itself, and involves numerous pathways that T-cell activation. Finally, as progress is made in
are not all directly relevant to NF-kB (Serhan and exploring these events it is becoming increasingly clear
Savill, 2005). Although the traditional view of NF-kB that the responses to different pathogens are tailored
would lead one to imagine that it would primarily based on the distribution of PRRs in different cell types
function by being turned off during the resolution phase and the ability of different cell types to, in turn, interact
of inflammation, recent work has suggested that NF-kB with T cells in a biasing manner (Iwasaki and
also has a more active role. Medzhitov, 2004).
During acute inflammation, there are multiple nega- Maturation of DCs following viral infection depends
tive feedback pathways that help to rein in inflammatory on nucleic acid-binding PRRs, including both TLRs and
responses. It has long been known that cells such as cytoplasmic RIG family molecules. Indeed, DC matura-
macrophages become resistant to repeated proinflam- tion during viral infection occurs normally in the
matory stimuli. BCL-3, which is induced late following absence of MyD88 or TLR3, as reported previously
Oncogene
NF-jB and immunology
MS Hayden et al
6772
(Lopez et al., 2003). Bacterial responses are either expression, human peripheral B cells express TLR1, 2, 4,
mediated through TLRs – DCs express TLRs 1, 2, 5 6 and 9 (Hornung et al., 2002).
and 6 – or other classes of PRRs. Murine CD8a þ DCs,
which tend to induce TH1 responses important in
clearance of viral and parasitic infections, express
TLR1, 2, 6, 9 and 11. In the absence of TLR11, for Role of NF-jB in the adaptive response
example, mice fail to mount a TH1 response against
T. gondii, owing to the failure of CD8a þ DCs to AgR signaling to NF-kB
recognize the TLR11 ligand (Yarovinsky et al., 2005). It The hallmark of the adaptive immune response is
remains unclear whether the ability of distinct TLR antigen specificity. In the section on hematopoiesis, we
ligands to induce TH1 versus TH2 responses is intrinsic discussed how NF-kB plays an important role in the
to the specific TLR/ligand, dose of ligand or the cell type selection of lymphocytes bearing somatically generating
within which this activation occurs; evidence to date AgRs. Signaling through these antigen-specific B-cell
points towards the latter. Even less clear are the roles of and T-cell receptors is therefore the central event of the
other PRRs in APC maturation and the initiation of adaptive immune response. Activation of NF-kB down-
adaptive responses. stream of BCR and TCR ligation facilitates antigen-
Finally, there is the question of the role of innate specific proliferation and maturation of lymphocytes
recognition of pathogens by lymphocytes themselves. into effector cells. Signaling through these two AgRs
Recently, it has been suggested that TLR signaling in B appears to be functionally analogous, although some of
cells is also required for optimal response to certain the components utilized differ.
antigens (Pasare and Medzhitov, 2005). Both B and BCR and TCR signaling are analogous in many
T cells express TLRs, although exactly which TLRs aspects, in particular with respect to the activation of
are expressed is debatable. At the level of mRNA NF-kB (Figure 7). The T-cell receptor complex consists

a b
Ag
Ag

TCR BCR

P
Syk Lyn
ZAP70 CARMA1 CARMA1
PDK1
P
BCL10 BCL10 P
Vav
Vav PI3K MALT1 MALT1

P TRAF6 Ras
Ras RIP2
DAG IP3
IP3
DAG
Ub AP-1
AP-1 NEMO
P
NFAT
NFAT

PP
NF- κB

Gene
Transcription

Figure 7 AgR signaling to NF-kB. Binding of TCR to peptide:MHC and co-stimulation through CD28:B7 interaction activates
NF-kB (a). Through multiple steps, initial phosphorylation events at the TCR complex lead to PI3K activation and recruitment of
PDK1. PDK1, in turn, mediates recruitment of the IKK complex through PKCy and the CARMA1 directly. PKCy phosphorylates
CARMA1 leading to the activation of IKK through BCL10, MALT1 and TRAF6. Binding to TI antigens or TD antigens in the
presence of co-stimulatory cytokines results in the activation of NF-kB through the BCR complex (b). Receptor proximal events of
BCR activation are highly analogous to those of the TCR. Although the role of PDK1 in B-cell signaling is not established, BCR
signaling to NF-kB requires PKCb as well as the CBM complex to achieve activation of IKK.

Oncogene
NF-jB and immunology
MS Hayden et al
6773
of a/b subunits that are associated with the CD3 protein et al., 2003; Hara et al., 2003). Similarly, BCL10 is
heterodimers g/e, d/e, and either Z/Z, Z/z or z/z, for a critical for NF-kB activation via the BCR and TCR, yet
total of eight membrane proteins. The BCR is, likewise, normal numbers of peripheral T cells are seen in BCL10
a multiprotein complex consisting of the surface knockouts, and no clear defects in B-cell development is
immunoglobulin receptor associated with a heterodimer observed (Ruland et al., 2001). BCL10 interacts with
of Iga and Igb. The AgR complexes associate with Src CARMA1 leading to BCL10 phosphorylation, although
family tyrosine kinases (SFKs), Lck and Fyn in T cells CARMA1 lacks kinase activity (Bertin et al., 2001;
and Lyn in B cells, which phosphorylate immunoreceptor Gaide et al., 2001).
tyrosine activation motifs (ITAMs) on CD3 and Iga/b Interestingly, genetic evidence of a role for RIP2 has
chains. The cytoplasmic tyrosine kinases ZAP70 or Syk recently been reported in T-cell signaling (Ruefli-Brasse
are then recruited via SH2 domains to the phosphory- et al., 2004). RIP2 associates with BCL10 and is
lated ITAMs and initiate activation of the IP3 and Ras necessary for TCR-induced BCL10 phosphorylation
family pathways. The IKK complex is rapidly recruited and IKK activation. It is not yet clear how TCR
to the immunological synapse and can be colocalized to signaling regulates RIP2 and, in turn, how RIP2 might
the TCR (Khoshnan et al., 2000; Weil et al., 2003). In affect IKK activity. BCL10 oligomerization has been
ZAP-70-deficient T cells, NF-kB activation can be implicated in IKK activation through a process that
rescued by directly targeting a chimeric NEMO to the involves ubiquitination of NEMO (Zhou et al., 2004).
immunological synapse, suggesting that signaling down- This ubiquitination event appears to be mediated by
stream of ZAP-70 may largely function for IKK MALT1, and perhaps TRAF6, although no TCR
recruitment (Weil et al., 2003). signaling deficits have been reported in TRAF6-deficient
The signaling pathway from the receptor to NF-kB mice (Lomaga et al., 1999; Sun et al., 2004). If this is
requires PKCy (PKCb in B cells), CARMA1/CARD11, true however, it is possible that the effect is mediated
BCL10 and MALT1 (Sun et al., 2000; Ruland et al., through the kinase TAK1, which functions in IKK
2001, 2003; Saijo et al., 2002; Hara et al., 2003; Ruefli- activation downstream of TRAF6 in other pathways.
Brasse et al., 2003;). Although there has been some However, B cells in which TAK1 has been conditionally
controversy, available data suggest that PKCy is largely inactivated have normal BCR signaling to NF-kB. More
essential for activation of NF-kB via T-cell stimulation work must be carried out with TAK1 conditional
(Sun et al., 2000; Pfeifhofer et al., 2003) and can mediate knockouts in both the BCR and TCR pathways to
the activation of IKK (Lin et al., 2000). PKCy is address the role of this kinase.
specifically recruited to the immunological synapse;
although how PKCy but not other PKC isoforms is
selectively recruited remains a mystery. In B cells PKCb T-cell responses mediated by NF-kB
is, likewise, required for recruitment of the IKK To become activated, naı̈ve T cells must receive two
complex to lipid rafts following BCR ligation (Su distinct signals: antigen-specific and co-stimulatory.
et al., 2002). PKCy is capable of directly interacting Antigen-specific activation signals emanate from the
with the IKK complex in primary T cells (Khoshnan binding of the TCR to cognate antigenic peptides
et al., 2000) and might, therefore, function by bringing presented in the binding cleft of MHC. Co-stimulatory
IKK to the receptor complex and into proximity with signaling is provided through ligation of CD28 by B7
other essential components in this pathway; namely molecules expressed on activated APCs. Stimulation of
CARMA1, BCL10 and MALT1 (collectively known as naı̈ve T cells results in the production of IL-2, which is
the CBM complex). Recently, studies have provided necessary for their proliferation and survival. These
evidence that the protein kinase PDK1 recruits PKCy activated naı̈ve T cell blasts proliferate rapidly and
and the IKK complex to lipid rafts. In addition, PDK1 simultaneously undergo differentiation into effector
can simultaneously recruit the CBM complex through cells. In the case of TH cells, proliferation leads to
binding to CARMA1 (Lee et al., 2005). The induced differentiation into immature effector cells, TH0, which
proximity of PKC and the CBM may allow phospho- subsequently differentiate into TH1 or TH2 cells
rylation of CARMA1 by PKCy/b (Matsumoto et al., depending on the predominant cytokine milieu. CD8 T
2005; Sommer et al., 2005). Interestingly, T-cell-specific cells are likewise activated by professional APCs,
PDK1 conditional deletion results in a defect in T-cell although they may receive secondary signals from
development, preventing the production of peripheral activated TH1 cells. The unavailability of CD8 condi-
T cells (Hinton et al., 2004). On the other hand, tional knockouts, and the selective loss of CD8 cells in
PCKy knockouts do not display defects in thymocyte the absence of NF-kB activity has prevented a thorough
development. characterization of the role of NF-kB in these cells.
The CBM complex is essential for both AgR signaling Rapidly proliferating activated T cells rely on NF-kB
in mature B and T cells. What is surprising however, as activity for protection from apoptosis as well as for the
mentioned above, is the lack of a role for this complex in production of cytokines supporting proliferation and
developing lymphocytes, and by extrapolation signaling differentiation. As expected, inhibition of NF-kB in
through the pre-AgRs. The MAGUK family protein activated T cells facilitates progression towards AICD
CARMA1 is required for activation of NF-kB in T cells or apoptosis (Ivanov and Nikolic-Zugic, 1997; Jeremias
following TCR ligation, but its loss has no effect on the et al., 1998). Indeed, stimulation of RelA-deficient naı̈ve
development of thymocyte (Gaide et al., 2002; Egawa T cells induces cell death (Wan and DeGregori, 2003).
Oncogene
NF-jB and immunology
MS Hayden et al
6774
Peripheral T cells lacking c-Rel do not undergo gene is repressed. Direct binding of T-bet to p65 that is
apoptosis, but nevertheless, fail to proliferate in associated with the IL-2 gene enhancer may mediate the
response to typical mitogenic stimuli (Köntgen et al., repression of IL-2 production in TH1 cells (Hwang et al.,
1995), and both RelA and c-Rel containing complexes 2005). Alternatively, in TH2 cells the lack of IL-2
accumulate in the nucleus following TCR/CD28 stimu- transcription may be due to the decreased levels of RelA
lation (Ghosh et al., 1993). Interestingly, c-Rel-deficient activation in TH2 cells (Lederer et al., 1994).
T cells appear to have a defect in TH1 proliferation
and production of IFNg, indicating a selective role for
NF-kB family members in TH1/TH2 differentiation, B-cell responses mediated by NF-kB
independent of that mediated by the innate response. B-cell responses can be classified into two groups:
Multiple transcriptional activators and repressors thymus-dependent (TD) or thymus-independent (TI).
regulate expression of IL-2. Among these, members of In response to T-dependent antigens, B cells require co-
the NF-kB family play multiple roles. In naı̈ve T cells, stimulatory signaling from TH cells expressing CD40L
which do not express IL-2, repressive p50 homo- and cytokines, such as IL-4. B cells from individuals
dimers are found associated with the IL-2 promoter with a mutation in CD40L are unable to undergo class
(Grundstrom et al., 2004). Failure of T-cell proliferative switch recombination in response to T-dependent
responses in c-Rel knockout mice is attributable to a antigens (Aruffo et al., 1993). Signaling through CD40
failure to produce IL-2 (Köntgen et al., 1995). In naı̈ve activates both canonical and non-canonical NF-kB
T cells, c-Rel is responsible for mediating chromatin pathways, although it is unclear which is operative in
remodeling across the IL-2 locus following CD3/CD28 the response to T-dependent antigens. For example,
co-stimulation (Rao et al., 2003). Naı̈ve T cells can be whereas B cells from p52/ mice mount inadequate
primed by exposure to inflammatory cytokines such that humoral responses to various T-dependent antigens,
they generate a more robust response to CD3/CD28 co- they exhibit a normal response following adoptive
stimulation. Overexpression of an IkBa super-repressor transfer into rag-1/ mice – indicating that this deficit
suggested that NF-kB is required for this priming event is not intrinsic to B cells (Franzoso et al., 1998).
in T cells (Mora et al., 2001a). More recent data indicate Furthermore, B cells from relB/ mice, although
that c-Rel is necessary for naı̈ve helper T-cell priming by crippled in their proliferative response, undergo normal
pro-inflammatory cytokines elicited following stimula- IgM secretion and class switching in response to various
tion with TLR ligands (Banerjee et al., 2005). NF-kB stimuli (Snapper et al., 1996a). Therefore, non-canonical
RelA-containing complexes, on the other hand, appear NF-kB pathway activation downstream of CD40 is
to function more traditionally in mediating transactiva- probably not required for class switching during
tion of IL-2 gene expression, and overexpression of T-dependent antigen responses.
RelA with c-Jun can overcome the requirement for Analysis of the canonical NF-kB pathway is compli-
co-stimulation in naı̈ve T cells (Parra et al., 1998). cated by more generalized defects in lymphocyte
However, as discussed below, these complexes may also response owing to the requirement for this pathway in
be the targets of negative regulation following T-cell AgR signaling. Nevertheless, whether downstream of
differentiation. CD40 or other stimuli, evidence supports canonical
Recent work in TH1/TH2 differentiation has focused pathway activation in the process of class switch
on the induction of specific transcription factors in these recombination. Following adoptive transfer, B cells
two effector cell types – T-bet and GATA3, respectively. from rela/ mice exhibit markedly diminished class
Interestingly, mice lacking p50 are unable to mount an switching, despite a modest loss of lymphocyte proli-
asthma-like airway TH2 response, and do not induce feration following various stimuli (Doi et al., 1997).
GATA-3 expression during T-cell stimulation under Likewise, c-Rel-deficient mice, or mice lacking the c-Rel
TH2 differentiating conditions (Das et al., 2001). C-terminal transactivation domain, fail to generate a
Consistent with this finding, BCL-3-deficient T cells productive humoral immune response suggesting a
also fail to undergo TH2 differentiation. Furthermore, requirement for c-Rel in class switch recombination
BCL-3 can induce expression of a reporter gene from a (Köntgen et al., 1995; Zelazowski et al., 1997; Carrasco
gata-3 promoter, suggesting that p50/BCL-3 complexes et al., 1998). B cells from nfkb1/ mice exhibit decreased
are crucial for TH2 differentiation (Corn et al., 2005). proliferation in response to mitogenic stimulation, and
Conversely, the same authors found that RelB-deficient p50/RelA double knockout B cells exhibit greater
T cells are deficient in TH1 differentiation and IFNg defects in proliferation and class switching (Snapper
production, and show decreased expression of T-bet; et al., 1996b; Horwitz et al., 1999). Therefore, analyses
likely through a failure to upregulate STAT4, which of knockout animals suggest that the canonical NF-kB
functions in signaling from IFN to T-bet induction. pathway likely has a role in maturation of the B-cell
Therefore, it appears that NF-kB activation during response in addition to directly mediating proliferative
TCR stimulation may render cells competent for both responses following BCR ligation.
proliferative and differentiating stimuli. As discussed above, signaling through TLRs has an
As TH cells differentiate into TH1 or TH2, they important role in the initiation of the adaptive immune
decrease their expression of IL-2 and, instead, become response via APCs of the innate immune system. In
dependent on TH1 and TH2 cytokines (e.g., IFNg and recent years, however, there has also been increasing
IL-4). As a corollary, NF-kB transactivation of the IL-2 interest in the ability of TLR signaling to directly
Oncogene
NF-jB and immunology
MS Hayden et al
6775
modulate the adaptive response. For example, it has In large part, these defects appear to be due to a loss
been observed that homeostatic polyclonal activation of of BCR signaling, as demonstrated in an elegant study
B cells, which results in the so-called serological that demonstrated that deletion of the BCR from
memory, that is, detectable antibody to antigens that mature B cells led to a complete loss of the peripheral
are no longer present in the host, can be induced/ B-cell pool (Kraus et al., 2004). Most likely this was due
maintained by TLR ligation (Bernasconi et al., 2002). to the loss of signaling to NF-kB in these cells because
Analogously, TLR2 is upregulated in CD4 þ T cells loss of IKKb, NEMO or components of the CBM
following TCR stimulation, and TLR2 ligands complex in mature B cells also results in a complete loss
may thus provide an activation/maintenance signal in of peripheral B cells (Pasparakis et al., 2002; Li et al.,
these cells (Komai-Koma et al., 2004). That signaling 2003; Thome, 2004).
through TLRs in these aspects of B-cell responses The non-canonical NF-kB pathway is also relevant to
requires NF-kB seems likely, but has yet to be B-cell survival, as the loss of IKKa results in striking
demonstrated. defects in B-cell survival (Kaisho et al., 2001; Senftleben
TI antigens have an intrinsic ability to activate B-cell et al., 2001a). However, rather than acting downstream
responses in the absence of T-cell help by acting as B-cell of tonic BCR signaling, recent studies have implicated
mitogens, for example by acting as TLR ligands or by signaling from BAFFR in this aspect of the B
binding with high avidity to the BCR through repetitive lymphocyte survival (reviewed in Mackay et al., 2003).
structural features. In such cases, it is expected that B-cell Together, these data suggest that a subset of Bcl-2
responses are more dependent on members of the family members, for example, the antiapoptotic factor
canonical pathway that have well-documented roles in A1, are regulated by p52/RelB-containing complexes
TLR signaling, or BCR signaling (as discussed above). and are necessary for the maintenance of mature B cells.
For example, c-Rel-deficient B cells are highly sensitive
to apoptosis following BCR cross-linking (Grumont
et al., 1998, 1999; Owyang et al., 2001). As mentioned
above, p50 and p50/pRelA double knockout B cells are Concluding remarks
deficient in responses to TI stimulation. Likewise,
IKKb-deficient B cells fail to mount TI or TD responses NF-kB was originally described as a transcriptional
(Li et al., 2003). These IKKb-deficient B cells also regulator in the adaptive immune response; however,
exhibit increased spontaneous apoptosis, suggesting that subsequent studies have revealed its importance in
NF-kB is important in survival of B cells. hematopoiesis, lymphoid organogenesis and innate
immunity. Investigations of mice with targeted deletions
and mutations have shed considerable light on the role
Maintenance and memory: a role for NF-kB in lymphoid of NF-kB in lymphoid organogenesis. For example,
cell survival characterization of the aly/aly mouse in part led to the
Lymphocyte homeostasis is dependent on the survival of discovery of the non-canonical NF-kB pathway and its
mature lymphocytes in addition to replenishment of the role in organogenesis. Progress continues to be made in
peripheral lymphocyte pool through lymphopoiesis. understanding AgR signaling to NF-kB, and this work
Consequently, there is increasing interest in the possible has led to the appreciation of regulatory ubiquitination
role of NF-kB in the survival of mature lymphocytes. It events in IKK activation (see reviews by Perkins, 2006;
is widely accepted that lymphocyte survival is mediated Scheidereit, 2006). The specificity of the requirement for
through tonic stimulation downstream of the AgR, as the CBM complex and PKCy/b in signaling by T-cell
well as certain cytokine receptors. As discussed above, and B-cell receptors opens the door to the development
genetic targeting experiments support an important role of equally specific NF-kB inhibitors. However, there
for NF-kB family members in lymphocyte survival. remain fundamental gaps in our understanding of how
Naı̈ve T cells require continued contact with MHC:self- these components mediate IKK activation and the
peptides, most likely expressed on lymphoid DCs, to possible role of regulatory ubiquitination in this process.
generate the tonic TCR signal that is essential for The role of NF-kB in hematopoiesis remains partially
continued survival. Survival of memory cells, on the defined, although there is little doubt about its
other hand, is independent of continued contact with importance. Genetic targeting in mice has allowed the
self-peptide:MHC complexes. enumeration of multiple steps in hematopoiesis at which
B cells are formed at a far higher rate than T cells and various NF-kB pathway components are required;
therefore B cells also undergo a significantly higher rate however, a cohesive picture of how NF-kB functions
of turnover. Nonetheless, B cells too require mainte- in these steps remains elusive. For example, whereas we
nance signals to achieve peripheral homeostasis. The know that canonical NF-kB activity is required in early
AgR on B cells most likely provides a basal level of lymphopoiesis, it is unclear whether this is in the
signaling, albeit independent of the presence of antigen, regulation of TNFa production or in mediating a
which is required for maintenance of mature B cells. Not survival signal in the developing lymphocyte. NF-kB is
surprisingly, B cells from RelA/, p100/, p105/ and required in the process of positive and negative
c-Rel/ mice display increased sensitivity to apoptosis selection, but again, its mechanism(s) of action remains
and/or decreased survival ex vivo (Grumont et al., 1998; poorly defined. Progress using in vitro systems for
Claudio et al., 2002; Prendes et al., 2003). studying lymphocyte development may allow a more
Oncogene
NF-jB and immunology
MS Hayden et al
6776
rigorous assessment of NF-kB function in these about the role of NF-kB in memory cells, although
processes. Likewise, advances in conditional gene recent advances in identifying and characterizing
targeting approaches and the generation of animals in memory precursors bodes well for future progress in
which defective versions of NF-kB genes have been this area.
knocked-in may help to overcome the problems of In summary, research to date has highlighted the
embryonic lethality and functional redundancy that importance of NF-kB in regulating genes that prevent
have sometimes made existing studies difficult to apoptosis and that promote differentiation and deve-
interpret. lopment in cells of the innate and adaptive immune
T- and B-cell responses require NF-kB as a prosurvi- systems. A large body of work has elucidated many of
val factor as well as for the regulation of genes involved the molecular mechanisms governing regulation of
in differentiation to effector cells. More recently, a NF-kB by engagement of innate or lymphocyte anti-
limited number of studies have suggested a role for gen-specific receptors. In turn, these data provide a
PRRs in lymphocyte activation, and the role of NF-kB trove of information that will likely prove useful in
in this process remains to be elucidated. The role of attempts to manipulate the immune system to prevent
NF-kB in the differentiation of TH cells is incompletely and treat disease.
understood, and requires further clarification. We also
know very little about the role of NF-kB in CD8 þ
Acknowledgements
activation, differentiation and function; progress in this
area awaits development of better tools for genetically Research in the authors’ laboratory was supported by
manipulating this cell population. Upon successful grants from the National Institutes of Health (to SG). MSH
clearance of pathogen, regulation of NF-kB allows was supported by NIH/National Institute of General
resolution of the response and facilitates the develop- Medical Sciences Medical Scientist Training Grant
ment of memory cells. To date, relatively little is known GM07205.

References

Abe K, Yarovinsky FO, Murakami T, Shakhov AN, Campos MA, Almeida IC, Takeuchi O, Akira S, Valente EP,
Tumanov AV, Ito D et al. (2003). Blood 101: 1477–1483. Procopio DO et al. (2001). J Immunol 167: 416–423.
Akira S, Uematsu S, Takeuchi O. (2006). Cell 124: 783–801. Carrasco D, Cheng J, Lewin A, Warr G, Yang H, Rizzo C
Alcamo E, Hacohen N, Schulte LC, Rennert PD, Hynes RO, et al. (1998). J Exp Med 187: 973–984.
Baltimore D. (2002). J Exp Med 195: 233–244. Catley MC, Chivers JE, Cambridge LM, Holden N,
Alcamo E, Mizgerd JP, Horwitz BH, Bronson R, Beg AA, Slater DM, Staples KJ et al. (2003). FEBS Lett 547:
Scott M et al. (2001). J Immunol 167: 1592–1600. 75–79.
Alexopoulou L, Holt AC, Medzhitov R, Flavell RA. (2001). Claudio E, Brown K, Park S, Wang H, Siebenlist U. (2002).
Nature 413: 732–738. Nat Immunol 3: 958–965.
Andrejeva J, Childs KS, Young DF, Carlos TS, Stock N, Coban C, Ishii KJ, Kawai T, Hemmi H, Sato S, Uematsu S
Goodbourn S et al. (2004). Proc Natl Acad Sci USA 101: et al. (2005). J Exp Med 201: 19–25.
17264–17269. Corn RA, Hunter C, Liou H-C, Siebenlist U, Boothby MR.
Aruffo A, Farrington M, Hollenbaugh D, Li X, Milatovich A, (2005). J Immunol 175: 2102–2110.
Nonoyama S et al. (1993). Cell 72: 291–300. Courtois G, Gilmore TD. (2006). Oncogene 25: 6831–6843.
Ayabe T, Satchell DP, Wilson CL, Parks WC, Selsted ME, Covert MW, Leung TH, Gaston JE, Baltimore D. (2005).
Ouellette AJ. (2000). Nat Immunol 1: 113–118. Science 309: 1854–1857.
Balachandran S, Thomas E, Barber GN. (2004). Nature 432: Das J, Chen CH, Yang L, Cohn L, Ray P, Ray A. (2001). Nat
401–405. Immunol 2: 45–50.
Banerjee D, Liou H-C, Sen R. (2005). Immunity 23: 445–458. Dejardin E, Droin NM, Delhase M, Haas E, Cao Y, Makris C
Barton GM, Kagan JC, Medzhitov R. (2006). Nat Immunol 7: et al. (2002). Immunity 17: 525–535.
49–56. Devin A, Cook A, Lin Y, Rodriguez Y, Kelliher M, Liu Z.
Batten M, Groom J, Cachero TG, Qian F, Schneider P, (2000). Immunity 12: 419–429.
Tschopp J et al. (2000). J Exp Med 192: 1453–1466. Devin A, Lin Y, Yamaoka S, Li Z, Karin M, Liu Z. (2001).
Beg AA, Sha WC, Bronson RT, Baltimore D. (1995). Genes Mol Cell Biol 21: 3986–3994.
Dev 9: 2736–2746. Didierlaurent A, Brissoni B, Velin D, Aebi N, Tardivel A,
Bernasconi NL, Traggiai E, Lanzavecchia A. (2002). Science Kaslin E et al. (2006). Mol Cell Biol 26: 735–742.
298: 2199–2202. Diebold SS, Kaisho T, Hemmi H, Akira S, Reis e Sousa C.
Bertin J, Wang L, Guo Y, Jacobson MD, Poyet JL, (2004). Science 303: 1529–1531.
Srinivasula SM et al. (2001). J Biol Chem 276: 11877–11882. Doi TS, Takahashi T, Taguchi O, Azuma T, Obata Y. (1997).
Bonizzi G, Karin M. (2004). Trends Immunol 25: 280–288. J Exp Med 185: 953–961.
Burkly L, Hession C, Ogata L, Reilly C, Marconi LA, Olson D Dougall WC, Glaccum M, Charrier K, Rohrbach K, Brasel K,
et al. (1995). Nature 373: 531–536. De Smedt T et al. (1999). Genes Dev 13: 2412–2424.
Burns K, Clatworthy J, Martin L, Martinon F, Plumpton C, Dutta j, Fan Y, Gupta N, Fan G, Gélinas C. (2006). Oncogene
Maschera B et al. (2000). Nat Cell Biol 2: 346–351. 25: 6800–6816.
Caamano JH, Perez P, Lira SA, Bravo R. (1996). Mol Cell Biol Eck SL, Perkins ND, Carr DP, Nabel GJ. (1993). Mol Cell
16: 1342–1348. Biol 13: 6530–6536.
Caamano JH, Rizzo CA, Durham SK, Barton DS, Raventos- Egawa T, Albrecht B, Favier B, Sunshine MJ, Mirchandani K,
Suarez C, Snapper CM et al. (1998). J Exp Med 187: 185–196. O’Brien W et al. (2003). Curr Biol 13: 1252–1258.
Oncogene
NF-jB and immunology
MS Hayden et al
6777
Feng B, Cheng S, Hsia CY, King LB, Monroe JG, Liou H-C. Horwitz BH, Zelazowski P, Shen Y, Wolcott KM, Scott ML,
(2004). Cell Immunol 232: 9–20. Baltimore D et al. (1999). J Immunol 162: 1941–1946.
Fitzgerald KA, Palsson-McDermott EM, Bowie AG, Jefferies Hsu H, Shu HB, Pan MG, Goeddel DV. (1996). Cell 84:
CA, Mansell AS, Brady G et al. (2001). Nature 413: 78–83. 299–308.
Fitzgerald KA, Rowe DC, Barnes BJ, Caffrey DR, Visintin A, Huang Q, Yang J, Lin Y, Walker C, Cheng J, Liu ZG et al.
Latz E et al. (2003). J Exp Med 198: 1043–1055. (2004). Nat Immunol 5: 98–103.
Franchi L, Amer A, Body-Malapel M, Kanneganti TD, Hueber AO. (2003). Cell Death Differ 10: 7–9.
Ozoren N, Jagirdar R et al. (2006). Nat Immunol 7: 576–582. Hwang ES, Hong JH, Glimcher LH. (2005). J Exp Med 202:
Francois S, El Benna J, Dang PM, Pedruzzi E, Gougerot- 1289–1300.
Pocidalo MA, Elbim C. (2005). J Immunol 174: 3633–3642. Inohara N, Nunez G. (2003). Nat Rev Immunol 3: 371–382.
Franzoso G, Carlson L, Poljak L, Shores EW, Epstein S, Inohara N, Koseki T, Lin J, del Peso L, Lucas PC, Chen FF
Leonardi A et al. (1998). J Exp Med 187: 147–159. et al. (2000). J Biol Chem 275: 27823–27831.
Franzoso G, Carlson L, Xing L, Poljak L, Shores EW, Ishii KJ, Coban C, Kato H, Takahashi K, Torii Y, Takeshita F
Brown KD et al. (1997). Genes Dev 11: 3482–3496. et al. (2006). Nat Immunol 7: 40–48.
French LE, Hahne M, Viard I, Radlgruber G, Zanone R, Ivanov VN, Nikolic-Zugic J. (1997). J Biol Chem 272:
Becker K et al. (1996). J Cell Biol 133: 335–343. 8558–8566.
Gaide O, Favier B, Legler DF, Bonnet D, Brissoni B, Iwasaki A, Medzhitov R. (2004). Nat Immunol 5: 987–995.
Valitutti S et al. (2002). Nat Immunol 3: 836–843. Janeway Jr CA. (1989). Cold Spring Harbor Symp Quant Biol
Gaide O, Martinon F, Micheau O, Bonnet D, Thome M, 54(Part 1): 1–13.
Tschopp J. (2001). FEBS Lett 496: 121–127. Jeremias I, Kupatt C, Baumann B, Herr I, Wirth T,
Gazzinelli RT, Ropert C, Campos MA. (2004). Immunol Rev Debatin KM. (1998). Blood 91: 4624–4631.
201: 9–25. Jimi E, Ghosh S. (2005). Immunol Rev 208: 80–87.
Ghosh P, Tan TH, Rice NR, Sica A, Young HA. (1993). Proc Jimi E, Phillips RJ, Rincon M, Voll R, Karasuyama H,
Natl Acad Sci USA 90: 1696–1700. Flavell R et al. (2005). Int Immunol 17: 815–825.
Gilmore TD. (2006). Oncogene 25: 6680–6684. Jurk M, Heil F, Vollmer J, Schetter C, Krieg AM, Wagner H
Goudeau B, Huetz F, Samson S, Di Santo JP, Cumano A, et al. (2002). Nat Immunol 3: 499.
Beg A et al. (2003). Proc Natl Acad Sci USA 100: 15800–15805. Kagan JC, Medzhitov R. (2006). Cell 125: 943–955.
Gross JA, Dillon SR, Mudri S, Johnston J, Littau A, Roque R Kaisho T, Takeda K, Tsujimura T, Kawai T, Nomura F,
et al. (2001). Immunity 15: 289–302. Terada N et al. (2001). J Exp Med 193: 417–426.
Grossmann M, Metcalf D, Merryfull J, Beg A, Baltimore D, Kajiura F, Sun S, Nomura T, Izumi K, Ueno T, Bando Y et al.
Gerondakis S. (1999). Proc Natl Acad Sci USA 96: (2004). J Immunol 172: 2067–2075.
11848–11853. Kang DC, Gopalkrishnan RV, Wu Q, Jankowsky E,
Grossmann M, O’Reilly LA, Gugasyan R, Strasser A, Pyle AM, Fisher PB. (2002). Proc Natl Acad Sci USA 99:
Adams JM, Gerondakis S. (2000). EMBO J 19: 6351–6360. 637–642.
Grumont RJ, Rourke IJ, Gerondakis S. (1999). Genes Dev 13: Kariko K, Buckstein M, Ni H, Weissman D. (2005). Immunity
400–411. 23: 165–175.
Grumont RJ, Rourke IJ, O’Reilly LA, Strasser A, Miyake K, Kato H, Sato S, Yoneyama M, Yamamoto M, Uematsu S,
Sha W et al. (1998). J Exp Med 187: 663–674. Matsui K et al. (2005). Immunity 23: 19–28.
Grundstrom S, Anderson P, Scheipers P, Sundstedt A. (2004). Kato H, Takeuchi O, Sato S, Yoneyama M, Yamamoto M,
J Biol Chem 279: 8460–8468. Matsui K et al. (2006). Nature 441: 101–105.
Hara H, Wada T, Bakal C, Kozieradzki I, Suzuki S, Suzuki N Kawai T, Adachi O, Ogawa T, Takeda K, Akira S. (1999).
et al. (2003). Immunity 18: 763–775. Immunity 11: 115–122.
Hayashi F, Smith KD, Ozinsky A, Hawn TR, Yi EC, Kawai T, Takahashi K, Sato S, Coban C, Kumar H, Kato H
Goodlett DR et al. (2001). Nature 410: 1099–1103. et al. (2005). Nat Immunol 6: 981–988.
Hayden MS, Ghosh S. (2004). Genes Dev 18: 2195–2224. Kelliher MA, Grimm S, Ishida Y, Kuo F, Stanger BZ,
Heil F, Hemmi H, Hochrein H, Ampenberger F, Kirschning C, Leder P. (1998). Immunity 8: 297–303.
Akira S et al. (2004). Science 303: 1526–1529. Khoshnan A, Bae D, Tindell CA, Nel AE. (2000). J Immunol
Hemmi H, Kaisho T, Takeuchi O, Sato S, Sanjo H, Hoshino K 165: 6933–6940.
et al. (2002). Nat Immunol 3: 196–200. Kinjyo I, Hanada T, Inagaki-Ohara K, Mori H, Aki D,
Hemmi H, Takeuchi O, Kawai T, Kaisho T, Sato S, Sanjo H Ohishi M et al. (2002). Immunity 17: 583–591.
et al. (2000). Nature 408: 740–745. Kinoshita D, Hirota F, Kaisho T, Kasai M, Izumi K, Bando Y
Hemmi H, Takeuchi O, Sato S, Yamamoto M, Kaisho T, et al. (2006). J Immunol 176: 3995–4002.
Sanjo H et al. (2004). J Exp Med 199: 1641–1650. Kishimoto H, Surh CD, Sprent J. (1998). J Exp Med 187:
Hettmann T, DiDonato J, Karin M, Leiden JM. (1999). J Exp 1427–1438.
Med 189: 145–158. Kobayashi K, Hernandez LD, Galan JE, Janeway Jr CA,
Hinton HJ, Alessi DR, Cantrell DA. (2004). Nat Immunol 5: Medzhitov R, Flavell RA. (2002). Cell 110: 191–202.
539–545. Koike R, Nishimura T, Yasumizu R, Tanaka H, Hataba Y,
Honda K, Yanai H, Mizutani T, Negishi N, Shimada N, Watanabe T et al. (1996). Eur J Immunol 26: 669–675.
Susuki N et al. (2003). Proc Natl Acad Sci USA 101 : Komai-Koma M, Jones L, Ogg GS, Xu D, Liew FY. (2004).
15416–15421. Proc Natl Acad Sci USA 101: 3029–3034.
Horng T, Barton GM, Flavell RA, Medzhitov R. (2002). Köntgen F, Grumont RJ, Strasser A, Metcalf D, Li R,
Nature 420: 329–333. Tarlinton D et al. (1995). Genes Dev 9: 1965–1977.
Hornung V, Rothenfusser S, Britsch S, Krug A, Jahrsdorfer B, Kopp E, Medzhitov R, Carothers J, Xiao C, Douglas I,
Giese T et al. (2002). J Immunol 168: 4531–4537. Janeway CA et al. (1999). Genes Dev 13: 2059–2071.
Horwitz BH, Scott ML, Cherry SR, Bronson RT, Baltimore D. Kraus M, Alimzhanov MB, Rajewsky N, Rajewsky K. (2004).
(1997). Immunity 6: 765–772. Cell 117: 787–800.
Oncogene
NF-jB and immunology
MS Hayden et al
6778
Kriehuber E, Bauer W, Charbonnier AS, Winter D, Meylan E, Burns K, Hofmann K, Blancheteau V, Martinon F,
Amatschek S, Tamandl D et al. (2005). Blood 106: 175–183. Kelliher M et al. (2004). Nat Immunol 5: 503–507.
Krug A, French AR, Barchet W, Fischer JA, Dzionek A, Meylan E, Curran J, Hofmann K, Moradpour D, Binder M,
Pingel JT et al. (2004). Immunity 21: 107–119. Bartenschlager R et al. (2005). Nature 437: 1167–1172.
Kurt-Jones EA, Popova L, Kwinn L, Haynes LM, Jones LP, Miao EA, Alpuche-Aranda CM, Dors M, Clark AE,
Tripp RA et al. (2000). Nat Immunol 1: 398–401. Bader MW, Miller SI et al. (2006). Nat Immunol 7: 569–575.
Lai WC, Zhou M, Shankavaram U, Peng G, Wahl LM. Miller LS, Sorensen OE, Liu PT, Jalian HR, Eshtiaghpour D,
(2003). J Immunol 170: 6244–6249. Behmanesh BE et al. (2005). J Immunol 174: 6137–6143.
Lawrence T, Bebien M, Liu GY, Nizet V, Karin M. (2005). Minakhina S, Steward R. (2006). Oncogene 25: 6749–6757.
Nature 434: 1138–1143. Miyawaki S, Nakamura Y, Suzuka H, Koba M, Yasumizu R,
Lawrence T, Gilroy DW, Colville-Nash PR, Willoughby DA. Ikehara S et al. (1994). Eur J Immunol 24: 429–434.
(2001). Nat Med 7: 1291–1297. Mizel SB, West AP, Hantgan RR. (2003). J Biol Chem 278:
Lederer JA, Liou JS, Todd MD, Glimcher LH, Lichtman AH. 23624–23629.
(1994). J Immunol 152: 77–86. Mora A, Youn J, Keegan A, Boothby M. (2001a). J Immunol
Lee KY, D’Acquisto F, Hayden MS, Shim JH, Ghosh S. 166: 2218–2227.
(2005). Science 308: 114–118. Mora AL, Stanley S, Armistead W, Chan AC, Boothby M.
Legler DF, Micheau O, Doucey MA, Tschopp J, Bron C. (2001b). J Immunol 167: 5628–5635.
(2003). Immunity 18: 655–664. Motoyama N, Wang F, Roth KA, Sawa H, Nakayama K,
Levitz SM. (2004). Microbes Infect 6: 1351–1355. Negishi I et al. (1995). Science 267: 1506–1510.
Li H, Kobayashi M, Blonska M, You Y, Lin X. (2006). J Biol Mueller T, Terada T, Rosenberg IM, Shibolet O, Podolsky
Chem 281: 13636–13643. DK. (2006). J Immunol 176: 5805–5814.
Li Q, Lu Q, Bottero V, Estepa G, Morrison L, Mercurio F Nakagawa R, Naka T, Tsutsui H, Fujimoto M, Kimura A,
et al. (2005). Proc Natl Acad Sci USA 102: 12425–12430. Abe T et al. (2002). Immunity 17: 677–687.
Li Q, Van Antwerp D, Mercurio F, Lee KF, Verma IM. Nakano H, Sakon S, Koseki H, Takemori T, Tada K,
(1999a). Science 284: 321–325. Matsumoto M et al. (1999). Proc Natl Acad Sci USA 96:
Li ZW, Chu W, Hu Y, Delhase M, Deerinck T, Ellisman M 9803–9808.
et al. (1999b). J Exp Med 189: 1839–1845. Oshiumi H, Matsumoto M, Funami K, Akazawa T, Seya T.
Li ZW, Omori SA, Labuda T, Karin M, Rickert RC. (2003). (2003). Nat Immunol 4: 161–167.
J Immunol 170: 4630–4637. Ouaaz F, Arron J, Zheng Y, Choi Y, Beg AA. (2002).
Liao G, Zhang M, Harhaj EW, Sun S-C. (2004). J Biol Chem Immunity 16: 257–270.
279: 26243–26250. Owyang AM, Tumang JR, Schram BR, Hsia CY, Behrens
Lien E, Chow JC, Hawkins LD, McGuinness PD, Miyake K, TW, Rothstein TL et al. (2001). J Immunol 167: 4948–4956.
Espevik T et al. (2001). J Biol Chem 276: 1873–1880. Ozinsky A, Smith KD, Hume D, Underhill DM. (2000).
Lin X, Wang D. (2004). Semin Immunol 16: 429–435. J Endotoxin Res 6: 393–396.
Lin X, O’Mahony A, Mu Y, Geleziunas R, Greene WC. Parra E, McGuire K, Hedlund G, Dohlsten M. (1998).
(2000). Mol Cell Biol 20: 2933–2940. J Immunol 160: 5374–5381.
Lomaga MA, Yeh WC, Sarosi I, Duncan GS, Furlonger C, Pasare C, Medzhitov R. (2005). Nature 438: 364–368.
Ho A et al. (1999). Genes Dev 13: 1015–1024. Pasparakis M, Schmidt-Supprian M, Rajewsky K. (2002).
Lopez CB, Garcia-Sastre A, Williams BR, Moran TM. (2003). J Exp Med 196: 743–752.
J Infect Dis 187: 1126–1136. Paxian S, Merkle H, Riemann M, Wilda M, Adler G,
Lund J, Sato A, Akira S, Medzhitov R, Iwasaki A. (2003). Hameister H et al. (2002). Gastroenterology 122: 1853–1868.
J Exp Med 198: 513–520. Perkins ND. (2006). Oncogene 25: 6717–6730.
Lund JM, Alexopoulou L, Sato A, Karow M, Adams NC, Pfeifhofer C, Kofler K, Gruber T, Tabrizi NG, Lutz C,
Gale NW et al. (2004). Proc Natl Acad Sci USA 101: Maly K et al. (2003). J Exp Med 197: 1525–1535.
5598–5603. Poligone B, Baldwin AS. (2001). J Biol Chem 276:
Mackay F, Schneider P, Rennert P, Browning J. (2003). Annu 38658–38664.
Rev Immunol 21: 231–264. Prendes M, Zheng Y, Beg AA. (2003). J Immunol 171:
Mambula SS, Sau K, Henneke P, Golenbock DT, Levitz SM. 3963–3969.
(2002). J Biol Chem 277: 39320–39326. Pyatt DW, Stillman WS, Yang Y, Gross S, Zheng JH, Irons
Mansell A, Smith R, Doyle SL, Gray P, Fenner JE, Crack PJ RD. (1999). Blood 93: 3302–3308.
et al. (2006). Nat Immunol 7: 148–155. Rao S, Gerondakis S, Woltring D, Shannon MF. (2003).
Massari P, Ram S, Macleod H, Wetzler LM. (2003). Trends J Immunol 170: 3724–3731.
Microbiol 11: 87–93. Rowe TM, Rizzi M, Hirose K, Peters GA, Sen GC. (2006).
Matsumoto R, Wang D, Blonska M, Li H, Kobayashi M, Proc Natl Acad Sci USA 103: 5823–5828.
Pappu B et al. (2005). Immunity 23: 575–585. Rudolph D, Yeh WC, Wakeham A, Rudolph B, Nallainathan
McDonald PP. (2004). Adv Immunol 82: 1–48. D, Potter J et al. (2000). Genes Dev 14: 854–862.
McDonald PP, Bald A, Cassatella MA. (1997). Blood 89: Ruefli-Brasse AA, French DM, Dixit VM. (2003). Science 302:
3421–3433. 1581–1584.
McWhirter SM, Fitzgerald KA, Rosains J, Rowe DC, Ruefli-Brasse AA, Lee WP, Hurst S, Dixit VM. (2004). J Biol
Golenbock DT, Maniatis T. (2004). Proc Natl Acad Sci Chem 279: 1570–1574.
USA 101: 233–238. Ruland J, Duncan GS, Elia A, del Barco Barrantes I,
Means TK, Wang S, Lien E, Yoshimura A, Golenbock DT, Nguyen L, Plyte S et al. (2001). Cell 104: 33–42.
Fenton MJ. (1999). J Immunol 163: 3920–3927. Ruland J, Duncan GS, Wakeham A, Mak TW. (2003).
Mebius RE. (2003). Nat Rev Immunol 3: 292–303. Immunity 19: 749–758.
Melmed G, Thomas LS, Lee N, Tesfay SY, Lukasek K, Saijo K, Mecklenbrauker I, Santana A, Leitger M, Schmedt C,
Michelsen KS et al. (2003). J Immunol 170: 1406–1415. Tarakhovsky A. (2002). J Exp Med 195: 1647–1652.
Oncogene
NF-jB and immunology
MS Hayden et al
6779
Samson SI, Memet S, Vosshenrich CA, Colucci F, Richard O, Tato CM, Mason N, Artis D, Shapira S, Caamano JC,
Ndiaye D et al. (2004). Blood 103: 4573–4580. Bream JH et al. (2006). Int Immunol 18: 505–513.
Sato S, Fujita N, Tsuruo T. (2000). Proc Natl Acad Sci USA Thomassen E, Renshaw BR, Sims JE. (1999). Cytokine 11:
97: 10832–10837. 389–399.
Sato S, Sanjo H, Takeda K, Ninomiya-Tsuji J, Yamamoto M, Thome M. (2004). Nat Rev Immunol 4: 348–359.
Kawai T et al. (2005). Nat Immunol 6: 1087–1095. Ting AT, Pimentel-Muinos FX, Seed B. (1996). EMBO J 15:
Scheidereit C. (2006). Oncogene 25: 6685–6705. 6189–6196.
Schiemann B, Gommerman JL, Vora K, Cachero TG, Vincenti MP, Brinckerhoff CE. (2002). Arthritis Res 4:
Shulga-Morskaya S, Dobles M et al. (2001). Science 293: 157–164.
2111–2114. Vincenti MP, Coon CI, Brinckerhoff CE. (1998). Arthritis
Schmidt-Supprian M, Tian J, Ji H, Terhorst C, Bhan AK, Rheum 41: 1987–1994.
Grant EP et al. (2004). J Immunol 173: 1612–1619. Voll RE, Jimi E, Phillips RJ, Barber DF, Rincon M, Hayday
Schwarz EM, Krimpenfort P, Berns A, Verma IM. (1997). AC et al. (2000). Immunity 13: 677–689.
Genes Dev 11: 187–197. Wald D, Qin J, Zhao Z, Qian Y, Naramura M, Tian L et al.
Senftleben U, Cao Y, Xiao G, Greten FR, Krahn G, (2003). Nat Immunol 4: 920–927.
Bonizzi G et al. (2001a). Science 293: 1495–1499. Wan YY, DeGregori J. (2003). Immunity 18: 331–342.
Senftleben U, Li ZW, Baud V, Karin M. (2001b). Immunity 14: Ward C, Chilvers ER, Lawson MF, Pryde JG, Fujihara S,
217–230. Farrow SN et al. (1999). J Biol Chem 274: 4309–4318.
Sentman CL, Shutter JR, Hockenbery D, Kanagawa O, Weih DS, Yilmaz ZB, Weih F. (2001). J Immunol 167:
Korsmeyer SJ. (1991). Cell 67: 879–888. 1909–1919.
Serhan CN, Savill J. (2005). Nat Immunol 6: 1191–1197. Weih F, Carrasco D, Durham SK, Barton DS, Rizzo CA,
Seth RB, Sun L, Ea CK, Chen ZJ. (2005). Cell 122: 669–682. Ryseck R-P et al. (1995). Cell 80: 331–340.
Sha WC, Liou H-C, Tuomanen EI, Baltimore D. (1995). Cell Weil R, Schwamborn K, Alcover A, Bessia C, Di Bartolo V,
80: 321–330. Israël A. (2003). Immunity 18: 13–26.
Sharma S, tenOever BR, Grandvaux N, Zhou GP, Lin R, Werner SL, Barken D, Hoffmann A. (2005). Science 309:
Hiscott J. (2003). Science 300: 1148–1151. 1857–1861.
Shim JH, Xiao C, Paschal AE, Bailey ST, Rao P, Hayden MS Wessells J, Baer M, Young HA, Claudio E, Brown K,
et al. (2005). Genes Dev 19: 2668–2681. Siebenlist U et al. (2004). J Biol Chem 279: 49995–50003.
Shinkura R, Kitada K, Matsuda F, Tashiro K, Ikuta K, Wetzler LM. (2003). Vaccine 21(Suppl 2): S55–S60.
Suzuki M et al. (1999). Nat Genet 22: 74–77. Wolfs TG, Buurman WA, van Schadewijk A, de Vries B,
Shoham S, Huang C, Chen JM, Golenbock DT, Levitz SM. Daemen MA, Hiemstra PS et al. (2002). J Immunol 168:
(2001). J Immunol 166: 4620–4626. 1286–1293.
Siebenlist U, Brown K, Claudio E. (2005). Nat Rev Immunol 5: Wu L, D’Amico A, Winkel KD, Suter M, Lo D, Shortman K.
435–445. (1998). Immunity 9: 839–847.
Smith KD, Andersen-Nissen E, Hayashi F, Strobe K, Wu M, Lee H, Bellas RE, Schauer SL, Arsura M, Katz D et al.
Bergman MA, Barrett SL et al. (2003). Nat Immunol 4: (1996). EMBO J 15: 4682–4690.
1247–1253. Wyllie DH, Kiss-Toth E, Visintin A, Smith SC, Boussouf S,
Snapper CM, Rosas FR, Zelazowski P, Moorman MA, Segal DM et al. (2000). J Immunol 165: 7125–7132.
Kehry MR, Bravo R et al. (1996a). J Exp Med 184: 1537–1541. Xia Y, Makris C, Su B, Li E, Yang J, Nemerow GR et al.
Snapper CM, Zelazowski P, Rosas FR, Kehry MR, Tian M, (2000). Proc Natl Acad Sci USA 97: 5243–5248.
Baltimore D et al. (1996b). J Immunol 156: 183–191. Xiao C, Shim JH, Kluppel M, Zhang SS, Dong C, Flavell RA
Sommer K, Guo B, Pomerantz JL, Bandaranayake AD, et al. (2003). Genes Dev 17: 2933–2949.
Moreno-Garcia ME, Ovechkina YL et al. (2005). Immunity Xiao G, Harhaj EW, Sun S-C. (2001). Mol Cell 7: 401–409.
23: 561–574. Xu LG, Wang YY, Han KJ, Li LY, Zhai Z, Shu HB. (2005).
Stetson DB, Medzhitov R. (2006). Immunity 24: 93–103. Mol Cell 19: 727–740.
Su TT, Guo B, Kawakami Y, Sommer K, Chae K, Humphries Yamamoto M, Sato S, Hemmi H, Sanjo H, Uematsu S,
LA et al. (2002). Nat Immunol 3: 780–786. Kaisho T et al. (2002). Nature 420: 324–329.
Sun L, Deng L, Ea CK, Xia ZP, Chen ZJ. (2004). Mol Cell 14: Yamamoto M, Sato S, Hemmi H, Uematsu S, Hoshino K,
289–301. Kaisho T et al. (2003). Nat Immunol 4: 1144–1150.
Sun Z, Arendt CW, Ellmeier W, Schaeffer EM, Sunshine MJ, Yarovinsky F, Zhang D, Andersen JF, Bannenberg GL,
Gandhi L et al. (2000). Nature 404: 402–407. Serhan CN, Hayden MS et al. (2005). Science 308:
Tada K, Okazaki T, Sakon S, Kobarai T, Kurosawa K, 1626–1629.
Yamaoka S et al. (2001). J Biol Chem 276: 36530–36534. Yeh WC, Shahinian A, Speiser D, Kraunus J, Billia F,
Takaesu G, Surabhi RM, Park KJ, Ninomiya-Tsuji J, Wakeham A et al. (1997). Immunity 7: 715–725.
Matsumoto K, Gaynor RB. (2003). J Mol Biol 326: Yilmaz ZB, Weih DS, Sivakumar V, Weih F. (2003). EMBO J
105–115. 22: 121–130.
Takeshita F, Leifer CA, Gursel I, Ishii KJ, Takeshita S, Yoneyama M, Kikuchi M, Matsumoto K, Imaizumi T,
Gursel M et al. (2001). J Immunol 167: 3555–3558. Miyagishi M, Taira K et al. (2005). J Immunol 175:
Takeuchi O, Hoshino K, Akira S. (2000). J Immunol 165: 2851–2858.
5392–5396. Yoneyama M, Kikuchi M, Natsukawa T, Shinobu N,
Takeuchi O, Kawai T, Muhlradt PF, Morr M, Radolf JD, Imaizumi T, Miyagishi M et al. (2004). Nat Immunol 5:
Zychlinsky A et al. (2001). Int Immunol 13: 933–940. 730–737.
Takeuchi O, Sato S, Horiuchi T, Hoshino K, Takeda K, Yujiri T, Ware M, Widmann C, Oyer R, Russell D, Chan E
Dong Z et al. (2002). J Immunol 169: 10–14. et al. (2000). Proc Natl Acad Sci USA 97: 7272–7277.
Tanabe T, Chamaillard M, Ogura Y, Zhu L, Qiu S, Zelazowski P, Carrasco D, Rosas FR, Moorman MA, Bravo
Masumoto J et al. (2004). EMBO J 23: 1587–1597. R, Snapper CM. (1997). J Immunol 159: 3133–3139.
Oncogene
NF-jB and immunology
MS Hayden et al
6780
Zhang D, Zhang G, Hayden MS, Greenblatt MB, Bussey C, Zhang SQ, Kovalenko A, Cantarella G, Wallach D. (2000).
Flavell RA et al. (2004). Science 303: 1522–1526. Immunity 12: 301–311.
Zhang G, Ghosh S. (2002). J Biol Chem 277: Zhou H, Wertz I, O’Rourke K, Ultsch M, Seshagiri S, Eby M
7059–7065. et al. (2004). Nature 427: 167–171.

Oncogene

Vous aimerez peut-être aussi