Download as pdf or txt
Download as pdf or txt
You are on page 1of 24

C H A P T E R

35

Tumor necrosis factor


Haichao Wang 1,2, Christopher]. Czura I a n d Keuin ]. Tracey 1
1North Shore-LIJ Research Institute, Manhasset, New York, USA
2North Shore University Hospital, Manhasset, New York, USA

Science is w o n d e r f u l l y e q u i p p e d to a n s w e r the q u e s t i o n 'How?'


b u t it gets terribly c o n f u s e d w h e n you ask the q u e s t i o n 'Why?'
Erwin Chargaff

INTRODUCTION STRUCTURE

Tumor necrosis factor (TNE TNF-a, cachectin) is a Tumor necrosis factor is synthesized as a 26-kDa type
pleiotropic pro-inflammatory cytokine that exerts II transmembrane precursor that is displayed on the
multiple biologic effects. TNF was originally identified plasma membrane, with the N-terminus in the cyto-
as an anti-tumor agent that induced necrotic cell plasm and the C-terminus exposed to the extracellu-
death in sarcomas and other tumor types. Localized, lar space. The TNF precursor is proteolytically cleaved
low-level expression of TNF participates in beneficial between alanine (-1) and valine (+ 1) to yield a bio-
tissue remodeling and host defense responses. The logically active 17-kDa mature TNF (Kriegler et al.,
expression of TNF is tightly controlled, because sys- 1988) that forms a trimer in solution (Smith and
temic overproduction of TNF activates inflammatory Baglioni, 1987). In solution, 17-kDa TNF is primarily
responses to infection and injury, and mediates comprised of two anti-parallel [3 sheets; these TNF
hypotension, diffuse coagulation, and widespread monomers self-associate in a head-to-tail fashion
tissue damage. The diverse activities of TNF led to the into non-covalently linked, symmetrical bell-shaped
simultaneous and apparently paradoxical pursuit of homotrimers (Smith and Baglioni, 1987; Jones et al.,
TNF as an anti-tumor strategy, and TNF inhibitors to 1989). The C-terminus of each subunit is embedded
attenuate lethal systemic inflammation. This chapter within the base of the trimer, while the N-terminus is
has been written as an introduction to the broad topic relatively free from the structure and is not critical for
of TNE its molecular biology, biochemistry, and TNF biologic activity (Creasey et al., 1987). Mutational
physiology; by necessity, only a portion of the primary analyses of mature TNF indicate that each trimer
literature for each topic is reviewed. has three receptor interaction sites located in the

The Cytokine Handbook, 4th Edition, edited by Angus W. Thomson & Michael T Lotze Copyright 9 2003 Elsevier Science Ltd.
ISBN 0-12-689663-1, London All rights of reproduction in any form reserved.
838 TUMOUR NECROSIS FACTOR

intersubunit grooves near the base of the trimer but forms an a-helical structure in the molten globule
(Zhang et al., 1992). The trimeric structure of TNF is (an intermediate structure produced by acid-induced
important for its biologic activity, because mutations unfolding of proteins). With the resulting increase
that destabilize the trimer or attenuate monomer in hydrophobicity at low pH, TNF becomes more
association result in the loss of TNF biologic activity cytolytic, perhaps by insertion into membrane to
(Lin, 1992). Recently identified TNF muteins have form sodium ion channels (Tracey et al., 1986;
been shown to possess decreased toxicity in mice Yoshimura and Sone, 1987; Baldwin et al., 1996). The
without loss of its antitumor effects (Van Ostade et al., conformational change at low pH may be physiologi-
1993; Barbara et al., 1994). For example, residues cally relevant, because a pH of about 3.6 has been
within the inter-subunit grooves at the base of the reported in the space between macrophages and their
trimer dictate receptor specificity: the double mutant substrates (Silver et al., 1988). Recent membrane con-
R32W/S86T exhibited wild-type binding to the 55- ductance studies have shown that the capacity of TNF
kDa receptor (TNFR1) with no measurable binding to to increase membrane conductance does not corre-
the 75-kDa TNF receptor (TNFR2). In contrast, the late with its ability to interact with membranes. These
D143N/A145R double mutant fails to bind TNFR1, but results suggest that TNF itself does not form a chan-
does bind TNFR2 with five- to 10-fold lower affinity nel, but may increase sodium flux via interaction with
than wild-type TNF (Loetscher et al., 1993). There endogenous ion channels or with plasma membrane
may be differences between species, because the proteins that are coupled to ion channels (van der
R32W/S86T double mutant, which was non-toxic in Goot et al., 1999).
mice, was significantly toxic to healthy baboons (Van
Zee et al., 1994). A recently identified triple mutant
($52I, Y56F, plus deletion of the seven N-terminal BIOSYNTHESIS AND
residues) has 11- and 71-fold lower affinity for the 55- REGULATION
kDa and 75-kDa receptors, respectively, and is 10-fold
less toxic than wild-type TNF (Cha et al., 1998). These TNF is produced by numerous immune cells in
findings have raised hope for an engineered TNF response to an assortment of activating stimuli (Table
protein that possesses tumor cytotoxicity activity, but 35.1). Among these stimuli, lipopolysaccharide (LPS)
with reduced systemic toxicity as compared with the (in monocytes), T-cell receptor activation (in T lym-
native cytokine. phocytes), crosslinking of surface immunoglobulin
Transmembrane pro-TNF is also biologically active, (sIg) (in B lymphocytes), ultraviolet light (in epithelial
but less is known about its structure-function rela- cells), and viral infections have been widely studied.
tionship. Prior to transport to the cell surface, pro- Suppressors of TNF expression in macrophages are
TNF is assembled into a bell-shaped trimer that is also highly diverse and include prostaglandin E 2
similar to the structure of mature TNF (Jones et al., (PGE2), transforming growth factor 13 (TGFI3), IFN~,
1989). A 20-residue linker sequence connects the IFNT, IL-4, IL-6, IL-10, G-CSE certain viral products
transmembrane domain with the N-terminus, which such as adenoviral proteins, dexamethasone, gluco-
facilitates but is not necessary for trimer formation. corticoids, cyclosporin A, spermine, pentoxifylline
During the interaction of effector and target cells, cAME and cholinergic agonists including acetyl-
membrane-bound pro-TNF in the effector cells binds choline and nicotine.
TNF receptors on the target cells, and induces TNF The biosynthesis of TNF is tightly controlled at
responses via receptor aggregation (Kriegler et al., many different levels to ensure the production of van-
1988). Pro-TNF is a more potent activator of TNF ishingly small quantities in quiescent cells, yet TNF is
receptor 2 (TNFR2) than mature TNF (Grell et al., capable of rapid and significant up-regulation in acti-
1994). vated cells (Beutler et al., 1985). TNF is expressed as
Highly acidic environments alter the tertiary struc- an immediate early gene, and a variety of stimuli
ture of TNF such that the 13-sheet structure is aban- induce high levels of TNF mRNA within 15-30 min-
doned in favor of ~ helices (Narhi et al., 1996). TNF is utes with no requirement for de novo protein synthe-
unusual in that it has no ~ helix in its native structure sis, suggesting that the factors necessary for the

THE CYTOKINES AND CHEMOKINES


BIOSYNTHESIS AND REGULATION 839

Table 35.1 Cell source a n d stimuli o f TNF production Y box SP1 NFAT NFAT ~:3 AP1 AP2
-232 -164 -149 -117 -89 -58 -27
I I , I, I ,I , I , I , I , [TNFGENE~"
Cell sources I m m u n e cells: ~:1 ~:2 Egr-1 Ets-1 CRE NFAT SP1 TATA
Macrophages/monocytes, natural -577 -201 -163 -118 -100 -76 -45 -20
killer cells, Kupffer cells, B cells, T
cells, basophils, eosinophils, glial FIGURE 35.1 Model of the TNF promoter. Putative
cells, mast cells cis-acting consensus sequences are denoted by boxes
N o n - i m m u n e cells: and numbers indicating the position in relation to the
Astrocytes, granulosa cells, transcription start site.
osteoblasts, cardiac myocytes,
fibroblasts, keratinocyte, neurons,
neutrophils, T cells, retinal pigment specific (Ets) binding site (Kramer et al., 1995); and an
epithelial cells, smooth muscle cells, early growth responsive- 1 (Egr-1) binding site
spermatogenic cells, tumor cells (Kramer et al., 1994). Although K3 matches the bind-
Stimuli Bacterial endotoxin (LPS), antibodies ing consensus sequence for the transcription factor
to LFA-3, calcium ionophores, C5a, NFKB, K3 appears to bind to NFATp instead of NFKB.
CD44, CD45, enterotoxin, GM-CSE The CRE element in TNF promoter binds mainly to
hypoxia, IL-1, leukotrienes, mellitin, ATF-2/Jun. Genetic analyses suggest that additional
MIP- 1~, mycobacterial
lipoarabinomannan, nitric oxide, regulatory elements may be important in regulating
oxygen radicals, parasites, phorbol TNF transcription (Wong and Goeddel, 1988). TNF
esters, synthetic lipid A, TNE toxic mRNA expression is negatively regulated by factors
shock toxin- 1, viruses that are less well characterized. Several regions within
Irradiation the TNF promoter, including b a s e s - 2 8 0 to -172,
-254 to -230, -125 to -82, and - 9 5 to -36, relative
induction of TNF expression pre-exist in unstimu- to the transcription start site, seem to be critical for
lated cells. The cell-type-specific regulation of TNF inhibition ofTNF mRNA expression (Fong et al., 1995;
synthesis is reflected by the differential use of regula- An et al., 1999). The transcriptional regulators that
tory elements on TNF biosynthesis in different cells. bind these regions remain enigmatic. While AP-1 and
Each one of the synthetic steps from sensing the pres- AP-2 consensus binding sequences have been noted
ence of a stimulus to TNF secretion is controlled by within these regions (An et al., 1999), evidence for
different molecular events regulating the TNF gene, binding of these repressors to the TNF promoter is
mRNA and protein. lacking (Fong et al., 1995). More recently, Singh et al.
(2001) have found that heat-shock factor 1 (HSF1)
binds to regions - 1080 to -845, -533 to -196 and
Transcriptional regulation -326 to - 3 9 of the TNF promoter relative to the
TNF is regulated at the transcriptional level by both transcription start site, and represses TNF gene
activators and repressors. Besides a TATA box pro- transcription.
moter located 20 base pairs (bp) upstream from the The regulation of TNF transcription is cell-type spe-
transcription start site, a number of regulatory cific, partially because of differential use of these reg-
sequences are also found upstream of the TNF gene ulatory elements on the TNF promoter. In calcium- or
(Figure 35.1), including three nuclear factor (NF)~:B T-cell ligand-activated At-5 T lymphocytes, a func-
sites designated ~:1, ~:2 and ~:3 (Goldfeld et al., 1993); tional interaction between NFATp on the K3 site and
three nuclear factor of activated T cells (NFAT) bind- ATF-2/Jun on the CRE element, but not AP-1 or AP-2,
ing sites for NFATp, NFAT-149, NFAT-117 and NFAT-76 plays a crucial role in the induction of TNF expression
(Tsai et al., 1996); one 'Y' box; one cAMP-responsive (Tsai et al., 1996). In calcium-activated A20 B lympho-
element (CRE) for activation transcription factor-2 cytes, NFATp on the K3 element is not required, but
(ATF-2)/Jun (Tsai et al., 1996); two SV40 promoter-1 NFATp on NFAT-76 participates in the induction of
(SP-1) sites (Kramer et al., 1994); one activating pro- TNF synthesis (Tsai et al., 1996). In phorbol myristate
tein- 1 (AP-1) and one AP-2 binding site for Fos/Jun acetate (PMA)-activated T cells, the interaction
(Leitman et al., 1992); one E26 transformation- between Ets and ATF/Jun is essential for both basal

THE CYTOKINES AND CHEMOKINES


840 T U M O U R NECROSIS FACTOR

and induced TNF expression. Post-translational mod- proteins, such as TIA-1, normally function as transla-
ification of the transcription factors that regulate TNF tional silencers (Piecyk et al., 2000), because
transcription, such as NFATp, NF)cB and AP-1, has macrophages derived from TIA-1 knockout mice have
been implicated in the regulation of TNF induction. a significantly higher proportion of TNF transcript
Protein kinases and phosphatases are also involved in associated with polysomes. TIA-1 knockout mice fail
the induction of TNF (Lee et al., 1994), as are pro- to inhibit TNF production and are exquisitely sensi-
teases and phospholipase D (Balboa et al., 1992). tive to endotoxemia (Piecyk et al., 2000). The 3' UTR of
Some of these enzymes modify the activity of TNF TNF mRNA is equally important for stimulating TNF
induction-related transcription factors such as NFKB synthesis, because a polymorphism (a GAU trinu-
and AP- 1 (see below). cleotide insertional mutation) present in the regula-
tory 3' UTR ofTNF mRNA of NZW mice prevents the
interaction of RNA-binding proteins with target
Translational regulation sequences, and significantly reduces TNF production
TNF synthesis is highly inducible upon cell stimula- (Di Marco et al., 2001).
tion, and is tightly regulated in quiescent cells; this
enforced inhibition of TNF expression protects
against the harmful effects of TNF overexpression. In
Signal transduction cascades
response to LPS stimulation, macrophages and
regulating TNF production
monocytic cells produce high levels of TNF protein, Mitogen-activated protein (MAP) kinases such as the
an effect that is blocked by pretreatment with dexam- extracellular signal-regulated kinase (ERIO, c-Jun N-
ethasone, spermine or CNI-1493 (a tetravalent terminal kinase (JNIO, p38 and Big MAP kinase
guanylhydrazone) (Bianchi et al., 1996; Beutler et al., (BMIO/ERK5 have a critical role in the regulation of
1986; Cohen et al., 1996; Zhang et al., 1997). Recent TNF production (Zhu et al., 2000). The roles of
studies have shown that cholinergic agonists such as ERK1/2- and p38 MAPK-dependent pathways in reg-
acetylcholine and nicotine inhibit TNF synthesis via ulating cytokine production have been demonstrated
post-transcriptional mechanisms (Borovikova et al., primarily through the use of specific inhibitors (such
2000). A key element for both negative and positive as PD098059 for ERK1/2 activation, and SB203580 for
translational regulation of TNF has been identified in p38 MAP kinase) (Cuenda et al., 1995; Alessi et al.,
the 3' untranslated region (UTR) of TNF mRNA and 1995), which prevent cytokine production in LPS-
many other cytokines, growth factors and oncopro- stimulated macrophage/monocytes (Feng et al., 1999;
teins (Caput et al., 1986; Han et al., 1990). The mini- Guha et al., 2001). p38 MAP kinase may be critical in
mal motif UUAUUUAUU renders the TNF mRNA the translational control of TNF synthesis, because
unstable (Shaw and Kamen, 1986; Zubiaga et al., activation of the MAP kinase cascade enhances the
1995), and the presence of a single octanucleotide translational efficiency of TNF mRNA (Lee et al.,
(UUAUUUAU) in the 3' UTR is sufficient to signifi- 1994). Mitogen-activated protein (MAP) kinase/extra-
cantly inhibit translation (Kruys et al., 1989). This cellular signal-regulated kinase (ERIO kinase (MEKIO
sequence physically interacts with the poly(A) tail of activates TNF induction-associated transcription fac-
the TNF mRNA, preventing the mRNA from forming tors such NFKB, c-Jun and p38 (Cohen et al., 1996; Lee
large polysomes (Crawford et al., 1997). Regulation of S.Y. et al., 1996). An abridged listing of transcriptional
mRNA turnover is mediated through this region by and translational control mechanisms in TNF synthe-
trans-acting proteins such as tristetraprolin (TTP) sis is shown in Figure 35.2. Multiple signaling path-
(Lai et al., 1999), HuR (Dean et al., 2001), TIA- 1 (Piecyk ways converge on NFKB, a ubiquitous transcription
et al., 2000), and TIAR (Gueydan et al., 1999), which factor involved in the transduction of activation sig-
bind AO-rich elements and stabilize or destabilize the nals from the cytoplasm to the nucleus. NFKB resides
transcript. Mice deficient in tristetraprolin (TTP), the in the cytosol as a trimer consisting of p50, p65
prototype of a family of CCCH zinc-finger proteins, and I~:B subunits. Upon stimulation, IKB is released,
develop an inflammatory syndrome mediated by and the p50/p65 heterodimer migrates to the nucleus
excess TNF (Lai et al., 1999). Other AO-rich-binding where it interacts with target DNA sequences

THE CYTOKINES A N D C H E M O K I N E S
BIOSYNTHESIS AND REGULATION 841

I Inflammatory stimuli , I modulate MMP levels (Panagakos et al., 1996), and


plasminogen and plasmin increase secretion of
MMPs and induce cleavage to their active forms (Lee
I aa,c/ 0dc421 et al., 1996).

IMEKK,,21 I ASK1 .....I Application


Uncontrolled synthesis of TNF has been implicated in
IMKK3,61 many human diseases of systemic inflammation,
including sepsis, arthritis and Crohn disease. Conse-
I ". I quently, there has been a great deal of interest in
developing therapeutic strategies to inhibit TNF activ-
I MAPKAP:K2 1 ity, and several inhibitors have been developed to
attenuate TNF activity, release, translation or tran-
scription. Corticosteroids, the most studied class of
compounds that suppress the production of TNF,
inhibit the transcription of TNF and other cytokines.
Y
j
One immunosuppressive mechanism of these drugs
is through the stimulation of IKB~ production
TNF DNA ~ ,. mRNA ,. Protein
(Auphan et al., 1995; Scheinman et al., 1995), which in
FIGURE 35.2 Schematic illustration of signal trans- turn results in the inactivation of NFKB. Corticos-
duction pathways regulating TNF transcription and teroids also inhibit gene transcription by preventing
translation in macrophages. the transcription factor, AP-1, from binding to its tar-
get promoter (Marx, 1995). Pentoxifylline and other
resulting in the transcription of TNF and other pro- protein kinase C (PKC) inhibitors regulate the pro-
inflammatory cytokines. duction of TNF and other cytokines by blocking PKC-
or PKA-catalyzed activation of NF~:B (Biswas et al.,
1994). Cytokine-suppressing anti-inflammatory drugs
Post-translational regulation (CSAIDs) (Young et al., 1994) have no significant effect
TNF is synthesized as a 233-amino-acid membrane- on the transcription of TNF or other cytokines, but
anchored prohormone that is subsequently processed rather suppress TNF translation by inhibiting the
proteolytically to yield the mature 157 residue activity of p38 MAP kinase (Lee et al., 1994). CNI- 1493,
cytokine. Analysis of the cleavage site sequences in a tetravalent guanylhydrazone that suppresses sys-
human TNF revealed significant homology with temic and central inflammation (Meistrell et al., 1997;
sites in collagen, %-inhibitory proteins, and %- Tracey, 1998), inhibits TNF by preventing phosphory-
macroglobulin cleaved by matrix metalloproteinases lation of p38 MAP kinase (Cohen et al., 1996; Denham
(MMPs). The TNFcz-converting enzyme (TACE) is an et al., 2000). At the post-translational level, thalido-
MMP-like enzyme (Black et al., 1997; Moss et al., mide selectively suppresses the release of TNF but not
1997), and metalloproteinase inhibitors block TNF of other cytokines (Sampaio et al., 1991). IL-4 and
processing (Gearing et al., 1994). Gearing and col- IL-10 suppress TNF activity by inhibiting the bio-
leagues (1995) have found that TACE activity is synthesis of matrix-destructive metalloproteinases
expressed ubiquitously, even in HeLa cells that do not (Lacraz et al., 1992, 1995) and down-regulating the
produce TNE and in insect cells where pro-TNF is proteolytic processing of pro-TNF to its mature form.
expressed by means of a baculovirus transfectant. A Furthermore, IL-4 inhibits the activation of the tran-
serine protease, proteinase 3 (PR-3), may also process scriptional factors NFKB and AP-1, providing yet
TNE and serve as an extracellular alternate processing another mechanism by which IL-4 can attenuate TNF
enzyme for TNF and IL-I[3 (Coeshott et al., 1999). activity. Like IL-4, IL-10 also inhibits TNF as well as
Endotoxin, as well as TNF and other cytokines, can other cytokines at multiple levels (Bogdan et al., 1992).

THE CYTOKINES AND CHEMOKINES


842 TUMOUR NECROSIS FACTOR

RECEPTORS seem to be sufficient to stimulate these functions


(Barbara et al., 1994). These observations suggest that
The pleiotropic effects of TNF are mediated through the role of TNFR2 may be to potentiate the effects of
two distinct but structurally homologous TNF recep- TNFR1. A unique 'passing model' has been suggested
tors, the 60-kDa receptor 1 (TNFR1, also known as to explain the role of TNFR2 in mediating TNF
p55 or p60) and the 80-kDa receptor 2 (TNFR2, also responses (Tartaglia et al., 1993b). The rapid rate of
known as p75 or p80), which are both type I trans- dissociation of the TNF-TNFR2 complex may facili-
membrane glycoproteins and members of the TNF tate the interaction of TNF with TNFR1 by recruiting
receptor superfamily. Members of this receptor family TNF to the vicinity of the plasma membrane. In this
share sequence homologies within their extracellular scenario, TNFR2 can be envisioned as passing TNF to
domains, consisting of multiple cystine-rich repeats TNFR1, the main TNF signal transducer. In some
of about 40 amino acids in length (Mallett and cases, however, TNFR2 is also believed to mediate
Barclay, 1991). TNFR1 and TNFR2 are present on several TNF effects, such as proliferation of T and B
virtually all cell types except for red blood cells cells (Tartaglia et al., 1991; Barbara et al., 1994), induc-
(Hohmann et al., 1989). TNFR1 is more ubiquitous, tion of NF~:B, and cytotoxicity (Hohmann et al., 1990;
but TNFR2 is more abundant on endothelial cells and Grell et al., 1994). TNF receptor activities are also sub-
cells of hematopoietic lineage (Hohmann et al., 1989; ject to regulation by a wide variety of agents such as
Brockhaus et al., 1990). Both receptors can bind to PKC and PKA modulators, LPS, retinoic acid, gluco-
TNF with high affinity; the dissociation constants (Ka) corticoids and cytokines including TNF; IL-2, IL-4,
are 2-5 • 10-~~ 3-7 • 10 -11, respectively. IL-6, IL-8; IFNct, 13,7; GM-CSF; and thyroid-stimulating
The precise roles of the TNF-induced receptor sig- hormone. Therefore, expression of either TNFR1 or
naling in mediating the effects of TNF continue to be TNFR2 is dependent upon cell type, as well as extra-
the focus of intensive study. TNFR1 is generally con- cellular stimuli.
sidered to be responsible for the majority of biologic Soluble receptors for TNF (sTNFR, also known as
actions of soluble TNE Direct signaling through TNF-BP for TNF-binding protein) are derived from
TNFR2 occurs less extensively and appears to be con- both TNFR1 and TNFR2 by proteolytic processing.
fined mainly to cells of the immune system, and may Both sTNFR1 and sTNFR2 are normally present in
be especially important during cell-cell contact blood and urine (Seckinger et al., 1989), and may be
(Kriegler et al., 1988). Fibroblasts derived from produced by monocytes and macrophages (Gatanaga
TNFRl-knockout mice are deficient in leukocyte et al., 1991; Leeuwenberg et al., 1994). Increased levels
adhesion, and display reduced expression of inter- of sTNFRs are found in many human diseases includ-
cellular adhesion molecule- 1 (ICAM-1), vascular cell ing rheumatoid arthritis, systemic lupus erythemato-
adhesion molecule-1 (VCAM-1), and CD44. These sus, malignancy, sepsis, after surgery and chronic
cells also fail to up-regulate major histocompatibility infection (Chikanza et al., 1993; Aderka, 1996). A num-
complex (MHC) class I expression, cytokine secretion, ber of stimuli, including TNE IL-1, PMA and endo-
cell proliferation, and NFKB activation. These activi- toxin, trigger proteolysis of the TNFRs, resulting in
ties appear to be specific attributes of the TNFR1 increased levels of sTNFRs in the circulation. The pro-
receptor, because activation of cells expressing only teases responsible for this processing have not yet
TNFR2 with exogenous TNF has no effect on these been identified, but may involve the same metallo-
functions (Mackay et al., 1994). Similar observations proteinases that process the TNF precursor to its
were made with TNF mutants that selectively bind mature form, because metalloproteinase inhibitors
TNFR1. TNFRl-selective TNF mutants that bind block the shedding of both TNF receptors (Crowe
TNFR2 poorly are equally cytotoxic and cytostatic to et al., 1995; Mullberg et al., 1995). Recently, matrix
carcinoma and leukemic cell lines, but are less pro- metalloproteinase processing of both precursor TNF
inflammatory, than wild-type TNE TNFRl-selective and the TNF receptors has been implicated in
mutants also recapitulate the hypotensive and proin- humans in vivo; administration of an MMP inhibitor
flammatory activities of wild-type TNF in vivo (Van prior to endotoxin infusion attenuated the release
Zee et al., 1994), whereas TNFR2 by itself does not of both TNF and TNFRs into the circulation. This

THE CYTOKINES AND CHEMOKINES


SIGNAL TRANSDUCTION 843

attenuation was accompanied by an increase in similarities with other known cell surface receptor
membrane-bound TNF and TNFRs in monocytes iso- signaling pathways. X-Ray data suggest that the TNF
lated from these patients (Dekkers et al., 1999). Solu- homotrimer induces receptor trimerization, which
ble TNFRs bind and neutralize both circulating and activates receptor signaling. Several downstream sig-
membrane-bound TNF (Seckinger et al., 1989; Kohno naling events initiated by the TNF receptor complex
et al., 1990). In vivo, administration of sTNFRs neu- have been characterized, and signaling molecules
tralizes or modulates TNF activity when sTNFRs are that mediate the initial interaction with the ligand-
present in high concentrations; lower concentrations, occupied receptor have been identified.
however, stabilize the TNF trimer, providing a TNF
reservoir (Mohler et al., 1993).
TNFRl-mediated signaling pathway
Multiple intracellular functional domains within
SIGNAL TRANSDUCTION TNFR1 transduce intracellular signals to coordinate
the biological activity of TNF (Tartaglia et al., 1993a)
TNF mediates or initiates diverse biologic responses by interacting with intracellular adaptor proteins
by interaction with TNFR1 or TNFR2. Investiga- (Figure 35.3). Three functional domains of TNFR1 -
tions into the signaling events activated by the lig- the C-terminal death domain (Tartaglia et al.,
and/receptor complex have been hampered by the 1993a), and the adjacent N-SMase (neutral sphingo-
diversity of cell-specific TNF responses, and a lack of myelinase) and A-SMase (acidic sphingomyelinase)

.SD I ASD Deathdomain TNFR1

I~N I I ~c-P,C I
I

N-terminus Death domain

N-SMase [ LA-sMase II P~C I


Ceramide
(membrane)
Ceramide
(endosome) I,~ I--IN'~ I L~'DD L ~-I P~D,P~-~ I
I MEKKI'2 II Caspase-2 II Caspase-8 I
I~ I
NF-KB /I-KB
complex IJNKI Protease cascade

~ er0r~
Cell growth Inflammation Anti-apoptosis Apoptosis

FIGURE 35.3 The TNF receptor 1 (TNFR1)-mediated signaling transduction pathways. TNF triggers different sig-
naling pathways via three functional domains of TNFR1 to mediate various activities.

THE CYTOKINES AND CHEMOKINES


844 TUMOUR NECROSIS FACTOR

activating domains (NSD and ASD) (Schutze et al., pathways: one that is dependent upon TRAF2 and
1992; Wiegmann et al., 1994) - transfer signals from activates inflammatory and antiapoptotic responses
extracellular TNF to intracellular adaptor proteins via NFKB and c-Jun; a second pathway that activates
that can contain additional death domains and/or caspase 2 through the activity of RIP to induce apop-
caspase recruitment domains (CARD). TRADD tosis; and a caspase 8-dependent apoptotic pathway
(TNFRl-associated death domain protein) binds to that is dependent upon FADD activity.
the death domain ofTNFR1 (Hsu et al., 1995) to medi- The TNFR1-TRADD complex initiates apoptotic
ate both apoptotic and antiapoptotic pathways. signaling through pathways dependent upon either
Proapoptotic effects of TNF are also mediated FADD or RIP. Overexpression of FADD (Chinnaiyan
through activation of phosphatidylcholine-specific et al., 1995; Boldin et al., 1996) or RIP (Stanger et al.,
phospholipase c (PC-PLC) by the ASD domain 1995) results in apoptosis; however, a dominant-
(Wiegmann et al., 1994; Machleidt et al., 1996), which negative mutant of FADD inhibits TNF-induced
shares receptor regions and function with the death apoptosis but not TNF-mediated NFKB activation
domain. The factor associated with N-SMase activa- (Hsu et al., 1996), demonstrating that the apoptotic
tion (FAN) binds the NSD domain of TNFR1 (Adam- and inflammatory cascades diverge at the level of the
Klages et al., 1996) to mediate a number of TRADD-FADD complex. Activated FADD recruits the
inflammatory and cell proliferation responses. FADD-homologous ICE/CED-3-1ike protease (FLICE),
Together, it is apparent that TNFR1 signaling diverges also referred to as pro-caspase 8 or MACH, which
into three functional branches at the level of the interacts with the caspase recruitment domains
receptor itself." one that is mediated through the death (CARD) of FADD, where it oligomerizes and auto-
domain to positively and negatively regulate apop- proteolytic activity generates catalytically active
totic responses, and two that are processed through capase-8 (Boldin et al., 1996; Muzio et al., 1996). The
sphingomyelinases to modulate apoptotic and FADD-FLICE interaction is negatively regulated by
inflammatory pathways. PED/PEA- 15, possibly by the dissociation of the com-
plex (Condorelli et al., 1999). The death-associated
protein (DAP) kinase is another death domain-
Death domain-mediated pathways
containing protein that operates downstream of
The TNFRl-associated death domain protein FLICE but upstream of other caspases to activate
(TRADD) mediates both apoptosis and antiapoptosis apoptosis (Cohen et al., 1999). The FADD-activated
pathways by activating the interleukin-1 converting caspase cascade induces apoptosis by proteolytically
enzyme (ICE) pathway or the NFKB pathway, respec- activating protein precursors, such as lamin, actin
tively (Hsu et al., 1995). The TRADD protein contains and polymerase, or by activating proteins that lead to
a death domain similar to the death domain of cell death (Figure 35.3).
TNFR1. While overexpression of an isolated TRADD TNFR1 can also induce apoptosis via the TRADD-
death domain can trigger cell death, overexpression of RIP pathway. All three RIP family members (RIP, RIP2
the full-length protein does not, suggesting that [also known as Rick or CARDIAK] or RIP3) possess
TRADD does not carry a caspase recruitment domain protein kinase and death domains, and transduce
(CARD). This has led to one currently accepted view of intracellular apoptotic signals. The death domain of
TRADD as an adaptor protein that links TNFR1 to RIP can mediate the death signal in the absence of the
other death effectors. Once bound to TNFR1, TRADD kinase domain, suggesting that RIP does not possess a
recruits MORT1/FADD (Fas-associating protein with CARD but rather induces apoptosis by recruiting
death domain) or RIP (receptor-interacting protein) other downstream effector molecule(s) through the
through its C-terminal death domain to activate RIP death domain. RIP binds preferentially to TRADD
divergent apoptotic pathways (Grimm et al., 1996), via protein-protein interaction between the death
and recruits TNF receptor-associated factor 2 (TRAF2) domains on both proteins (Hsu et al., 1996), but can
through its N-terminal TRAF-interacting domain also bind weakly to TNFR1 (Stanger et al., 1995).
(Hsu et al., 1996). Thus, TNFR1 death domain signal- TNFR1-TRADD-RIP activate apoptosis via the inter-
ing diverges at the level of TRADD into three distinct mediate effector, RAIDD (for RIP-associated Ich-l/

THE CYTOKINES AND CHEMOKINES


SIGNAL TRANSDUCTION 845

CED-3 homologous protein with death domain), the C-terminal WD repeats of FAN and a cytoplasmic,
which binds RIP via a death domain (Duan and Dixit, nine-amino-acid binding motif (residues 310-318) of
1997). The RAIDD/RIP complex recruits caspase-2 TNFR1 (Adam-Klages et al., 1996). FAN specifically
(Ich-1) to activate the protease cascade leading to activates N-SMase, and not A-SMase (see ASD-
apoptosis. mediated pathway below), which modulates the pro-
RIP can also bind to TRAF2, which can be recruited duction of ceramide, a product of the sphingomyelin
to TRADD simultaneously with RIP. The significance (SM) degradation. Ceramide is generated at several
of RIP binding to TRADD and TRAF2 as well as TNFR1 distinct subcellular locations that influence its action
is still obscure, and it is possible that RIP may coordi- (Wiegmann et al., 1994). Activated N-SMase func-
nate these signaling pathways. Overexpression of tions at the outer leaflet of the plasma membrane to
TRAF2 activates NF~:B (Rothe et al., 1995) but TRAF2 trigger the degradation of sphingomyelin (SM) into
is not required for the induction of apoptosis (Hsu ceramide, which in turn mediates further down-
et al., 1996). A serine-threonine kinase known as stream events in NSD-mediated TNF response.
NFKB-inducing kinase (NIK) binds specifically to Ceramide is also produced in endosomes by A-SMase,
TRAF2 and mediates TRAF2-dependent activation of where ceramide mediates a distinct signaling path-
NF~cB (Malinin et al., 1997). The binding between way. On the plasma membrane, ceramide further
TRAF2 and NIK is mainly dependent on the C-terminus activates ceramide-activated protein kinase (CAPK)
of TRAF2, but also involves the N-terminal of TRAF2. (Winston and Riches, 1995), which phosphorylates
Because NIK can be autophosphorylated and has cytoplasmic raf-1. Activated raf-1 can activate the
sequence similarity to several kinases that participate MAP kinase cascade and activate MAPK/ERK (extra-
in MAP kinase cascades, it may act in a kinase cascade cellular signal-regulated kinase), which in turn
to activate NF~cB. The cascade downstream of NIK activates PI2K2 (Lin et al., 1993). PUK2 may be respon-
that is responsible for NFKB activation, however, sible for the generation of the arachidonic acid
is still unclear. TRAF2-dependent activation of metabolites, leukotrienes and prostaglandins, which
MEKK and c-Jun amino-terminal kinase (JNK) / stress- contribute to TNF proinflammatory activities.
activated protein kinase (SAPK) through TNFR1 has MAPK/ERK also induces the expression of c-myc and
been reported (Liu et al., 1996; Natoli et al., 1997). As other genes to regulate cell proliferation and differen-
MEKK has been shown to induce the phosphorylation tiation. Activation of ERK may act as a negative-
and degradation of IKB~, resulting in the activation feedback loop to attenuate TNF-induced apoptosis,
of NFKB (Lee et al., 1997), MEKK may function as because inhibition of ERK is required for induction of
a downstream factor of NIK and mediate TRADD- apoptosis in some systems (Xia et al., 1995).
TRAF2-directed activation of NFKB to prevent TNF-
induced apoptosis (Figure 35.3). It is plausible that
ASD-mediated pathway
this may be one of the mechanisms of TNF self-
tolerance. Because NF~:B and JNK are major factors N-SMase and A-SMase are activated independently
regulating the inflammatory response, this pathway by different cytoplasmic domains ofTNFR1, and elicit
may also play a role in the proinflammatory effect of discrete signaling pathways (Wiegmann et al., 1994).
TNE Like the death domain, the ASD can induce activation
of NFKB to protect against apoptosis (Van Antwerp
et al., 1996; Wang et al., 1996), but is also involved in
NSD- mediated pathway
induction of apoptosis (Pena et al., 1997). The ASD
NSD (neutral sphingomyelinase domain) occupies a maps to the same stretch of residues within the death
central role in mediating a number of TNF activities, domain, and may in fact be the same domain
including inflammatory responses and cell pro- (Tartaglia et al., 1993a; Wiegmann et al., 1994). The
liferation through the MAP kinase ERK and PIA2, ASD mediates activation of A-SMase via PC-PLC
respectively (Figure 35.3). The novel WD repeat ({X6_94- (Schutze et al., 1992). The connection between the
[GH-X23_41-WD]}N4_8) protein FAN couples TNFR1 to ASD in TNFR1 and PC-PLC is still obscure, but
N-SMase via protein-protein interactions between appears to involve the activity of protein kinase C

THE CYTOKINES AND CHEMOKINES


846 T U M O U R NECROSIS FACTOR

(PKC) (Mtiller et al., 1995). Activated PC-PCL pro- BIOLOGIC EFFECTS


duces 1,2-diacylglycerol (DAG), which activates A-
SMase and contributes to the activation of NFKB; TNF was originally characterized as a protein induc-
TNF-induced A-SMase and NFKB activity is blocked ing necrosis of methylcholanthrene (Meth A)-induced
by the specific PC-PLC inhibitor, D609, which effec- sarcomas in vivo (Carswell et al., 1975) and believed to
tively inhibits DAG production (Schutze et al., 1992). be a growth modulatory agent that exhibits primarily
A-SMase produces ceramide in the acidic endoso- antitumor activity. The discovery of the systemic
mal/lysosomal compartment (Spence, 1993). How- inflammatory role of TNF in non-malignant disease
ever, the precise role of A-SMase-induced ceramide (Tracey et al., 1986b) led to a focus on the pathogenic
is unclear, because cells that are deficient in role of TNF in many immune-mediated diseases. The
ceramidase, and therefore accumulate intracellular net effects of TNF are influenced by a complex array of
ceramide, are equally responsive to apoptotic signals cell- and tissue-specific factors. The diverse role of
from TNFR1. In addition, TNF- or CD95-induced TNF in mediating cellular responses is summarized in
apoptosis does not induce the degradation of labeled Table 35.2.
sphingomyelin (Segui et al., 2000). A-SMase activity in
TNF-induced NFKB may in fact be cell-type specific,
because a recent study by Kouba et al. (2001) revealed
Central nervous system
that A-SMase activity can be used as a surrogate Systemic TNF produces fever and anorexia via hypo-
mediator of NF~:B activation. thalamic centers that regulate body temperature and
appetite (Tracey et al., 1988; Plata-Salaman, 1991).
The anorectic effect of TNF is attenuated by insulin
TNFR2-mediated signaling pathway
and appears to be independent of serotonergic sig-
Direct signaling through TNFR2 is less well character- nals (Tracey and Cerami, 1990). Cells of the central
ized, and appears to be confined primarily to cells of nervous system (CNS), including microglia, astrocytes
the immune system. TNFR2 mediates specific TNF and neurons (Cheng et al., 1994), express TNF and
effects including proliferation of T and B cells high-affinity TNF receptors (Smith and Baglioni,
(Tartaglia et al., 1991; Barbara et al., 1994), induction 1992). Furthermore, elevated TNF levels have been
of NFKB and cytotoxicity (Hohmann et al., 1990a, detected in brain injury and certain CNS diseases
1990b; Grell et al., 1994), and may potentiate the including stroke (Liu et al., 1994), meningococcal
effects of TNFR1. TNFR2 can down-regulate human meningitis (Leist et al., 1988), human immuno-
and murine activated T cells by inducing apoptosis deficiency virus infection (Grimaldi et al., 1991),
(Zheng et al., 1995). TNFR2 lacks an intracellular Alzheimer's disease (Fillit et al., 1991) and multiple
death domain, suggesting that TNFR2 uses distinct sclerosis (Hofman et al., 1989). These findings
signaling pathway(s) to induce apoptosis. Down- strongly suggest that TNF is involved in various
regulation of the anti-apoptotic protein Bcl-xL (Boise biologic processes within the CNS.
and Thompson, 1997) has been correlated with The inflammatory effects of TNF are pivotal to the
TNFR2-induced apoptosis (Lin et al., 1997), but the development of cerebral inflammation and edema
precise mechanisms triggering TNFR2-induced apop- during meningitis. High levels of TNF in cerebrospinal
tosis remain to be elucidated. Whether TNFR2 alone fluid during meningitis are predictive of poor out-
can mediate the activation of NFKB is controversial. come (Leist et al., 1988; Waage et al., 1989; Saukkonen
Although TRAF2 interacts with TNFR2, and TRAF2- et al., 1990). The in vivo inflammatory effects of TNF
TRAF1 (Rothe et al., 1995) and TRAF2-TANK (Cheng have been implicated in the development of the
and Baltimore, 1996) heterocomplexes are able to plaques characteristic of multiple sclerosis, and in
mediate the activation of NFKB, the results of a recent cerebral ischemia in experimental models of stroke,
study (Chainy et al., 1996) indicate that TNFR2 does in which immunologic or pharmacologic inhibition of
not participate in the activation of NFKB in many cell TNF activity can reduce the severity of brain damage
types, including immune cells. (Saukkonen et al., 1990; Meistrell et al., 1997).

THE CYTOKINES A N D C H E M O K I N E S
BIOLOGIC EFFECTS 847

TABLE 35.2 Selected list of biological effects of TNF


Immune cells Non-immune cells In vivo
Mo nocytes/macro p hages Vascular endothelial cells Central nervous system
Autocrine induction of TNF Modulation of angiogenesis Fever
Induction of IL- 1, IL-8, Increasing permeability Anorexia
GM-CSF, M-CSE IFNT, Enhanced expression of MHC 1 Altered pituitary hormone
NGF, TGF[3, DGF and E G 2 Procoagulant and antifibrinolytic secretion
Chemotaxis Increasing permeability of
Stimulation of metabolism albumin Cardiovascular
Inhibition of differentiation Suppression of proliferation Shock
Suppression of proliferation Rearrangement of cytoskeleton ARDS
Internalization of Induction of NO synthase, IL-1, Capillary leakage syndrome
complement-coated virus IL-3 receptor, G-CSE GM-CSE
ICAM- 1, VCAM- 1, P- and E-selectin, Gastrointestinal
Polymorphonuclear leukocytes surface antigen, platelet-activating Ischemia
Priming of integrin response factor, prostacyclin Colitis
Release of granule Inhibition of integrin B30, Hepatic necrosis
components thromomodulin, glutathione, Inhibition of albumin
Increasing phagocytic protein S expression
capacity Decreased catalase activity
Enhanced production of Fibroblasts in liver
superoxide Induction of proliferation Suppression of HBV gene
Increased adherence to Induction of IL-1, 6, IFN~2, expression
extracellular matrix leukemia inhibitory factor,
Suppression of chemotaxis to metalloproteinases (MMTs) Metabolic
N- formyl- 1-1eucyl- 1-phenylalanine Suppression of respiratory activity LPL suppression
Suppression of cell surface Inhibition of collagen synthesis, Net protein catabolism
expression of sialophlorin CD43 MMT inhibitor Net lipid catabolism
Stress hormone release
Lymphocytes Adipocytes Insulin resistance
Induction ofT-cell colony Enhanced release of free fatty acids
formation and glycerol Inflammatory
Induction of superoxide in B cells Suppression of lipoprotein lipase Activation of cell cytotoxicity
Activation of cytotoxic T-cell (LPL) Enhanced NK cell function
invasiveness Mediation of IL-2 tumor
Induction of apoptosis in mature Endocrine system toxicity
T cells Stimulation of adrenocorticotrophic Protective role in cutaneous
hormone and prolactin leishmaniasis
Inhibition of thyroid-stimulating
hormone
Follicle-stimulating hormone and
growth hormone
Enhancing IL- 1 inhibition of
steroidogenesis

Although high levels of TNF play a pivotal role in proliferative effects of TNF may be dependent upon
tissue injury, accumulating evidence also indicates the production of nerve growth factor (NGF) (Gadient
that low levels of TNF play critical roles in the repair et al., 1990), which is protective in tissue damage
and regeneration of damaged tissue in the CNS (Mattson et al., 1993). Mice genetically deficient for
(Tracey et al., 1989). TNF prevents neuronal death both TNFR1 and TNFR2 are more sensitive to central
following metabolic excitotoxic insults (Cheng et al., ischemic insult and have more extensive cortical
1994), and activates CNS tissue repair by inducing damage (Bruce et al., 1996), suggesting that TNF can
proliferation of astrocytes (Selmaj et al., 1990) and be protective in ischemic brains. This contrasts with
neuronal progenitor cells (Mehler et al., 1993). The the overexpression of TNF during cerebral ischemia

THE CYTOKINES A N D C H E M O K I N E S
848 T U M O U R NECROSIS FACTOR

in normal rats subjected to focal infarction, where effects including a decrease in peripheral vascular
TNF significantly increases brain damage (Meistrell resistance, falling cardiac output and loss of intravas-
et al., 1997). cular volume through capillary leakage (Tracey et al.,
1986a, 1987b; Natanson et al., 1989). TNF-induced
NO production in endothelium has been implicated
Cardiovascular system
in decreases of both peripheral vascular tone and
Endothelial cells serve as a semipermeable barrier in cardiac function (Finkel et al., 1992), whereas
blood vessels, and mediate coagulation. During sys- TNF-induced rearrangement of actin filaments and
temic inflammation, sepsis, multiple organ failure and loss of tight junctions are responsible for the capillary
other acute TNF-mediated diseases, TNF enhances leakage syndrome (Sato et al., 1986; Stephens et al.,
tissue factor expression and suppresses co-factor 1988; Brett et al., 1989). TNF is extremely toxic to the
activity for the anticoagulant protein C on endothelial lungs and is pivotal in the development of adult respi-
cells (Bevilacqua et al., 1986; Nawroth et al., 1986). TNF ratory distress syndrome (ARDS), characterized by
also directly induces actin filament rearrangement, pulmonary edema, hypoxia and a high mortality rate.
leading to endothelial cell damage and loss of tight ARDS develops because of TNF-induced activation of
junctions; this, in turn, causes capillary leakage syn- pulmonary endothelium, margination of leukocytes
drome as plasma proteins and water leak into tissues with degranulation of granulocytes and capillary
(Sato et al., 1986; Stephens et al., 1988; Brett et al., leakage, which precipitates the collection of edema-
1989). TNF exerts a negative inotropic effect on tous fluid in alveoli and prevents adequate perfusion
myocardial contractility through a mechanism that (Stephens et al., 1988).
remains largely unknown. Evidence suggests that TNF
induces the release of platelet-activating factor (PAF)
Human diseases
(Alloatti et al., 1999), as well as stimulation of nitric
oxide (NO) production (Finkel et al., 1992; Habib et al., Although TNF can preserve cellular and biochemical
1996; Alloatti et al., 1999). Both epithelial cells and car- homeostasis thereby participating in beneficial tissue
diac myocytes express TNF receptors and are capable remodeling and host-defense responses, overproduc-
of synthesizing TNF under certain forms of stress tion of TNF is harmful, even lethal, to the host (Tracey
(Giroir et al., 1994; Kapadia et al., 1995). et al., 1989). Therefore, either insufficient or excessive
Elevated levels of TNF have been identified in production of TNF can be deleterious, and TNF has
patients with advanced heart failure (Moriarty et al., been directly implicated in the pathogenesis of many
1990; Katz et al., 1994). TNF may play a central role in disease states (Table 35.3).
the pathogenesis of heart failure, and the known bio-
logic effects of TNF may contribute to the develop-
Septic shock
ment of many of the clinical hallmarks of heart failure,
including left ventricular dysfunction, cardiomyopa- TNF plays a pivotal role in septic shock, because it ful-
thy, cachexia and pulmonary edema (Millar et al., fils Koch's postulates, a logical chain of evidence that
1989; Natanson et al., 1989; Suffredini et al., 1989; can be modified here for considering cytokine biol-
Hegewisch et al., 1990). The negative inotropic effects ogy. First, TNF is released systemically during over-
of TNF on isolated cardiac myocytes require TNF whelming sepsis, and in some cases leads to lethality
concentrations as low as 0.7-1.4 x 10-9 M (Yokoyama (Girardin et al., 1988; Waage et al., 1989). Persistent
et al., 1993; Kapadia et al., 1995), which have been increases in TNF are associated with the development
reported in congestive heart failure (Levine et al., of multiple organ failure and mortality (Girardin et al.,
1990). 1988; Calandra et al., 1990; van der Poll and Lowry,
Direct injection of TNF produces hypotension, 1995). Second, administration of exogenous TNF
metabolic acidosis, hemaconcentration and death causes shock and tissue injury that is physiologi-
within minutes, mimicking the cardiovascular cally, metabolically, hematologically and pathologi-
response in endotoxin-induced septic shock (Tracey cally indistinguishable from septic shock syndrome
et al., 1986a). This is caused by several TNF-mediated (Tracey et al., 1986a, 1987a). Third, neutralization of

THE CYTOKINES A N D C H E M O K I N E S
BIOLOGIC EFFECTS 849

TABLE 35.3 Selected TNF-associated diseases

Infectious diseases Autoimmune diseases Cancer and others


Bacterial Glomerulonephritis Hairy cell leukemia
Meningococcal disease Guillain-Barr6 syndrome T-cell leukemia
Leprosy Inflammatory bowel disease Chronic lymphocytic leukemia
Tuberculosis Systemic lupus erythematosus Malignant lymphoma
Septic shock syndrome Insulin-dependent diabetes Colorectal cancer
Nocardia brasiliensis mellitus Lung cancer
Cryptogenic fibrosing Rheumatoid arthritis Cervical cancer
alveolitis Dermatitis
Pneurnocystis carinii Celiac disease Euthyroid sick syndrome
pneumonia Myasthenia gravis Hemorrhagic shock
Systemic sclerosis Disseminated intravascular
Viral coagulopathy
AIDS Anemia
HIV-related lung disease Myocardial ischemia
Measles Obesity
Hepatitis Shock
Viral meningitis Stroke
Murine retrovirus infection Rejection of transplanted organs
Pulmonary fibrosis
Parasitic Chronic osteomyelitis
Cerebral malaria Graves' disease
Toxoplasma gondii Asthma
Dysentery Injury
Shigella dysenteriae Inflammatory hyperalgesia
Children with pertussis Down's syndrome
Cachexia

TNF with antibodies prevents septic shock syndrome be of predictive value for clinical outcome (Girardin
during lethal bacteremia (Tracey et al., 1987b). In sup- et al., 1992).
port of this view, TNFRl-deficient knockout mice are
resistant to endotoxemic shock (Pfeffer et al., 1993).
Cancer
TNF induces other mediators of sepsis such as IL-1,
and infusion of anti-TNF antibody attenuates the TNF was originally characterized as an antitumor pro-
activity of these mediators (Tracey et al., 1987b; Fong tein inducing necrosis of Meth A sarcomas in vivo
et al., 1989). (Carswell et al., 1975). TNF has been widely studied
TNF and soluble receptors ofTNF (sTNFRs) are lib- and explored as an antitumor drug; at present it is
erated during sepsis (Girardin et al., 1992). As men- used clinically as a locally delivered intratumoral
tioned above, sTNFRs are inhibitors of TNF biologic agent and has some efficacy (Lienard et al., 1992). The
activities but may also serve as a reservoir of TNF, and therapeutic application of TNF as a single and sys-
increased levels of sTNFRs are found in many TNF- temic antitumor agent is limited by toxic side-effects
associated diseases. In fact, increased levels of sTN- at tumoricidal doses (Blick et al., 1987). Several strate-
FRs correlate with TNF levels and outcome in severe gies have been proposed to avoid the limitations
meningococcemia (Girardin et al., 1992). In general, imposed by toxicity, including: (1) high-dose intra-
sTNFR concentrations are associated with TNF levels arterial administration of TNF directly into the tumor
when the TNF level is relatively low, but not when it is compartment (Ohkawa et al., 1989); (2) administra-
high. The imbalance between TNF and sTNFRs is tion of non-toxic levels of TNF to tumors sensitive to
probably pathophysiologically important, as reflected its therapeutic effects; (3) administration of non-toxic
by the observation that their ratio on admission may doses of TNF in combination with other antitumor

THE CYTOKINES AND CHEMOKINES


850 TUMOUR NECROSIS FACTOR

agents - synergistic effects have been identified regulation and the inflammatory response, and
(Zimmerman et al., 1989), but enhanced side-effects affects the outcome of human diseases, polymor-
have also been reported (Schiller etal., 1992); (4) iden- phisms within the TNF locus are considered possible
tification and development of TNF-like molecules genetic bases for these diseases. Some studies have
with maximum efficacy as an antitumor agent, but indicated that genetic variation of TNF and closely
minimal side-effects. In addition, chemical modifica- linked loci are important in determining suscepti-
tion of TNF with compounds such as polyethylene bility to a significant number of human diseases,
glycol (PEG) has been found selectively to increase the including autoimmune diseases such as diabetes,
antitumor potency of TNF while effectively reducing rheumatoid arthritis and multiple sclerosis, parasitic,
its systemic toxic side-effects (Tsutsumi et al., 1995). bacterial and viral infections and cancer.
A few mechanisms have been suggested for the TNF responsiveness is controlled by variable
tumoricidal effect of TNE including direct cytotoxicity genetic elements within the MHC region. A polymor-
against tumor cells, activation of immune antitumor phism located at -308 bp upstream of the TNF tran-
response and selective damage of tumor blood vessels scriptional start site, with two allelic forms referred to
(North and Havell, 1988). Direct cytotoxicity to tumor as TNF1 and TNF2, has been demonstrated directly
cells has been supported by most in vitro studies to affect TNF production (Wilson et al., 1993, 1997);
(Duerksen-Hughes et al., 1992). TNF also activates TNF2 is associated with higher constitutive and
other cytokines (Tracey and Cerami, 1994) and cyto- inducible transcriptional levels than the TNF1 allele
toxic factors such as NO (Estrada et al., 1992), which (Wilson et al., 1994, 1997). There is also a correlation
in turn participate in immune antitumor responses of human leukocyte antigen (HLA)-DR2 with low TNF
and direct tumor cell killing. A study by Sato et al. production (Bendtzen et al., 1988; Molvig et al., 1988),
(1996) reported that it might be feasible to use TNF and of HLA-DR3 and HLA-DR4 with high TNF pro-
gene-transduced tumor cells as a vaccine, since duction (Jacob et al., 1990; Abraham et al., 1993).
TNF is a powerful activator of the immune response. These polymorphisms are associated with the suscep-
TNF selectively eradicates vascularized tumors but is tibility or severity of certain diseases. Thus, the TNF
much less effective in killing avascular implants locus may contribute to the pathogenesis of diseases
(Yoshimura and Sone, 1987; Thomson, 1997). In con- by influencing the TNF levels produced in those
trast, TNF is produced by some tumors or tumor cells, diseases.
and can act as an autocrine growth factor (Sugarman
et al., 1985; Buck et al., 1990).
Susceptibility to infectious diseases
MHC alleles affect outcome in several infectious
ASSOCIATED DISEASE diseases. TNF2 homozygotes with associated higher
SUSCEPTIBILITY TNF levels are at increased risk of cerebral malaria
(McGuire et al., 1994) or post-traumatic sepsis
The TNF locus with two tandemly arranged and (Majetschak et al., 1999). This is consistent with the
closely linked genes, TNF and lymphotoxin-tz, lies in phenotypic observation that Gambian children with
the class III region of the MHC on the short arm of cerebral malaria have higher TNF levels than children
human chromosome 6. A striking feature of the MHC with mild malaria (Kwiatkowski et al., 1990), and that
is the high degree of polymorphism of genes in this TNF levels are highest in children who die or develop
region. At least five polymorphisms (Partanen and neurologic sequelae due to cerebral malaria. In
Koskimies, 1988; D'Alfonso and Richiardi, 1994) and contrast, the HLA-B53 locus, which correlates with
five microsatellites (Jongeneel et al., 1991; Udalova depressed levels of circulating TNF, appears to be pro-
et al., 1993) have been described at the TNF locus. tective against several forms of malaria (Hill, 1992).
These polymorphisms have been linked to the vari- The TNFB2 allele, also associated with higher levels of
ability of TNF production in different individuals TNF production, is linked to lower survival in patients
(Molvig et al., 1988). Because TNF production has with multiple organ failure secondary to severe sepsis
been implicated as an important factor in immune (Stuber et al., 1996). Susceptibility to mucotaneous

THE CYTOKINES AND CHEMOKINES


ASSOCIATED DISEASE SUSCEPTIBILITY 85 1

leishmaniasis, which is accompanied by high circulat- disease is strongly influenced by the genes in MHC,
ing TNF levels, is linked directly with the regulatory and is associated with the HLA-A1-B8-DR3-DQ2
polymorphisms affecting TNF production (Cabrera haplotype (Ahmed et al., 1993) and more strongly
et al., 1995). In addition, HLA-DQ and HLA-DP alleles associated with the DQA*0501/DQB*0201 het-
are associated with generalized and localized forms of erodimer (Sollid et al., 1989). Wilson and colleagues
onchocerciasis (Meyer et al., 1994). As the under- (1993) found that the TNF2 allele in the TNF locus is
standing of allelic regulatory differences improves, it closely associated with HLA-A1-B8-DR3-DQ2, sug-
should be possible to more effectively translate our gesting that the association of this haplotype with
basic research advances in TNF mechanisms into celiac disease and high TNF production may be
clinically useful methods to prevent the pathologic related to polymorphisms within the TNF locus. At the
sequelae of excessive TNF, and/or to improve the same time, susceptibility to celiac disease is strongly
potential benefits of its controlled local production related to TNF2 (Manus et al., 1996), supporting this
(e.g. cancer). speculation. The TNF2 allele appears to be involved
in a variety of MHC-linked diseases including sys-
temic lupus erythematosus, dermatitis herpetiformis
Susceptibility to cancer and insulin-dependent diabetes mellitus (IDDM).
Alleles of HLA-A, -B and -DR loci are associated with Microsatellite polymorphisms (TNFa2 and TNFb3)
resistance to lung cancer. Investigators have located close to the TNF genes are associated with celiac dis-
one such element to the TNF locus and found that ease, an observation that could have functional signif-
TNFB2 homozygosity is associated with disease icance because the TNFa2 allele correlates with high
resistance and a better prognosis in patients with lung TNF production (McManus et al., 1996).
cancer (Shimura et al., 1994), in keeping with a previ- Rheumatoid arthritis (RA) is a debilitating disease
ous study that demonstrated a strong correlation characterized by progressive joint dysfunction and
between HLA-A1 and a lower i-year survival rate chronic systemic inflammation (Pincus et al., 1994).
(Markman et al., 1984). As TNFB2 is associated with TNF has been implicated in the pathogenesis of RA,
higher levels of TNF production, these results are con- although its precise mechanisms of action in the dis-
sistent with the original characterization of TNF as an ease remain poorly understood. Genetic analyses
antitumor factor. It has been reported that germline have implicated the TNFcl allele as well as the -238
allelic frequencies for the TNF locus are significantly GG genotype, but not the TNFa allele, in rheumatoid
different in patients with colorectal cancer compared arthritis (RA) susceptibility (Fabris et al., 2002). Ele-
with those in normal controls. Most strikingly, the R3 vated concentrations of TNF have been identified in
allele in patients with colorectal cancer has an allelic the serum and synovial fluid of RA patients (Chu et al.,
frequency of nearly 30% but is not detected in the 1991), and plasma TNF concentrations correlate with
normal control group (Campbell et al., 1994). Other disease severity in terms of joint function and pain
studies have shown that class II haplotypes such as (Beckham et al., 1992). Because TNF induces the
DRB 1"1501-DQB 1"0602 are positively associated with expression of endothelial cell adhesion molecules
the development of cervical cancer, but that DR13- (Moser et al., 1989), TNF may recruit inflammatory
positive haplotypes are negatively associated (Apple cells into affected joints. TNF also induces the expres-
et al., 1994). sion of matrix metalloproteinases from fibroblasts
and chondrocytes, and may therefore accelerate joint
degradation and dysfunction in RA (Shingu et al.,
Susceptibility to a u t o i m m u n e diseases 1993). Therapeutic interventions that inhibit the
The outcome of a large number of studies suggests activity of TNF have proven efficacious for the treat-
that TNF genetics is an important contributing factor ment of both adult and juvenile RA (Maini et al., 1999;
to autoimmune disease susceptibility. Celiac disease Weinblatt et al., 1999; Lovell et al., 2000).
(CD) is an immune disease triggered by the cereal The role of various TNF alleles as susceptibility fac-
antigen gliadin, resulting in villus atrophy in the small tors for systemic lupus erythematosus (SLE) is cur-
intestine. Susceptibility to the development of celiac rently under intense investigation. The alleles TNFB 1

THE CYTOKINES AND CHEMOKINES


852 T U M O O R NECROSIS FACTOR

(Bettinotti et al., 1993) and TNF2 (Wilson et al., 1993) 1987b; Ashkenazi et al., 1991; Peppel et al., 1991).
are associated with systemic lupus erythematosus Another is to suppress TNF synthesis using glucocor-
(SLE), but the association is not independent of ticoids, pentoxifylline, CNI-1493, spermine, thalido-
HLA-DR3 (Welch et al., 1988). The HLA-DR3 and mide and cytokines such as IL-10 (Beutler et al., 1986;
TNF-308A loci appear to be independent susceptibil- Lilly et al., 1989; Sampaio et al., 1991; Cohen et al.,
ity loci for SLE (Rood et al., 2000; van der Linden et al., 1996; Zhang et al., 1997). The third approach is to
2001), but others challenge this view (Tsuchiya et al., induce TNF tolerance with repetitive administration
2001). In another study (D'Alfonso et al., 1996), 123 of low-dose TNE Inhibitors of receptor-mediated
patients with SLE and 199 matched controls were endocytosis of TNF decrease TNF-induced gene
analyzed. Three TNF-238/A homozygous patients expression, thus providing yet another potential point
but no homozygous control were detected, suggesting of intervention (Bradley et al., 1993). In addition,
that the -238/AA genotype is a marker of a particular pharmacologic inhibitors of metalloproteinases have
clinical subtype; this has recently been confirmed in been developed to inhibit TNF processing, although
a cohort of 51 Mexican SLE patients (Zuniga et al., clinical efficacy has not been addressed (McGeehan
2001). Because TNF has been genetically implicated et al., 1994). Finally, TNF cytotoxicity may be inhibited
as a mediator of SLE, several studies have attempted by targeting its secondary mediators such as IL-1
to neutralize TNF as a therapeutic approach to SLE. (McNamara et al., 1993). Some of these therapies have
Although immunologic or pharmacologic inhibition been demonstrated to have salutory effects in animal
of TNF production has reduced the clinical manifesta- models of sepsis, yet preliminary clinical trials have
tions of experimental SLE in mice (Segal et al., 2001), not been as encouraging (van der Poll and Lowry,
aberrant TNF activity may promote humoral auto- 1995).
immunity and allow the development of autoreactive Strategies to directly inhibit TNF activity with anti-
B cells (Via et al., 2001). These findings suggest that, bodies or sTNFRs are effective in animal models
while abrogation of TNF activity may be therapeuti- (Mohler et al., 1993) (Table 35.2), and have been
cally beneficial for the treatment of established SLE, tested in clinical trials of Crohn's diseases (Sandborn
depressed TNF levels may play a causative role in the and Hanauer, 1999; Shanahan, 2000; Kam and Targan,
development of this autoimmune disease. 2000), rheumatoid arthritis (Taylor, 2001; Schwarz
TNF has also been implicated in the pathogenesis et al., 2000; Feldmann and Maini, 2001; Alldred, 2001),
of IDDM as a mediator in the immune-mediated and sepsis (Dhainaut et al., 1995; Cronin et al., 1995;
destruction of pancreatic [3 cells (Mandrup-Poulsen Abraham et al., 1997, 1998). For instance, a chimeric
et al., 1987). Studies of TNF genetics in patients with monoclonal anti-TNF antibody, infliximab, has
IDDM show a weak association between TNF2 and shown efficacy in moderately to severely active
IDDM (Pociot et al., 1993b; Cox et al., 1994). However, Crohn's disease, and has recently been approved
another study demonstrated a higher frequency of the by the U.S. Food and Drug Administration (FDA)
TNFa2 allele in DR3-DR4 heterozygotic patients (Sandborn and Hanauer, 1999). A recombinant
with IDDM than in controls. As this marker is associ- human soluble TNF receptor, Etanercept (Enbrel), has
ated with higher TNF production independently of recently been approved by the U.S. FDA for the
class II alleles, the TNF locus may play a direct role in treatment of rheumatoid arthritis, which has shown
susceptibility to IDDM (Pociot et al., 1993a). a statistically significant reduction in swollen and
inflamed joint counts, and a significant improve-
ments in quality of life measures (Schwarz et al., 2000;
Anti-TNF therapy Taylor, 2001; Feldmann and Maini, 2001; Alldred,
Several strategies have been proposed to prevent TNF 2001). Although anti-TNF therapy has been success-
toxicity in sepsis (Tracey and Cerami, 1994). One fully employed in the treatment of rheumatoid arthri-
approach is direct inhibition of TNF activity with anti- tis, a significant survival advantage has not been
bodies or native and chimeric sTNFRs (Tracey et al., observed in the treatment of sepsis.

THE CYTOKINES A N D C H E M O K I N E S
REFERENCES 853

Regulation of TNF by the cholinergic Alessi, D.R., Cuenda, A., Cohen, P. et al. (1995). PD 098059 is
a specific inhibitor of the activation of mitogen-activated
anti-inflammatory pathway protein kinase kinase in vitro and in vivo. ]. Biol. Chem.
270, 27489-27494.
The cholinergic a n t i - i n f l a m m a t o r y p a t h w a y is a neu- Alldred, A. (2001). Etanercept in rheumatoid arthritis. Exp.
ral p a t h w a y t h a t rapidly a n d directly a t t e n u a t e s the Opin. Pharmacother. 2, 1137-1148.
p r o d u c t i o n of TNE In r e s p o n s e to e n d o t o x i n or Alloatti, G., Penna, C., De Martino, A. et al. (1999). Role of
nitric oxide and platelet-activating factor in cardiac alter-
i s c h e m i a / reperfusion injury, efferent p a r a s y m p a - ations induced by tumor necrosis factor-alpha in the
thetic signals t h r o u g h the vagus nerve increase h e a r t guinea-pig papillary muscle. Cardiovasc. Res. 41, 611-619.
rate, stabilize b l o o d pressure, a n d a t t e n u a t e s e r u m An, J., Ribeiro, R.C., Webb, P. et al. (1999). Estradiol repression
of tumor necrosis factor-alpha transcription requires
a n d tissue TNF levels (Borovikova et al., 2000a, 2000b; estrogen receptor activation function-2 and is enhanced
Bernik e t al., 2001, 2002). Acetylcholine, the p r i m a r y by coactivators. Proc. N a t l Acad. Sci. USA 96,
n e u r o t r a n s m i t t e r of the vagus nerve, b i n d s alpha- 15161-15166.
Apple, R.J., Erlich, H.A., Klitz, W. et al. (1994). HLA DR-DQ
b u n g a r o t o x i n - s e n s i t i v e nicotinic cholinergic recep- associations with cervical carcinoma show papillo-
tors e x p r e s s e d on m a c r o p h a g e s to inhibit the mavirus-type specificity. Nat. Genet. 6, 157-162.
p r o d u c t i o n of p r o i n f l a m m a t o r y mediators, i n c l u d i n g Ashkenazi, A., Marsters, S.A., Capon, D.J. et al. (1991).
Protection against endotoxic shock by a tumor necrosis
TNF a n d IL-1 (Borovikova et al., 2000a, 2000b). Elec- factor receptor immunoadhesin. Proc. Natl Acad. Sci. USA
trical s t i m u l a t i o n of the vagus nerve can r e c a p i t u l a t e 88, 10535-10539.
or a u g m e n t the i n n a t e central r e s p o n s e to p e r i p h e r a l Auphan, N., Di Donato, J.A., Rosette, C. et al. (1995). Immuno-
suppression by glucocorticoids: inhibition of NFKB activ-
i n f l a m m a t i o n (Borovikova et al., 2000a, 2000b; Bernik ity through induction of I~:-B synthesis. Science 270,
et al., 2001, 2002); b e c a u s e vagus nerve s t i m u l a t o r s 286-290.
are u s e d clinically for the t r e a t m e n t of d e p r e s s i o n a n d Balboa, M.A., Balsinde, J., Aramburu, J. et al. (1992).
Phospholipase D activation in human natural killer cells
epilepsy (Goodnick et al., 2001; B e n b a d i s a n d Tatum, through the KP43 and CD16 surface antigens takes place
2001), vagus nerve s t i m u l a t i o n m a y provide a n e w by different mechanisms. Involvement of the phospho-
a p p r o a c h for the t r e a t m e n t of i n f l a m m a t o r y diseases, lipase D pathway in tumor necrosis factor-~ synthesis.
J. Exp. Med. 176, 9-17.
b e c a u s e it inhibits TNF a n d o t h e r p r o i n f l a m m a t o r y Baldwin, R.L, Stolowitz, M.L, Hood, L. et al. (1996). Structural
mediators. changes of tumor necrosis factor ~ associated with mem-
brane insertion and channel formation. Proc. Natl Acad.
Sci. USA 93, 1021-1026.
Barbara, J.A., Smith, W.B., Gamble, J.R. et al. (1994). Dissocia-
tion of TNF-R cytotoxic and proinflammatory activities
REFERENCES by p55 receptor- and p75 receptor-selective TNF-~
mutants. EMBO ]. 13, 843-650.
Abraham, E., Glauser, M.P., Butler, T. et al. (1997). p55 Tumor Beckham, J.C., Caldwell, D.S., Peterson, B.L. et al. (1992).
necrosis factor receptor fusion protein in the treatment Disease severity in rheumatoid arthritis: relationships of
of patients with severe sepsis and septic shock. A ran- plasma tumor necrosis factor-alpha, soluble interleukin
domized controlled multicenter trial. Ro 45-2081 Study 2-receptor, soluble CD4/CD8 ratio, neopterin, and fibrin
Group. ]AMA 277, 1531-1538. D-dimer to traditional severity and functional measures.
Abraham, E., Anzueto, A., Gutierrez, G. et al. (1998). Double- ]. Clin. I m m u n o l . 12, 353-361.
blind randomised controlled trial of monoclonal anti- Benbadis, S.R. and Tatum, W.O. IV. (2001). Advances in the
body to human tumour necrosis factor in treatment of treatment of epilepsy. Am. Faro. Physician 64, 91-98.
septic shock. NORASEPT II Study Group. Lancet 351, Bendtzen, K., Morling, N., Fomsgaard, A. et al. (1988).
929-933. Association between HLA-DR2 and production of
Abraham, L.J., French, M.A. and Dawkins, R.L (1993). tumour necrosis factor (~ and interleukin 1 by mono-
Polymorphic MHC ancestral haplotypes affect the activ- nuclear cells activated by lipopolysaccharide. Scand. J.
ity of tumour necrosis factor-a. Clin. Exp. I m m u n o l . 92, I m m u n o l . 28, 599-606.
14-18. Bernik, T.R., Ivanova, S.M., Ochani M. et al. (2001). Vagus
Adam-Klages, S., Adam, D., Wiegmann, K. et al. (1996). FAN, nerve stimulation attenuates cardiac TNF production in
a novel WD-repeat protein, couples the p55 TNF-receptor endotoxic shock. Shock 15 (Supp.), 27.
to neutral sphingomyelinase. Cell 86, 937-947. Bernik, T.R., DiRaimo, R., Friedman, S.G. et al. (2002).
Aderka, D. (1996). The potential biological and clinical sig- Systemic anti-inflammatory action of CNI-1493 is
nificance of the soluble tumor necrosis factor receptors. mediated by the cholinergic anti-inflammatory pathway.
Cytokine Growth Factor Rev. 7, 231-240. ]. Exp. Med., 18, 781-788.
Ahmed, A.R., Yunis, J.J., Marcus-Bagley, D. et al. (1993). Major Bettinotti, M.P., Hartung, K., Deicher. H. et al. (1993).
histocompatibility complex susceptibility genes for der- Polymorphism of the tumor necrosis factor [3gene in sys-
matitis herpetiformis compared with those for gluten- temic lupus erythematosus: TNFB-MHC haplotypes.
sensitive enteropathy. ]. Exp. Med. 178, 2067-2075. I m m u n o g e n e t i c s 37, 449-454.

THE CYTOKINES AND CHEMOKINES


854 TUMOUR NECROSIS FACTOR

Beutler, B., Greenwald, D., Hulmes, J.D. et al. (1985). Identity Calandra, T., Baumgartner, J.D., Grau, G.E. et al. (1990).
of tumour necrosis factor and the macrophage-secreted Prognostic values of tumor necrosis factor/cachectin,
factor cachectin. Nature 316, 552-554. interleukin-1, interferon-a and interferon-7 in the serum
Beutler, B., Krochin, N., Milsark, I.W. et al. (1986). Control of of patients with septic shock. Swiss-Dutch J5 immuno-
cachectin (tumor necrosis factor) synthesis: mechanisms globulin study group. J. Infect. Dis. 161,982-987.
of endotoxin resistance. Science 232, 977-980. Campbell, D.A., Field, M., McArdle, C.S. et al. (1994).
Bevilacqua, M.P., Pober, J.S., Majeau, G.R. et al. (1986). Polymorphism at the tumour necrosis factor locus: a
Recombinant tumor necrosis factor induces procoagu- marker of genetic predisposition to colorectal cancer?
lant activity in cultured human vascular endothelium: Lancet 343, 293-294 (letter).
characterization and comparison with the actions of Caput, D., Beufler, B., Hartog, K. et al. (1986). Identification
interleukin 1. Proc. Natl Acad. Sci. USA 83, 4533-4537. of a common nucleotide sequence in the 3'-untranslated
Bianchi, M., Bloom. 0., Raabe, T. et al. (1996). Suppression of region of mRNA molecules specifying inflammatory
proinflammatory cytokines in monocytes by a tetravalent mediators. Proc. Natl Acad. Sci. USA. 83, 1670-1674.
guanylhydrazone. L Exp. Med. 183, 927-936. Carswell, E.A., Old, L.J., Kassel, R.L. et al. (1975). An endo-
Biswas, D.K., Ahlers, C.M., Dezube, B.J. et al. (1994). toxin-induced serum factor that causes necrosis of
Pentoxifylline and other protein kinase C inhibitors tumors. Proc. Natl Acad. Sci. USA 72, 3666-3670.
down-regulate HIV-LTR NF-•B-induced gene expression. Cha, S.S., Kim, J.S., Cho, H.S. et al. (1998). High resolution crys-
Mol. Med. 1, 31-43. tal structure of a human tumor necrosis factor-a mutant
Black, R.A., Rauch, C.T., Kozlosky, C.J. et al. (1997). A metal- with low systemic toxicity. ]. Biol. Chem. 273, 2153-2160.
loproteinase disintegrin that releases tumour-necrosis Chainy, G.B., Singh, S., Raju, U. et al. (1996). Differential acti-
factor-a from cells. Nature 385, 729-733. vation of the nuclear factor-~:B by TNF muteins specific
Blick, M., Sherwin, S.A., Rosenblum, M. et al. (1987). Phase I for the p60 and p80 TNF receptors. ]. I m m u n o l . 157,
study of recombinant tumor necrosis factor in cancer 2410-2417.
patients. Cancer Res. 47, 2986-2989. Cheng, B., Christakos. S. and Mattson, M.P. (1994). Tumor
Bogdan, C., Paik, J., Vodovotz, Y. et al. (1992). Contrasting necrosis factors protect neurons against metabolic-
mechanisms for suppression of macrophage cytokine excitotoxic insults and promote maintenance of calcium
release by transforming growth factor-~. ]. Biol. Chem. homeostasis. N e u r o n 12, 139-153.
267, 23301-23308. Cheng, G. and Baltimore, D. (1996). Tank, a co-inducer with
Boise, LH. and Thompson, C.B. (1997). Bcl-x(L) can inhibit TRAF2 of TNF- and CD 401-mediated NF~:B activation.
apoptosis in cells that have undergone Fas-induced pro- Genes Dev. 10, 963-973.
tease activation. Proc. Natl Acad. Sci. USA 94, 3759-3764. Chikanza, I.C., Roux-Lombard, P., Dayer, J.M. et al. (1993).
Boldin, M.E, Goncharov, T.M., Goltsev, Y.V. et al. (1996). Tumour necrosis factor soluble receptors behave as acute
Involvement of mach, a novel MORT1/FADD-interacting phase reactants following surgery in patients with
protease, in Fas/Apo-1- and TNF receptor-induced cell rheumatoid arthritis, chronic osteomyelitis and
death. Cell 85, 803-815. osteoarthritis. Clin. Exp. I m m u n o l . 92, 19-22.
Borovikova, L.V., Ivanova, S., Nardi, D. et al. (2000a). Role of Chinnaiyan, A.M., O'Rourke, K., Tewari, M. et al. (1995).
vagus nerve signaling in CNI- 1493-mediated suppression FADD, a novel death domain-containing protein, inter-
of acute inflammation. Auton. Neurosci. 85, 141-147. acts with the death domain of Fas and initiates apoptosis.
Borovikova, L.V., Ivanova, S., Zhang, M. et al. (2000b). Vagus Cell 81,505-512.
nerve stimulation attenuates the systemic inflammatory Chu, C.Q., Field, M., Feldman, M. et al. (1991). Localization
response to endotoxin. Nature 405, 458-462. of tumor necrosis factor-a in synovial tissues and at the
Bradley, J.R., Johnson, D.R. and Pober, J.S. (1993). Four differ- cartilage-pannus junction in patients with rheumatoid
ent classes ofinhibitors of receptor-mediated endocytosis arthritis. Arthritis R h e u m . 34, 1125-1132.
decrease tumor necrosis factor-induced gene expression Coeshott, C., Ohnemus, C., Pilyavskaya, A. et al. (1999).
in human endothelial cells. ]. I m m u n o l . 150, 5544-5555. Converting enzyme-independent release of tumor necro-
Brett, J., Gerlach, H., Nawroth, P. et al. (1989). Tumor necro- sis factor alpha and IL-lbeta from a stimulated human
sis factor/cachectin increases permeability of endothelial monocytic cell line in the presence of activated neu-
cell monolayers by a mechanism involving regulatory trophils or purified proteinase 3. Proc. NatlAcad. Sci. USA
G proteins. ]. Exp. Med. 169, 1977-1991. 96, 6261-6266.
Brockhaus, M., Schoenfeld. H.J., Schlaeger, E.J. et al. (1990). Cohen, O., Inbal, B., Kissil, J.L. et al. (1999). DAP-kinase par-
Identification of two types of tumor necrosis factor ticipates in TNF-alpha- and Fas-induced apoptosis and
receptors on human cell lines by monoclonal antibodies. its function requires the death domain. ]. Cell Biol. 146,
Proc. Natl Acad. Sci. USA 87.3127-3131. 141-148.
Bruce, A.]., Boling, W., Kindy, M.S. et al. (1996). Altered neu- Cohen, P.S., Nakshatri. H., Dennis, J. et al. (1996). CNI-1493
ronal and microglial responses to excitotoxic and inhibits monocyte/macrophage tumor necrosis factor by
ischemic brain injury in mice lacking TNF receptors. Nat. suppression of translation efficiency. Proc. Natl Acad. Sci.
Med. 2, 788-794. USA. 93, 3967-3971.
Buck, C., Digel, W., Schoniger, W. et al. (1990). Tumor necro- Condorelli, G., Vigliotta, G., Cafieri, A. et al. (1999). PED/
sis factor-a, but not lymphotoxin, stimulates growth of PEA-15: an anti-apoptotic molecule that regulates
tumor cells in hairy cell leukemia. L e u k e m i a 4, 431-434. FAS/TNFRl-induced apoptosis. Oncogene 18, 4409-4415.
Cabrera, M., Shaw, M.A., Sharpies, C. et al. (1995). Poly- Cox, A., Gonzalez, A.M., Wilson, A.G. et al. (1994). Compara-
morphism in tumor necrosis factor genes associated tive analysis of the genetic associations of HLA-DR3
with mucocutaneous leishmaniasis. 1. Exp. Med. 182, and tumour necrosis factor a with h u m a n IDDM.
1259-1264. Diabetologia 37, 500-503.

THE CYTOKINES AND CHEMOKINES


REFERENCES 855

Crawford, E.K., Ensor, J.E., Kalvakolanu, I. et al. (1997). The Extracellular signal-related kinase (ERK) and p38
role of 3' poly(A) tail metabolism in tumor necrosis mitogen-activated protein (MAP) kinases differentially
factor-alpha regulation. 1. Biol. Chem. 272, 21120-21127. regulate the lipopolysaccharide-mediated induction of
Creasey, AA., Doyle, L.V., Reynolds, M.T. et al. (1987). inducible nitric oxide synthase and IL- 12 in macrophages:
Biological effects of recombinant human tumor necrosis Leishmania phosphoglycans subvert macrophage IL-12
factor and its novel muteins on tumor and normal cell production by targeting ERK MAP kinase. ]. I m m u n o l . 163,
lines. Cancer Res. 47, 145-149. 6403-6412.
Cronin, L., Cook, D.J., Carlet, J. et al. (1995). Corticosteroid Fillit, H., Ding, W.H., Buee, L. et al. (1991). Elevated circulat-
treatment for sepsis: a critical appraisal and meta- ing tumor necrosis factor levels in Alzheimer's disease.
analysis of the literature. Crit. Care Med. 23, 1430-1439. Neurosci. Lett. 129, 318-320.
Crowe, P.D., Walter, B.X., Mohler, K.M. et al. (1995). A metal- Finkel, M.S., Oddis, C.V., Jacob, T.D. et al. (1992). Negative
loprotease inhibitor blocks shedding of the 80-kD TNF inotropic effects of cytokines on the heart mediated by
receptor and TNF processing in T lymphocytes. I. Exp. nitric oxide. Science 257, 387-389.
Med. 181, 1205-1210. Fong, C.W., Siddiqui, A.H. and Mark, D.E (1995). Characteri-
Cuenda, A., Rouse, J., Doza, Y.N. et al. (1995). SB 203580 is a zation of protein complexes formed on the repressor
specific inhibitor of a MAP kinase homologue which is elements of the human tumor necrosis factor alpha gene.
stimulated by cellular stresses and interleukin-1. FEBS ]. Interferon Cytokine Res. 15, 887-895.
Lett. 364, 229-233. Fong, Y., Tracey. K.J., Moldawer, L.L. et al. (1989). Antibodies
D'Alfonso, S. and Richiardi, RM. (1994). A polymorphic vari- to cachectin/tumor necrosis factor reduce interleukin 113
ation in a putative regulation box of the TNFA promoter and interleukin 6 appearance during lethal bacteremia.
region. Immunogenetics 39, 150-154. ]. Exp. Med. 170, 1627-1633.
D'Alfonso, S., Colombo, G., Della Bella, S. et al. (1996). Gadient, R.A., Cron, K.C. and Otten, U. (1990). Interleukin- 113
Association between polymorphisms in the TNF region and tumor necrosis factor-a synergistically stimulate
and systemic lupus erythematosus in the Italian popula- nerve growth factor (NGF) release from cultured rat
tion. Tissue Antigens 47, 551-555. astrocytes. Neurosci. Lett. 117, 335-340.
Dean, J.L., Wait, R., Mahtani, K.R. et al. (2001). The 3' untrans- Gatanaga, T., Hwang, C.D., Gatanaga, M. et al. (1991). The
lated region of tumor necrosis factor alpha mRNA is a tar- regulation of TNF receptor mRNA synthesis, membrane
get of the mRNA-stabilizing factor HuR. Mol. Cell Biol. 21, expression, and release by PMA- and LPS-stimulated
721-730. human monocytic THP-1 cells in vitro. Cell I m m u n o l .
Dekkers, RE., Lauw, F.N., ten Hove, T. et al. (1999). The effect 138, 1-10.
of a metalloproteinase inhibitor (GI5402) on tumor Gearing, A.J., Beckett, P., Christodoulou, M. et al. (1994).
necrosis factor-alpha (TNF-alpha) and TNF-alpha recep- Processing of tumour necrosis factor-a precursor by
tors during human endotoxemia. Blood 94, 2252-2258. metalloproteinases. Nature 370, 555-557.
Denham, W., Yang, J., Wang, H. et al. (2000). Inhibition of p38 Gearing. A.J., Beckett, R, Christodoulou, M. et al. (1995).
mitogen activated kinase attenuates the severity of pan- Matrix metalloproteinases and processing of pro-TNF-a.
creatitis-induced adult respiratory distress syndrome. ]. Leukoc. Biol. 57, 774-777.
Crit. Care Med. 28, 2567-2572. Girardin, E., Grau, G.E., Dayer, J.M. et al. (1988). Tumor
Dhainaut, J.E, Vincent, J.L., Richard, C. et al. (1995). CDP571, necrosis factor and interleuldn-1 in the serum of children
a humanized antibody to human tumor necrosis factor- with severe infectious purpura. N. Engl. ]. Med. 319,
alpha: safety, pharmacokinetics, immune response, and 397-400.
influence of the antibody on cytoldne concentrations in Girardin, E., Roux-Lombard, R, Grau, G.E. et al. (1992).
patients with septic shock. CPD571 Sepsis Study Group. Imbalance between tumour necrosis factor-a and soluble
Crit. Care Med. 23, 1461-1469. TNF receptor concentrations in severe meningococ-
Di Marco, S., Hel, Z., Lachance, C. et al. (2001). Polymorphism caemia. The J5 Study Group. I m m u n o l o g y 76, 20-23.
in the 3'-untranslated region ofTNFalpha mRNA impairs Giroir, B.R, Horton, J.W.,White, D.J. et al. (1994). Inhibition of
binding of the post-transcriptional regulatory protein tumor necrosis factor prevents myocardial dysfunction
HuR to TNFalpha mRNA. Nucleic Acids Res. 29, 863-871. during burn shock. Am. ]. Physiol. 267, H118-H124.
Duan, H. and Dixit, V.M. (1997). RAIDD is a new 'death' adap- Goldfeld, A.E., McCaffrey, RG., Strominger, J.L. et al. (1993).
tor molecule. Nature 385, 86-89. Identification of a novel cyclosporin-sensitive element in
Duerksen-Hughes, RJ., Day, D.B., Laster, S.M. et al. (1992). the human tumor necrosis factor ~ gene promoter. ]. Exp.
Both tumor necrosis factor and nitric oxide participate in Med. 178, 1365-1379.
lysis of simian virus 40-transformed cells by activated Goodnick, RJ., Rush, A.J., George, M.S. et al. (2001). Vagus
macrophages. ]. I m m u n o l . 149, 2114-2122. nerve stimulation in depression. Expert Opin. Pharmaco-
Estrada, C., Gomez, C., Martin, C. et al. (1992). Nitric oxide ther. 2, 1061-1063.
mediates tumor necrosis factor-a cytotoxicity in endo- Grell, M., Douni, E., Wajant, H. et al. (1995). The transmem-
thelial cells. Biochem.Biophys.Res. C o m m u n . 186,475-482. brane form of tumor necrosis factor is the prime activat-
Fabris, M., Di, RE., D'Elia, A. et al. (2002). Tumor necrosis ing ligand of the 80 kDa tumor necrosis factor receptor.
factor-alpha gene polymorphism in severe and mild- Cell 83, 793-802.
moderate rheumatoid arthritis. ]. R h e u m a t o l . 29, Grimaldi, L.M., Martino, G.V., Franciotta, D.M. et al. (1991).
29-33. Elevated a-tumor necrosis factor levels in spinal fluid
Feldmann, M. and Maini, R.N. (2001). Anti-TNF alpha ther- from HW-l-infected patients with central nervous sys-
apy of rheumatoid arthritis: what have we learned? Annu. tem involvement. Ann. Neurol. 29, 21-25.
Rev. I m m u n o l . 19, 163-196. Grimm, S., Stanger, B.Z. and Leder, P. (1996). RIP and FADD:
Feng, G.J., Goodridge, H.S., Harnett, M.M. et al. (1999). two 'death domain'-containing proteins can induce

THE CYTOKINES AND CHEMOKINES


856 TUMOUR NECROSIS FACTOR

apoptosis by convergent, but dissociable, pathways. Proc. Relation to nitric oxide-dependent vasodilation in the
Natl Acad. Sci. USA 93, 10923-10927. forearm circulation. Circulation 90, 12-16.
Gueydan, C., Droogmans, L., Chalon, P. et al. (1999). Kohno, T., Brewer, M.T., Baker, S.L. et al. (1990). A second
Identification of TIAR as a protein binding to the transla- tumor necrosis factor receptor gene product can shed a
tional regulatory AU-rich element of tumor necrosis fac- naturally occurring tumor necrosis factor inhibitor. Proc.
tor alpha mRNA. J. Biol. Chem. 274, 2322-2326. Natl Acad. Sci. USA 87, 8331-8335.
Guha, M., O'Connell, M.A., Pawlinski, R. et al. (2001). Kouba, D.J., Nakano, H., Nishiyama, T. et al. (2001). Tumor
Lipopolysaccharide activation of the MEK-ERK1/2 path- necrosis factor-a induces distinctive NF-KB signaling
way in human monocytic cells mediates tissue factor within human dermal fibroblasts. ]. Biol. Chem. 276,
and tumor necrosis factor alpha expression by inducing 6214-6224.
Elk-1 phosphorylation and Egr-1 expression. Blood 98, Kramer, B., Meichle, A., Hensel, G. et al. (1994). Characteri-
1429-1439. zation of a KROX-24/EGR-l-responsive element in the
Habib, EM., Springall, D.R., Davies, G.J. et al. (1996). Tumour h u m a n tumor necrosis factor promoter. Biochim.
necrosis factor and inducible nitric oxide synthase in Biophys.Acta 1219, 413-421.
dilated cardiomyopathy. Lancet. 347, 1151-1155. Kramer. B., Wiegmann, K. and Kronke, M. (1995). Regulation
Han, J., Thompson, P. and Beutler, B. (1990). Dexamethasone of the human TNF promoter by the transcription factor
and pentoxifylline inhibit endotoxin-induced cachectin! Ets. ]. Biol. Chem. 270, 6577-6583.
tumor necrosis factor synthesis at separate points in the Kriegler, M., Perez, C., De Fay, K. et al. (1988). A novel form of
signaling pathway. ]. Exp. Med. 172, 391-394. TNF/cachectin is a cell surface cytotoxic transmembrane
Hegewisch, S., Weh, H.J. and Hossfeld, D.K. (1990). TNF- protein: ramifications for the complex physiology of TNE
induced cardiomyopathy. Lancet 335, 294-295 (letter). Cell 53, 45-53.
Hill, A.V. (1992). Malaria resistance genes: a natural selection. Kruys, V., Marinx, O., Shaw, G. et al. (1989). Trans-
Trans. R. Soc. Trop. Med. Hyg. 86, 225-226, 232. lational blockade imposed by cytokine-derived UA-rich
Hofman, P.M., Hinton, D.R., Johnson, K. et al. (1989). Tumor sequences. Science 245, 852-855.
necrosis factor identified in multiple sclerosis brain. Kwiatkowski, D., Hill, A.V., Sambou, I. et al. (1990). TNF
]. Exp.,Med. 170, 607-612. concentration in fatal cerebral, non-fatal cerebral, and
Hohmann, H.P., Remy. R., Brockhaus, M. et al. (1989). Two uncomplicated P l a s m o d i u m f a l c i p a r u m malaria. Lancet
different cell types have different major receptors for 336, 1201-1204.
human tumor necrosis factor (TNF-a). J. Biol. Chem. 264, Lacraz, S., Nicod, L., Galve-de Rocnemonteix, B. et al. (1992).
14927-14934. Suppression of metalloproteinase biosynthesis in human
Hohmann, H.P., Brockhaus, M., Baeuerle, P.A. et al. (1990). alveolar macrophages by intefleukin-4. L Clin. Invest. 90,
Expression of the types A and B tumor necrosis factor 382-388.
(TNF) receptors is independently regulated, and both Lacraz, S., Nicod, L.P., Chicheportiche, R. et al. (1995). IL-10
receptors mediate activation of the transcription factor inhibits metalloproteinase and stimulates TIMP-1 pro-
NF-~:B. TNF-a is not needed for induction of a biological duction in human mononuclear phagocytes. I. Clin.
effect via TNF receptors. J. Biol. Chem. 265, 22409-22417. Invest. 96, 2304-2310.
Hsu, H., Xiong, J. and Goeddel, D.V. (1995). The TNF receptor Lai, W.S., Carballo, E., Strum, J.R. et al. (1999). Evidence that
1-associated protein TRADD signals cell death and NF- tristetraprolin binds to AU-rich elements and promotes
~:B activation. Cell 81,495-504. the deadenylation and destabilization of tumor necrosis
Hsu. H., Shu, H.B., Pan, M.G. et al. (1996). TRADD-TRAF2 factor alpha mRNA. Mol. Cell, Biol. 19, 4311-4323.
and TRADD-FADD interactions define two distinct TNF Lee, E., Vaughan, D.E., Parikh, S.H. et al. (1996). Regulation of
receptor 1 signal transduction pathways. Cell 84, matrix metalloproteinases and plasminogen activator
299-308. inhibitor-1 synthesis by plasminogen in cultured human
Jacob, C.O., Fronek, Z., Lewis, G.D. et al. (1990). Heritable vascular smooth muscle cells. Circ. Res. 78, 44-49.
major histocompatibility complex class II-associated Lee. ES., Hagler. J., Chen, Z.J. et al. (1997). Activation of the
differences in production of tumor necrosis factor ~: bcBa kinase complex by MEKK1, a kinase of the JNK path-
relevance to genetic predisposition to systemic lupus way. Cell 88, 213-222.
erythematosus. Proc. Natl Acad. Sci. USA 87, 1233-1237. Lee, J.C., Laydon, J.T., McDonnell, P.C. et al. (1994). A protein
Jones, E.Y., Stuart, D.I. and Walker, N.P. (1989). Structure of kinase involved in the regulation of inflammatory
tumour necrosis factor. Nature 338, 225-228. cytokine biosynthesis. Nature 372, 739-746.
Jongeneel, C.V., Briant, L., Udalova, I.A. etal. (1991). Extensive Leeuwenberg, J.E, Jeunhomme, T.M. and Buurman, W.A.
genetic polymorphism in the human tumor necrosis fac- (1994). Slow release of soluble TNF receptors by mono-
tor region and relation to extended HLA haplotypes. Proc. cytes in vitro. ]. I m m u n o l . 152, 4036-4043.
Natl Acad. Sci. USA 88, 9717-9721. Leist, T.P., Frei, I.C., Kam-Hansen, S. et al. (1988). Tumor
Kam, L.Y. and Targan, S.R. (2000). TNF-alpha antagonists necrosis factor-a in cerebrospinal fluid during bacterial,
for the treatment of Crohn's disease. Expert Opin. but not viral, meningitis. Evaluation in murine model
Pharmacother. 1, 615-622. infections and in patients. ]. Exp. Med. 167, 1743-1748.
Kapadia, S., Torre-Amione, G., Yokoyama, T. et al. (1995). Leitman, D.C., Mackow, E.R., Williams, T. et al. (1992). The
Soluble TNF binding proteins modulate the negative core promoter region of the tumor necrosis factor a gene
inotropic properties of TNF-~ in vitro. Am. ]. Physiol. 268, confers phorbol ester responsiveness to upstream tran-
H517-H525. scriptional activators. Mol. Cell Biol. 12, 1352-1356.
Katz, S.D., Rao, R., Berman, J.W. "et al. (1994). Patho- Levine, B., Kalman, J., Mayer, L. et al. (1990). Elevated circu-
physiological correlates of increased serum tumor necro- lating levels of tumor necrosis factor in severe chronic
sis factor in patients with congestive heart failure. heart failure. N. Engl]. Med. 323, 236-241.

THE CYTOKINES AND CHEMOKINES


REFERENCES 857

Lienard, D., Ewalenko. P., Delmotte. J.J. et al. (1992). Histocompatibility antigens in small cell carcinoma of
High-dose recombinant tumor necrosis factor r in com- the lung. Cancer 54, 2943-2945.
bination with interferon 7 and melphalan in isolation Marx, J. (1995). How the glucocorticoids suppress immunity.
perfusion of the limbs for melanoma and sarcoma. Science 270, 232-233.
]. Clin. Oncol. 10, 52-60. Mattson, M.P., Zhang, Y. and Bose, S. (1993). Growth factors
Lilly, C.M., Sandhu, J.S., Ishizaka, A. et al. (1989). Pento- prevent mitochondrial dysfunction, loss of calcium
xifylline prevents tumor necrosis factor-induced lung homeostasis, and cell injury, but not ATP depletion in
injury. Am. Rev. Respir. Dis. 139, 1361-1368. hippocampal neurons deprived of glucose. Exp. Neurol.
Lin, L.L., Wartmann, M., Lin, A.Y. etal. (1993). CPLA2 is phos- 121, 1-13.
phorylated and activated by MAP kinase. Cell 72, 269-278. McGeehan, G.M., Becherer, J.D., Bast, R.C., Jr. et al.
Lin, L.S. (1992). In: Beutler, B (ed.) Tumor Necrosis Factor: (1994). Regulation of tumour necrosis factor-R process-
The Molecules a n d Their Emerging Role in Medicine. New ing by a metalloproteinase inhibitor. N a t u r e 370,
York: Raven Press, pp. 33-48. 558-561.
Lin, R.H., Hwang, Y.W.,Yang, B.C. et al. (1997). TNF receptor- McGuire, W., Hill, A.V., Allsopp, C.E. et al. (1994). Variation in
2-triggered apoptosis is associated with the down- the TNF-a promoter region associated with susceptibility
regulation of Bcl-xL on activated T cells and can be to cerebral malaria. Nature 371,508-510.
prevented by CD28 costimulation. ]. I m m u n o l . 158, McManus, R., Moloney, M., Borton, M. et al. (1996). Associa-
598-603. tion of celiac disease with microsatellite polymorphisms
Liu, T., Clark, R.K., McDonnell, PC. et al. (1994). Tumor close to the tumor necrosis factor genes. H u m . I m m u n o l .
necrosis factor-alpha expression in ischemic neurons. 45, 24-31.
Stroke 25, 1481-1488. McNamara, M.J., Norton, J.A., Nauta, R.J. et al. (1993).
Liu, Z.G., Hsu, H., Goeddel, D.V. et al. (1996). Dissection of Interleukin- 1 receptor antibody (IL-1RAB) protection
TNF receptor 1 effector functions: JNK activation is not and treatment against lethal endotoxemia in mice. ]. Surg.
linked to apoptosis while NF-~:B activation prevents cell Res. 54, 316-321.
death. Cell 87, 565-576. Mehler, M.F., Rozental., R., Dougherty, M. et al. (1993).
Loetscher, H., Stueber, O., Banner, D. et al. (1993). Human Cytokine regulation of neuronal differentiation of hip-
tumor necrosis factor ~ (TNF ~) mutants with exclusive pocampal progenitor cells. Nature 362, 62-65.
specificity for the 55-kDa or 75-kDa TNF receptors. ]. Biol. Meistrell, M., III, Botchkina. G.I., Wang, H. et al. (1997). TNF
Chem. 268, 26350-26357. is a brain-damaging cytokine in cerebral ischemia. Shock
Lovell, D.J., Giannini, E.H., Reiff, A. et al. (2000). Etanercept 8, 341-348.
in children with plyarticular juvenile rheumatoid arthri- Meyer, C.G., Gallin, M., Erttmann, K.D. et al. (1994). HLA-D
tis. N. Engl ]. Med. 342, 763-769. alleles associated with generalized disease, localized dis-
Machleidt, T., Kramer, B., Adam, D. et al. (1996). Function of ease, and putative immunity in Onchocerca volvulus
the p55 tumor necrosis factor receptor 'death domain' infection. Proc. Natl Acad. Sci. USA 91, 7515-7519.
mediated by phosphatidylcholine-specific phospholi- Millar, A.B., Foley, N.M., Singer, M. et al. (1989). Tumour
pase C. ]. Exp. Med. 184, 725-733. necrosis factor in b r o n c h o p u l m o n a r y secretions of
Mackay, E, Rothe, J., Bluethmann, H. et al. (1994). patients with adult respiratory distress syndrome. Lancet
Differential responses of fibroblasts from wild-type and ii, 712-714.
TNF-r55-deficient mice to mouse and h u m a n TNF-~ Mohler, K.M., Torrance, D.S., Smith, C.A. et al. (1993). Soluble
activation. ]. I m m u n o l . 153, 5274-5284. tumor necrosis factor (TNF) receptors are effective thera-
Maini, R., St. Clair, E.W., Breeveld, F. et al. (1999). Infliximab peutic agents in lethal endotoxemia and function simul-
(chimeric anti-tumour necrosis factor-~ monoclonal taneously as both TNF carriers and TNF antagonists.
antibody) versus placebo in rheumatoid arthritis patients ]. I m m u n o l . 151, 1548-1561.
receiving concomitant methotrexate: a randomised MoMg, J., Baek, L., Christensen, P. et al. (1988). Endotoxin-
phase III trial. Lancet 354, 1932-1939. stimulated h u m a n monocyte secretion of interleukin 1,
Majetschak, M., Flohe, S., Obertacke, U. et al. (1999). tumour necrosis factor ~, and prostaglandin E2 shows
Relation of a TNF gene polymorphism to severe sepsis stable interindividual differences. Scand. ]. I m m u n o l . 27,
in trauma patients. Ann. Surg. 230, 207-214. 705-716.
Malinin, N.L, Boldin, M.P., Kovalenko, A.V. et al. (1997). Moriarty, T.M., Padrell. E., Carty, D.J. et al. (1990). GO protein
MAP3K-related kinase involved in NF-KB induction by as signal transducer in the pertussis toxin-sensitive phos-
TNF, CD95 and IL-1. Nature 385, 540-544. phatidylinositol pathway. Nature 343, 79-82.
Mallett, S. and Barclay, A.N. (1991). A new superfamily of cell Moser, R.B., Schleiffenbaum, B., Groscurth, P. et al. (1989).
surface proteins related to the nerve growth factor recep- Interleukin 1 and tumor necrosis factor stimulate human
tor. I m m u n o l . Today 12, 220-223. vascular endothelial cells to promote transendothelial
Mandrup-Poulsen, T., Bendtzen, K., Dinarello, C.A. et al. neutrophil passage. ]. Clin. Invest. 83, 444-455.
(1987). Human tumor necrosis factor potentiates h u m a n Moss, M.L., Jin, S.L., Milla, M.E. et al. (1997). Cloning of a
interleukin 1-mediated rat pancreatic [3-cell cytotoxicity. disintegrin metalloproteinase that processes precursor
]. I m m u n o l . 139, 4077-4082. tumour-necrosis factor-~. Nature 385, 733-736.
Manus, R.M., Wilson, A.G., Mansfield, J. et al. (1996). TNF2, a Mullberg, J., Durie, F.H., Otten-Evans, C. et al. (1995). A
polymorphism of the tumour necrosis-a gene promoter, metalloprotease inhibitor blocks shedding of the IL-6
is a c o m p o n e n t of the celiac disease major histo- receptor and the p60 TNF receptor. ]. I m m u n o l . 155,
compatibility complex haplotype. Eur. ]. I m m u n o l . 26, 5198-5205.
2113-2118. Mtiller, G., Ayoub, M., Storz, P. et al. (1995). PKC zeta is a
Markman, M., Braine, H.G. and Abeloff, M.D. (1984). molecular switch in signal transduction of TNF-alpha,

THE CYTOKINES AND CHEMOKINES


858 TUMOUR NECROSIS FACTOR

bifunctionally regulated by ceramide and arachidonic Pociot, E, Wilson, A.G., Nerup, J. et al. (1993b). No independ-
acid. EMBO]. 14, 1961-1969. ent association between a tumor necrosis factor-a pro-
Muzio, M., Chinnaiyan, A.M., Kischkel, F.C. et al. (1996). moter region polymorphism and insulin-dependent
FLICE, a novel FADD-homologous ICE/CED-3-1ike pro- diabetes mellitus. Eur. ]. I m m u n o l . 23, 3050-3053.
tease, is recruited to the CD95 (Fas/APO-1) death- Rood, M.J., van Krugten, M.V., Zanelli, E. et al. (2000). TNF-
inducing signaling complex. Cell 85, 817-827. 308A and HLA-DR3 alleles contribute independently to
Narhi, L.O., Philo, J.S., Li, T. et al. (1996). Induction of susceptibility to systemic lupus erythematosus. Arthritis
a-helix in the ~-sheet protein tumor necrosis factor-a: Rheum. 43, 129-134.
acid-induced denaturation. Biochemistry 35, Rothe, M., Sarma, V., Dixit, V.M. et al. (1995). TRAF2-
11454-11460. mediated activation of NF-KB by TNF receptor 2 and
Natanson, C., Eichenholz, P.W., Danner, R.L. et al. (1989). CD40. Science 269, 1424-1427.
Endotoxin and tumor necrosis factor challenges in dogs Sampaio, E.E, Sarno, E.N., Galilly, R. et al. (1991). Thalido-
simulate the cardiovascular profile of human septic mide selectively inhibits tumor necrosis factor a produc-
shock. ]. Exp. Med. 169, 823-832. tion by stimulated human monocytes. ]. Exp. Med. 173,
Natoli, G., Costanzo, A., Ianni, A. et al. (1997). Activation of 699-703.
SAPK/JNK by TNF receptor 1 through a noncytotoxic Sandborn, W.J. and Hanauer, S.B. (1999). Antitumor necrosis
TRAF2-dependent pathway. Science 275, 200-203. factor therapy for inflammatory bowel disease: a review
Nawroth, PP., Bank, I., Handley, D. et al. (1986). Tumor necro- of agents, pharmacology, clinical results, and safety.
sis factor/cachectin interacts with endothelial cell recep- Inflamm. Bowel Dis. 5, 119-133.
tors to induce release of interleukin 1. ]. Exp. Med. 163, Sato, N., Goto, T., Haranaka, K. et al. (1986). Actions of tumor
1363-1375. necrosis factor on cultured vascular endothelial cells:
North, R.J. and Havell, E.A. (1988). The antitumor function of morphologic modulation, growth inhibition, and cyto-
tumor necrosis factor (TNF). II. Analysis of the role of toxicity. ]. Natl Cancer Inst. 76, 1113-1121.
endogenous TNF in endotoxin-induced hemorrhagic Sato, Y., Koshita, Y., Hirayama, M. et al. (1996). Augmented
necrosis and regression of an established sarcoma. 1. Exp. antitumor effects of killer cells induced by tumor necro-
Med. 167, 1086-1099. sis factor gene-transduced autologous tumor cells from
Ohkawa, S., Wright, K.C., Mahajan, H. et al. (1989). Hepatic gastrointestinal cancer patients. Hum. Gene Ther. 7,
arterial infusion of human recombinant tumor necrosis 1895-1905.
factor-a. An experimental study in dogs. Cancer 63, Saukkonen, K., Sande, S., Cioffe, C. et al. (1990). The role of
2096-2102. cytokines in the generation of inflammation and tis-
Panagakos, ES., O'Boskey, J.E, Jr. and Rodriguez, E. (1996). sue damage in experimental Gram-positive meningitis.
Regulation of pulp cell matrix metalloproteinase produc- ]. Exp. Med. 171,439-448.
tion by cytokines and lipopolysaccharides. 1. Endod. 22, Scheinman, R.I., Cogswell, EC., Lofquist, A.K. et al. (1995).
358-361. Role of transcriptional activation of IKB a in mediation
Partanen. J. and Koskimies. S. (1988). Low degree of DNA of immunosuppression by glucocorticoids. Science 270,
polymorphism in the HIA-linked lymphotoxin (tumour 283-286.
necrosis factor 13) gene. Scand. 1. I m m u n o l . 28, Schiller, J.H., Win, EL. Storer, B. et al. (1992). Clinical and bio-
313-316. logic effects of combination therapy with 7-interferon
Pena, L.A., Fuks, Z. and Kolesnick, R. (1997). Stress-induced and tumor necrosis factor. Cancer69, 562-571.
apoptosis and the sphingomyelin pathway. Biochem. Schutze, S., Potthoff, K., Machleidt, T. et al. (1992). TNF acti-
Pharmacol. 53, 615-621. vates NF-KB by phosphatidylcholine-specific phospholi-
Peppel, K., Crawford, D. and Beutler, B. (1991). A tumor pase C-induced 'acidic' sphingomyelin breakdown. Cell
necrosis factor (TNF) receptor-IGG heavy chain chimeric 71,765-776.
protein as a bivalent antagonist of TNF activity. 1. Exp. Schwarz, E.M., Looney, R.J. and O'Keefe, R.J. (2000). Anti-
Med. 174, 1483-1489. TNF-alpha therapy as a clinical intervention for peripros-
Pfeffer, K., Matsuyama, T., Kundig, T.M. et al. (1993). Mice thetic osteolysis. Arthritis Res. 2, 165-168.
deficient for the 55 kD tumor necrosis factor receptor are Seckinger, P., Isaaz, S. and Dayer, J.M. (1989). Purification
resistant to endotoxic shock, yet succumb to L. monocy- and biologic characterization of a specific tumor necrosis
togenes infection. Cell 73, 457-467. factor a inhibitor. ]. Biol. Chem. 264, 11966-11973.
Piecyk, M., Wax, S., Beck, A.R. et al. (2000). TIA-1 is a transla- Segal, R., Dayan, M., Zinger, H. et al. (2001). Suppres-
tional silencer that selectively regulates the expression of sion of experimental systemic lupus erythematosus
TNF-alpha. EMBO ]. 19, 4154-4163. (SLE) in mice via TNF inhibition by an anti-TNFa
Pincus, T., Brooks, R.H. and Callahan, L.E (1994). Prediction monoclonal antibody and by pentoxiphylline. Lupus 10,
of long-term mortality inpatients with rheumatoid 23-31.
arthritis according to simple questionnaire and joint S6gui, B., Bezombes, C., Uro-Coste, E. et al. (2000). Stress-
count measures. Ann. Intern. Med. 120, 26-34. induced apoptosis is not mediated by endolysosomal
Plata-Salaman, C.R. (1991). Immunoregulators in the nerv- ceramide. FASEB ]. 14, 36-47.
ous system. Neurosci. Biobehav. Rev. 15, 185-215. Selmaj, K.W., Farooq, M., Norton, W.T. et al. (1990). Prolifera-
Pociot, E, Briant, L, Jongeneel, C.V. et al. (1993a). Association tion of astrocytes in vitro in response to cytokines. A pri-
of tumor necrosis factor (TNF) and class II major histo- mary role for tumor necrosis factor. ]. I m m u n o l . 144,
compatibility complex alleles with the secretion of TNF-a 129-135.
and TNF-[3 by human mononuclear cells: a possible link Shanahan, E (2000). Anti-TNF therapy for Crohn's disease: a
to insulin-dependent diabetes mellitus. Eur. 1. I m m u n o l . perspective (infliximab is not the drug we have been
23, 224-231. waiting for). Inflamm. Bowel Dis. 6, 137-139.

THE CYTOKINES AND CHEMOKINES


REFERENCES 859

Shaw, G. and Kamen, R. (1986). A conserved AU sequence Tracey, K.J. (1998). Suppression of TNF and other proinflam-
from the 3' untranslated region of GM-CSF mRNA medi- matory cytokines by the tetravalent guanylhydrazone
ates selective mRNA degradation. Cell 46, 659-667. CNI-1493. Prog. Clin. Biol. Res. 397, 335-343.
Shimura, T., Hagihara, M., Takebe, K. et al. (1994). The study Tracey, K.J. and Cerami, A. (1990). Metabolic responses to
of tumor necrosis factor [3 gene polyrnorphism in lung cachectin/TNE A brief review. Ann. N Y Acad. Sci. 587,
cancer patients. Cancer 73, 1184-1188. 325-331.
Shingu, M., Nagai, Y., Isayama, T. et al. (1993). The effects of Tracey, K.J. and Cerami, A. (1994). Tumor necrosis factor: a
cytokines on metalloproteinase inhibitors (TIMP) and pleiotropic cytokine and therapeutic target. Annu. Rev.
collagenase production by h u m a n chondrocytes and Med. 45, 491-503.
TIMP production by synovial cells and endothelial cells. Tracey, K.J., Beutler, B., Lowry, S.E et al. (1986a). Shock and
Clin. Exp. I m m u n o l . 94, 145-149. tissue injury induced by recombinant h u m a n cachectin.
Singh, I.S., He, J.R., Calderwood, S. et al. (2001). A high affin- Science 234, 470-474.
ity HSF- 1 binding site in the 5'-untranslated region of the Tracey K.J., Lowry S.E, Beutler B. et al. (1986b). Cachectin/
murine tumor necrosis factor- (alpha) gene is a transcrip- tumor necrosis factor mediates changes of skeletal
tional repressor. 1. Biol. Chem. 15, 4981-4988. muscle plasma membrane potential. 1. Exp. Med. 164,
Silver, I.A., Murrills, R.J. and Etherington, D.J. (1988). Micro- 1368-1373.
electrode studies on the acid microenvironment beneath Tracey, K.J., Fong, Y., Hesse, D.G. et al. (1987a). Anti-
adherent macrophages and osteoclasts. Exp. Cell Res. cachectin/TNF monoclonal antibodies prevent septic
175, 266-276. shock during lethal bacteraemia. Nature 330, 662-664.
Smith, R.A. and Baglioni, C. (1987). The active form of tumor Tracey, K.J., Lowry. S.E, Fahey, T.J., III. et al. (1987b).
necrosis factor is a trimer. 1. Biol. Chem. 262, 6951-6954. Cachectin/tumor necrosis factor induces lethal shock
Smith, R.A. and Baglioni, C. (1992). Characterization of TNF and stress hormone respenses in the dog. Surg. Gynecol.
receptors. I m m u n o l . Ser. 56, 131-147. Obstet. 164, 415-422.
Sollid, L.M., Markussen, G., Ek, J. et al. (1989). Evidence for Tracey, K.J., Wei, H., Manogue, K.R. et al. (1988). Cachectin/
a primary association of celiac disease to a particular tumor necrosis factor induces cachexia, anemia, and
HLA-DQ~/[3 heterodimer. 1. Exp. Med. 169, 345-350. infammation. ]. Fxp. Med. 167, 1211-1227.
Spence, M.W. (1993). Sphingomyelinases. Adv. Lipid Res. 26, Tracey, K.J., Vlassara, H. and Cerami, A. (1989). Cachectin/
3-23. tumour necrosis factor. Lancet i, 1122-1126.
Stanger, B.Z., Leder, P., Lee, T.H. etal. (1995). RIP: a novel pro- Tsai, E.Y., Yie, J., Thanos, D. et al. (1996). Cell-type-specific
tein containing a death domain that interacts with regulation of the h u m a n tumor necrosis factor-a gene in
FAS/APO-1 (CD95) in yeast and causes cell death. Cell81, B cells and T cells by NFATP and ATF-2/Jun. Mol. Cell BioL
513-523. 16, 5232-5244.
Stephens, K.E., Ishizaka, A., Wu, Z.H. et al. (1988). Tsuchiya, N., Kawasaki, A., Tsao, B.P. et al. (2001). Analysis of
Granulocyte depletion prevents tumor necrosis factor- tbe association of HLA-DRB1, TNFalpha promoter and
mediated acute lung injury in guinea pigs. Am. Rev. TNFR2 (TNFRSF1B) polymorphisms with SLE using
Respir. Dis. 138, 1300-1307. transmission disequilibrium test. Genes I m m u n . 2,
Stuber, E, Petersen, M., Bokelmann, E et al. (1996). A 317-322.
genomic polymorphism within the tumor necrosis factor Tsutsumi, Y., Kihira, T., Tsunoda, S. et al. (1995). Molecular
locus influences plasma tumor necrosis factor-a concen- design of hybrid tumour necrosis factor-a with polyethyl-
trations and outcome of patients with severe sepsis. Crit. ene glycol increases its anti-tumour potency. Br. I. Cancer
Care Med. 24, 381-384. 71,963-968.
Suffredini, A.E, Fromm, R.E., Parker, M.M. et al. (1989). The Udalova, I.A., Nedospasov, S.A., Webb, G.C. et al. (1993).
cardiovascular response of normal h u m a n s to the Highly informative typing of the h u m a n TNF locus
administration of endotoxin. N. Engl 1. Med. 321, using six adjacent polymorphic markers. Genomics 16,
280-287. 180-186.
Sugarman, B.J., Aggarwal., B.B., Hass, P.E. et al. (1985). Van Antwerp, D.J., Martin, S.J., Kafri, T. et al. (1996).
Recombinant human tumor necrosis factor-a: effects on Suppression of TNF-a-induced apoptosis by NF-KB.
proliferation of normal and transformed cells in vitro. Science 274, 787-789.
Science 230, 943-945. van der Goot, EG., Pugin, J., Hribar, M. et al. (1999). Mem-
Tartaglia, L.A., Weber, R.F., Figari, I.S. et al. (1991). The two brane interaction of TNF is not sufficient to trigger
different receptors for tumor necrosis factor mediate increase in membrane conductance in mammalian cells.
distinct cellular responses. Proc. Natl Acad. Sci. USA 88, FEBS Lett. 460, 107-111.
9292-9296. van der Linden, M.W., van der Slik, A.R., Zanelli, E. et al.
Tartaglia, L.A., Ayres, T.M., Wong, G.H. et al. (1993a). A novel (2001). Six microsatellite markers on the short arm of
domain within the 55 kD TNF receptor signals cell death. chromosome 6 in relation to HLA-DR3 and TNF-308A in
Cell 74, 845-853. systemic lupus erythematosus. Genes I m m u n . 2, 373-380.
Tartaglia, L.A., Pennica, D. and Goeddel, D.V. (1993b). Ligand van der Poll, T. and Lowry, S.E (1995). Tumor necrosis factor
passing: the 75-kDA tumor necrosis factor (TNF) receptor in sepsis: mediator of multiple organ failure or essential
recruits TNF for signaling by the 55-kDa TNF receptor. part of host defense? Shock 3, 1-12 (editorial).
]. Biol. Chem. 268, 18542-18548. Van Ostade, X., Vandenabeele, P., Everaerdt, B. et al. (1993).
Taylor, PC. (2001). Anti-tumor necrosis factor therapies. Human TNF mutants with selective activity on the p55
Curr. Opin. Rheumatol. 13, 164-169. receptor. Nature 361,266-269.
Thomson, A.M. (1997). Neuroscience. More than just fre- Van Zee, K.J., Stackpole, S.A., Montegut, W.J. et al. (1994). A
quency detectors? Science275, 179-180. human tumor necrosis factor (TNF) a mutant that binds

THE CYTOKINES AND CHEMOKINES


860 TUMOUR NECROSIS FACTOR

exclusively to the p55 TNF receptor produces toxicity in manganous superoxide dismutase by tumor necrosis
the baboon. ]. Exp. Med. 179, 1185-1191. factor: possible protective mechanism. Science 242,
Via, C.S., Shustov, A., Rus, V. et al. (2001). In vivo neutraliza- 941-944.
tion of TNF-alpha promotes humoral autoimmunity by Xia, Z., Dickens, M., Raingeaud, J. et al. (1995). Opposing
preventing the induction of CTL. 1. I m m u n o l . 167, effects of ERK and JNK-p38 MAP kinases on apoptosis.
6821-6826. Science 270, 1326-1331.
Waage, A., Brandtzaeg, P., Halstensen, A. et al. (1989). The Yokoyama, T., Vaca, L., Rossen, R.D. et al. (1993). Cellular
complex pattern of cytokines in serum from patients with basis for the negative inotropic effects of tumor necrosis
meningococcal septic shock. Association between inter- factor-a in the adult mammalian heart. ]. Clin. Invest. 92,
leukin 6, interleukin 1. and fatal outcome. ]. Exp. Med. 2303-2312.
169, 333-338. Yoshimura, T and Sone, S. (1987). Different and synergistic
Wang, C.Y., Mayo, M.W. and Baldwin, A.S., Jr. (1996). TNF- actions of human tumor necrosis factor and interferon-7
and cancer therapy-induced apoptosis: potentiation by in damage of liposome membranes. J. Biol. Chem. 262,
inhibition of NF-KB. Science 274, 784-787. 4597-4601.
Weinblatt, M.E., Kremer, J.M., Bankhurst, A.D. et al. (1999). A Young, P., McDonnell, P., Laydon, J. et al. (1994). A novel MAP
trial of etanercept, a recombinant tumor necrosis fac- kinase regulates the production of IL-1 and TNF in LPS
tor:Fc fusion protein, in patients with rheumatoid arthri- activated human monocytes. Cytokine 6, 564.
tis receiving methotrexate. N. Engl]. Med. 340, 253-259. Zhang, M., Caragine, T., Wang, K. et al. (1997). Spermine
Welch, T.R., Beischel, L.S., Balakrishnan, K. et al. (1988). inhibits proinflammatory cytokine synthesis in human
Major histocompatibility complex extended haplotypes mononuclear cells. J. Exp. Med. 185, 1759-1768.
in systemic lupus erythematosus. Dis. Markers 6, Zhang, X.M., Weber, I. and Chen, M.J. (1992). Site-directed
247-255. mutational analysis of human tumor necrosis factor-a
Wiegmann, K., Schutze, S., Machleidt, T. et al. (1994). receptor binding site and structure-functional relation-
Functional dichotomy of neutral and acidic sphin- ship. ]. Biol. Chem. 267, 24069-24075.
gomyelinases in tumor necrosis factor signaling. Cell 78, Zheng, L., Fisher, G., Miller, R.E. et al. (1995). Induction of
1005-1015. apoptosis in mature T cells by tumour necrosis factor.
Wilson, A.G., de Vries, N., Pociot, E et al. (1993). An allelic Nature 377, 348-351.
polymorphism within the human tumor necrosis factor- Zhu, W., Downey, J.S., Gu, J. et al. (2000). Regulation of TNF
promoter region is strongly associated with HLA A1, B8, expression by multiple mitogen-activated protein kinase
and DR3 alleles. ]. Exp. Med. 177, 557-560. pathways. J. I m m u n o l . 164, 6349-6358.
Wilson, A.G., Gordon. C., Di Giovine, ES. etal. (1994). A genetic Zimmerman, R.J., Chan, A. and Leadon, S.A. (1989).
association between systemic lupus erythematosus and Oxidative damage in murine tumor cells treated in vitro
tumor necrosis factor-a. Eur. ]. I m m u n o l . 24, 191-195. by recombinant human tumor necrosis factor. Cancer
Wilson, A.G., Symons, J.A., McDowell, T.L. et al. (1997). Res. 49, 1644-1648.
Effects of a polymorphism in the human tumor necrosis Zubiaga, A.M., Belasco, J.G. and Greenberg, M.E. (1995). The
factor-a promoter on transcriptional activation. Proc. nonamer UUAUUUAUU is the key AU-rich sequence
Natl Acad. Sci. USA 94, 3195-3199. motif that mediates mRNA degradation. Mol. Cell Biol.
Winston, B.W. and Riches, D.X. (1995). Activation of 15, 2219-2230.
p42MAPK/ERK2 following engagement of tumor necrosis Zuniga, J., Vargas-Alarcon, G., Hernandez-Pacheco, G. et al.
factor receptor CD120A (p55) in mouse macrophages. (2001). Tumor necrosis factor-alpha promoter polymor-
]. I m m u n o l . 155, 1525-1533. phisms in Mexican patients with systemic lupus erythe-
Wong, G.H. and Goeddel, D.V. (1988). Induction of matosus (SLE). Genes I m m u n . 2, 363-366.

THE CYTOKINES AND CHEMOKINES

You might also like