You are on page 1of 8

663

Exp Physiol 90.5 pp 663670

Experimental Physiology Hot Topic Review

Actions of TNF- on glutamatergic synaptic transmission


in the central nervous system
Mark Pickering, Derval Cumiskey and John J. OConnor
Department of Human Anatomy and Physiology, Conway Institute of Biomolecular and Biomedical Research, University College Dublin,
Belfield, Dublin 4, Ireland

Increasing attention is being paid to the role of inflammatory and immune molecules in the
modulation of central nervous system (CNS) function. Tumour necrosis factor- (TNF-) is
a pro-inflammatory cytokine, the receptors for which are expressed on neurones and glial cells
throughout the CNS. Through the action of its two receptors, it has a broad range of actions
on neurones which may be either neuroprotective or neurotoxic. It plays a facilitatory role in
glutamate excitotoxicity, both directly and indirectly by inhibiting glial glutamate transporters
on astrocytes. Additionally, TNF- has direct effects on glutamate transmission, for example
increasing expression of AMPA receptors on synapses. TNF- also plays a role in synaptic
plasticity, inhibiting long-term potentiation (LTP), a process dependent on p38 mitogen activated
kinase (p38 MAP) kinase. In the following review we look at these and other effects of TNF-
in the CNS.
(Received 4 May 2005; accepted after revision 7 June 2005; first published online 00 Month 2005)
Corresponding author J. J. OConnor: Department of Human Anatomy and Physiology, Conway Institute of
Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland.
Email: john.oconnor@ucd.ie

For many decades the general consensus was that the


immune and central nervous systems were relatively
independent due to the inaccessibility of the brain
to the immune cells because of the bloodbrain
barrier. This outlook has changed in recent years as
it has become apparent that many immune molecules
may be used by the nervous system in intercellular
communication (Boulanger et al. 2001; Chun, 2001).
This review addresses one such immune molecule,
tumour necrosis factor- (TNF-) in relation to its
role in neurotoxicity, synaptic transmission and synaptic
plasticity.
The pro-inflammatory cytokine TNF- is a 17-kDa
peptide and forms multimers, which are active in binding
TNF- receptors that are constitutively expressed on
both neurones and glia in the central nervous system
(Benveniste & Benos, 1995). TNF- can be synthesized
and released in the brain by astrocytes, microglia and some
neurones (Lieberman et al. 1989; Chung & Benveniste,
1990; Morganti-Kossman et al. 1997). Brain TNF- levels
are typically increased in a wide range of CNS disorders,
including trauma (Goodman et al. 1990), ischaemia (Liu
et al. 1994) and multiple sclerosis (Rieckmann et al. 1995).
Under such pathological conditions, the expression and
release of TNF- is rapid and in some cases as early as 1 h

C The Physiological Society 2005

after the brain insult and well before neuronal death (Liu
et al. 1994; Wang et al. 1994; Allan & Rothwell, 2001).
Two different receptors for TNF- (p55, or TNF-R1
and p75, or TNF-R2) have been identified (Beutler & Van
Huffel, 1994a,b; Wajant & Scheurich, 2001) and shown
to mediate differential cellular responses using distinct
pathways (Kinouchi et al. 1991; Tartaglia et al. 1991). These
receptors have been shown to exist to varying degrees in the
brainstem, cortex, cerebellum, thalamus and basal ganglia
(Kinouchi et al. 1991). The TNF-R1 and TNF-R2 are
present in both neurones and glia (Boka et al. 1994). The
pathways activated by TNF-R1 and TNF-R2 are diverse,
and include G-protein-mediated activation of protein
kinase A (PKA), phospholipase C and phospholipase A2,
activation of the sphingomyelinase pathway, production
of nitric oxide and ceramide, free radical formation and
phosphorylation of other membrane receptors by protein
kinases (Kronke et al. 1990; Rothe et al. 1992; Beyaert &
Fiers, 1994). The pathways activated by the two receptors
are summarized in Fig. 1. Note that while either receptor
can lead to the activation of transcription factors, only
TNF-R1 activation can lead to activation of the caspase
pathways leading to apoptosis.
In the context of this review, it is probably most
relevant to note that the signal transduction pathway
DOI: 10.1113/expphysiol.2005.030734

664

M. Pickering and others

used by the p55 TNF-R1 can result in the activation


of the transcription factor nuclear factor kappa B (NFB) (Kolesnick & Golde, 1994; Goodman & Mattson,
1996; Mattson et al. 1997a,b), which is pivotal in
controlling diverse cellular processes, including immune
responses, cell proliferation and differentiation (Israel,
2000; Silverman & Maniatis, 2001; Ghosh & Karin, 2002;
Li & Verma, 2002). Increasingly, it has become evident that
NF-B also plays important roles in the CNS (ONeill &
Kaltschmidt, 1997).
A role for TNF- in glutamate neurotoxicity

Although TNF- was originally named for its


degeneration-inducing action in some types of tumour
cells, the relationship between TNF- and cell toxicity and
death is not a simple one. On the one hand, TNF- has
been shown to induce cell death in septo-hippocampal
cultures. Zhao et al. (2001) detected -spectrin fragments
in these septo-hippocampal cultures treated with TNF-
and found elevated 120kDa fragments, indicative of
caspase-3 activity, but not 145-kDa fragments, indicative
of calpain activity. Unlike calpain, which is associated
with both necrotic and apoptotic cell death, caspase-3
is exclusively characteristic of apoptosis-like cell death
(Armstrong et al. 1996; Wang et al. 1996; Nath et al. 1998).
On the other hand, a number of investigators have
presented evidence that, under different conditions,
TNF- may play a protective role against neuronal cell
death. For example, while it has been shown that TNF-
mediates damage to myelin and oligodendrocytes (Selmaj
& Raine, 1998), Garcia et al. (1992) found that it was

Figure 1. Schematic diagram of some of the relevant TNF-R


signalling pathways
TNF- binds to both TNF-R1 and TNF-R2 on the cell membrane. Both
receptors activate tumour necrosis factor associated factor 2 (TRAF2),
which causes the activation of the transcription factors activator
protein 1 (AP1) via Jun N-terminal kinase (JNK) and NF-B via
NF-B-inducing kinase (NIK). While both receptors activate this
signalling pathway, only TNF-R1 activation leads to activation of
caspases and ceramide which can lead to apoptosis.

Exp Physiol 90.5 pp 663670

not toxic to rat cultured CNS neurones. TNF- under


in vitro conditions may protect neurones against
metabolic, excitotoxic or oxidative insults by promoting
maintenance of intracellular calcium homeostasis,
suppression of reactive oxygen species (Cheng et al. 1994),
and by activation of transcription factor NF-B (Barger
et al. 1995). Mice genetically deficient in TNF-R1 or both
TNF-R1 and TNF-R2, also show exacerbated neuronal
damage compared to wild-type controls following middle
cerebral artery occlusion (Bruce et al. 1996; Gary et al.
1998).
Another aspect of this complexity may relate to the
role of TNF- in the interactions between neurones and
glia. Increasingly glial cells are no longer seen as passive
supporters of neurones, but as active participants in
information processing (for review see Haydon, 2001;
Volterra & Steinhauser, 2004). Of particular interest is the
relationship between neurones, glial cells and TNF- in
glutamate excitotoxicity.
Excitotoxicity in general is linked to excessive
glutamate activation of receptors, particularly the
N -methyl-d-aspartate (NMDA) receptor. Cell death
resulting from excessive levels of glutamate and over
stimulation of glutamate receptors is known to be caused
by impaired uptake of glutamate by glial cells (Choi,
1988). In vivo, it has been shown that mice lacking
expression of the excitatory amino acid transporter,
EAAT2/GLT-1 develop epilepsy and increased
susceptibility to acute injury as a result of excessive
extracellular glutamate levels (Tanaka et al. 1997). The
expression of this transporter has been shown recently
to be both positively and negatively regulated by NF-B
(Sitcheran et al. 2005). This study showed that the
increased binding of NF-B to the EAAT2 promoter in
H4 astroglioma cells was regulated by epidermal growth
factor (EGF), but decreased expression was caused by
TNF- inducing the classical I kappa B (IB) degradation
pathway to trigger NF-B nuclear translocation and
DNA binding to repress EAAT2 expression. In this
situation, the presence of elevated TNF- concentrations
leads to elevated extracellular glutamate concentration,
thereby increasing the risk of glutamate excitotoxicity.
Hermann et al. (2001) were the first to demonstrate
that the combination of glutamate and TNF- provoked
an amplified neurotoxic effect that was contingent on
the AMPA receptor. Interestingly, Zou & Crews (2005)
showed that in rat organotypic hippocampal slice cultures,
which possess a cytoarchitecture comparable to that in
vivo, TNF- increased glutamate neurotoxicity. They also
demonstrated that the effect was mediated by NMDA and
not AMPA receptors.
In addition to this, TNF- and glutamate have also
been implicated in -amyloid-induced microglia-related
cell death. Abundant activated microglia are prominent
in the brains of Alzheimer patients (Griffin et al. 1989)

C The Physiological Society 2005

Exp Physiol 90.5 pp 663670

TNF- and glutamate in the CNS

and are associated with -amyloid plaques (Griffin et al.


1995; Frautschy et al. 1998). It has been proposed that
inefficient phagocytosis of peptide by microglia could lead
to hyperactivation of cells and release of inflammatory
mediators and neurotoxic factors, thereby contributing to
neurodegenerative processes (Akiyama et al. 2000). It is
widely believed that the microglia play a direct role in the
neuronal death seen in Alzheimers disease. By applying
media from -amyloid-stimulated microglial cultures to
neurones, Floden et al. (2005) showed that neuronal
cell death is dependent on the synergistic co-activation
of TNF- and NMDA receptors; the NMDA receptor
antagonists memantine and 2-amino-5-phosphopetanoic
acid, as well as soluble TNF- receptor applied to the
neurones, protected them from cell death. It is interesting
that blockade of either the TNF- receptor or the NMDA
receptor alone was insufficient to induce neuronal cell
death.
There is, however, evidence that other glutamate
receptors are involved in the relationship between
neuronal cell death, glial cells and TNF-. Taylor et al.
(2005) examined the effect of metabotropic glutamate
receptor (mGluR) stimulation on TNF- release. They
found that stimulating rat primary cultured microglial
mGluRs for 24 h induced microglial activation. This in
turn induced caspase-3 activation in cerebellar granule
neurones in culture. This neurotoxicity was mediated
by TNF- released by the microglia via neuronal
TNF-R1 and caspase-3 activation (Fig. 1). Importantly,
it was the specific group II mGluR agonist 2S,2 R,3 R-2(2 ,3 -dicarboxycyclopropyl)glycine that led to the release
of TNF- and the consequent toxicity. N -acetyl-laspartyl-l-glutamate, a specific mGluR3 agonist, did not
induce microglial activation or neurotoxicity. TNF-
was only neurotoxic in the presence of microglia or
conditioned medium from microglia, a fact possibly due
to the presence of microglial-derived Fas ligand. However,
TNF--dependent neurotoxicity was prevented when the
neurones were exposed to conditioned medium from
microglia stimulated by the specific group III agonist
l-2-amino-4-phosphono-butyric acid (Taylor et al. 2005).
Glialglial interactions also play a role in this system.
Bezzi et al. (2001) showed that astrocyte glutamate
release induced by activation of the chemokine receptor
CXCR4 is accompanied by release of TNF-, and that
the TNF- release is dramatically enhanced by microglia.
In light of this evidence of glialglial and glialneuronal
interactions, it is possible to see how a cascading
glialneuronal interaction could occur, leading to cell
death. Activation of TNF- receptors on astrocytes leads
to increased extracellular glutamate concentration, which
may lead to neurotoxicity in itself, while also activating
mGluR2 on microglia, which release more TNF- in
addition to other pro-inflammatory cytokines.

C The Physiological Society 2005

665

TNF- and glutamatergic synaptic transmission

The subject of TNF- in glialneuronal interactions


also emerges when looking at glutamatergic synaptic
transmission. Beattie et al. (2002) showed that glial TNF causes an increase in surface expression of neuronal
AMPA receptors, thereby increasing synaptic efficacy.
Indeed, they showed that removal of endogenous TNF had a negative effect on AMPA receptor expression.
It has recently been shown that this AMPA receptor
exocytosis is mediated by activation of TNF-R1 through
a phosphatidylinositol 3-kinase (PI3)-dependent pathway
(Stellwagen et al. 2005). The newly expressed AMPA
receptors were shown to have lower stoichiometric
amounts of GluR2, making the receptors permeable to
calcium ions. Additionally, this study also showed a
surprising concurrent endocytosis of inhibitory GABA
receptors induced by TNF-. Furukawa & Mattson
(1998) presented further evidence that TNF- alters
glutamatergic transmission and neuronal excitability.
They showed, using whole-cell perforated patch-clamp
recording of cultured hippocampal neurones, that longterm treatment (2448 h) with TNF- caused an increase
in calcium current by 30%. Selective blockade of calcium
channel showed that this increase was largely due to
L-type calcium channels, whose currents increased by
4050%, whereas the N- and Q-type currents showed
increases of 1025%. They measured the actual calcium
concentrations using fluorescence imaging and showed
that while 24-h TNF- exposure did not alter resting
intracellular calcium concentration, it did increase the
elevated calcium concentration caused by 50 mm KCl
depolarization.
Furukawa & Mattson (1998) also showed that whole-cell
currents in these cultured hippocampal neurones induced
by glutamate, NMDA, AMPA and kainite were decreased
by 24-h exposure to TNF-. In addition, fluorescence
calcium imaging was used to show that the increase in
intracellular calcium concentration caused by application
of glutamate receptor agonists was decreased after 24-h
TNF- exposure. NF-B was also shown to be important
in regulating these responses to TNF-; no effect was
seen in response to shorter exposure to TNF-, on a
timescale suggesting possible altered gene expression,
and the effect of TNF- was abolished by co-treatment
with decoy DNA (Furukawa & Mattson, 1998). The
difference between calcium responses to depolarization
and glutamate receptor activation suggests that the effect
of TNF- is more complex than simply altering calcium
homeostasis.
The relationship between glutamatergic transmission
and TNF- was also examined in the nucleus of the
solitary tract. It has been shown that neurones excited
by gastric distension in the nucleus of the solitary tract
are excited by TNF- (Emch et al. 2000). Emch et al.

666

M. Pickering and others

(2001), using the immediate early gene product c-fos as a


marker for neuronal activation, found that c-fos expression
in that region, elicited by TNF- injection, was impaired
by AMPA and NMDA receptor blockers, suggesting that
TNF- activation of these neurones is dependent on
altered glutamate neurotransmission resulting from the
presence of TNF-.
TNF- and synaptic plasticity

These TNF--induced changes in neuronal excitability


may have important implications for synaptic plasticity
(Carroll et al. 2001). Indeed, in addition to its role in
apoptotic events, TNF- is known to act as a regulator
of synaptic plasticity. TNF- levels are elevated in several
neuropathological states that are associated with learning
and memory deficits, leading to the search for a possible
role in plasticity. To this end, much work has been

Figure 2. The p38 MAP kinase inhibitor SB 203580 reverses


early- but not late-phase-mediated inhibition of LTP by TNF-
A, SB 203580 (1 M) was added at time 10 min, TNF- (4.5 ng ml1 )
at time 40 min, and high frequency stimulation (HFS) was delivered to
the slice at time 60 min. SB 203580 reversed the inhibitory effect of
TNF- 1 h post tetanus (174 5%, versus 120 7%, n = 6;
P < 0.001). At 3 h post tetanus SB 203580 had a reduced but highly
significant antagonistic effect on TNF--mediated depression of LTP. B,
comparison of slices treated with TNF- in the presence of SB 203580
(as A) to control LTP. The rectangles represent the period of perfusion
of the designated cytokine/drug. HFS is represented by the arrow.
Figure reprinted from Butler et al. (2004). Neuroscience, 124,
319326, copyright (2004) with permission from Elsevier.

Exp Physiol 90.5 pp 663670

carried out in the hippocampus. TNF- has been shown


to regulate the development of the hippocampus, as
TNF-R1 and TNF-R2 knockout mice demonstrate
decreased arbourization of the apical dendrites of the
CA1 and CA3 regions and accelerated dentate gyrus
development (Golan et al. 2004), probably via activation
of TNF-R2, which, as mentioned earlier, does not lead to
caspase-3 activation, but is known to transduce the trophic
effect of TNF- (Yang et al. 2002).
The two forms of synaptic plasticity seen in the
hippocampus, long-term potentiation (LTP) and longterm depression (LTD), involve glutamate receptor
activation and increased intracellular calcium levels,
with induction of LTP dependent on the activation of
calciumcalmodulin kinase II, protein kinase C (PKC) and
PKA, and induction of LTD dependent on activation of
serine/threonine phosphatases 1, 2A and 2B (Mayford et al.
1995; Coussens & Teyler, 1996; Silva et al. 1997). LTP is a
long-lasting increase in synaptic efficacy, which is thought
to be an important underlying mechanism of learning and
memory formation (Bliss & Collingridge, 1993).
Several pro-inflammatory cytokines have been shown
to act as inhibitors of the induction of LTP, for example
interleukin 1 (Cunningham et al. 1996; Curran et al.
2003) and interleukin 18 (Curran & OConnor, 2001).
Like these other cytokines, TNF- has also been shown to
inhibit LTP in the CA1 and dentate gyrus regions of the rat
hippocampus at pathophysiological levels (Tancredi et al.

Figure 3. Schematic diagram of the putative role of the mGluR


and TNF-R in long-term potentiation
TNF- inhibits the early phase of LTP by activation of TNF-R1 (p55) and
is dependent on p38 activation. Additionally, TNF- causes an increase
in NF-B, which may lead to inhibition of late LTP by an as yet
unknown mechanism. Activation of the G-protein-linked mGluRs leads
to inositol-1,4,5-trisphosphate (IP3 ) receptor-mediated calcium release
via phospholipase C (PLC). TNF- may also alter calcium
concentrations. Combined activation of TNF-R1 and mGluRs may lead
to sufficiently high calcium concentrations to impair the formation of
LTP, although the exact nature of the interaction between TNF-R1 and
mGluR activation remains unknown.

C The Physiological Society 2005

Exp Physiol 90.5 pp 663670

TNF- and glutamate in the CNS

1992; Cunningham et al. 1996; Butler et al. 2004). However


there would appear to be differences in the mechanisms by
which TNF- inhibits early phase (less than 2 h) and late
phase LTP (greater than 2 h post induction (Butler et al.
2004). Using electrophysiological and immunohistological
techniques it was found that there was a major role played
by the p38 MAP kinase in the inhibitory effect of TNF- on
early LTP (Butler et al. 2004; Fig. 2). The p38 inhibitor SB
203580 reversed the effect of TNF- on early LTP without
affecting late LTP.
It has also been shown that application of TNF-
causes an increase in RGS7 (a regulator of G-protein
signalling), and that this increase is dependent on
activation of p38 MAP kinase (Benzing et al. 2002).
The RGS7 protein modulates G-protein signalling by
accelerating the intrinsic GTPase activity of G i and
Gq subunits. Increased RGS7 levels therefore leads to
increased Gq levels, giving rise to increased calcium
levels. Combined with the aforementioned evidence from
Stellwagen et al. (2005) that TNF- causes increased
expression of AMPA receptors which may be more calcium
permeable, the intracellular calcium concentrations may
become sufficiently elevated in the presence of TNF- to
contribute to the impairment of LTP.
mGLuRs may also play a role in the inhibitory
effects of TNF- on early LTP. We have recently shown
that inhibition of mGluR1 and mGluR5 can block the
TNF--dependent inhibition of early and late LTP
(Cumiskey et al. 2004). Activation of these subtypes of
mGluRs would also lead to an increase in intracellular
calcium concentration. Thus a complex and as yet
undiscovered interaction between TNF- and mGlu
receptors and the p38 MAP kinase would be required for
inhibition of LTP to occur (Fig. 3).
If the TNF--dependent inhibition of the early stage
of LTP is mediated by a p38 MAP kinase pathway and
activation of mGluRs, then it may be that the late phase
inhibition of LTP by TNF- is regulated by altered protein
synthesis (Frey et al. 1993; Huang & Kandel, 1994; Nguyen
& Kandel, 1996). Exactly how TNF- might interfere with
new protein synthesis in LTP remains to be elucidated, but
a likely candidate is the transcription factor, NF-B (Butler
et al. 2002; Fig. 3).
In addition to the effect of TNF- on LTP, it has been
shown that TNF-?receptor knockout mice demonstrate
an impairment of LTD in the CA1 region of the
hippocampus (Albensi & Mattson, 2000). The effect is
mimicked by kappa B (B) decoy DNA, which implicates
the TNF- NFB signalling pathway in LTD as well as
LTP.
It is interesting that the findings relating to the effect
of TNF- on synaptic plasticity seems to have some
behavioural correlates in vivo. TNF- knockout mice
showed increased performance in spatial memory and
learning as measured in the Morris water maze task

C The Physiological Society 2005

667

when compared to wild-type animals (Golan et al.


2004). Conversely, Aloe et al. (1999) demonstrated a
significant impairment in spatial learning in two lines
of mice that over-expressed human recombinant TNF, also using the same water maze task. However, these
results must be interpreted in the context of the study
showing developmental differences mentioned previously;
the substrate of learning and memory cannot be seen as
the same in the knockout and wild-type mice.
Concluding remarks

While undoubtedly recent work has greatly expanded


our knowledge of the role of the interactions between
the pro-inflammatory cytokine, TNF- and glutamate in
the CNS, a great many questions remain unanswered.
Inflammatory and immunological challenges, which lead
to the increase in TNF- in the CNS, will normally lead to a
coincident increase in other inflammatory mediators. The
complexity of the interactions between these mediators
should not be underestimated. It is not unlikely that as
more information about TNF- and other inflammatory
and immunological mediators active in the CNS becomes
available, interactions between these agents may manifest
as either synergistic or occlusive effects, due to convergent
or complimentary intracellular signalling pathways and
intercellular interactions. With this in mind, it is unlikely
that a full understanding of the roles of TNF- and
glutamate in the CNS can be achieved until they can be
viewed in the context of a better understanding of the
neuroimmunological system.
References
Akiyama H, Barger S, Barnum S, Bradt B, Bauer J, Cole GM
et al. (2000). Inflammation and Alzheimers disease.
Neurobiol Aging 21, 383421.
Albensi BC & Mattson MP (2000). Evidence for the
involvement of TNF and NF-kappaB in hippocampal
synaptic plasticity. Synapse 35, 151159.
Allan SM & Rothwell NJ. (2001). Cytokines and acute
neurodegeneration. Nat Rev Neurosci 2, 734744.
Aloe L, Properzi F, Probert L, Akassoglou K, Kassiotis G,
Micera A & Fiore L (1999). Learning abilities, NGF and
BDNF brain levels in two lines of TNFalpha transgenic mice,
one characterized by neurological disorders, the other
phenotypically normal. Brain Res 840, 125137.
Armstrong RC, Aja T, Xiang J, Gaur S, Krebs JF, Hoang K, Bai
X, Korsmeyer SJ, Karanewsky DS, Fritz LC & Tomaselli KJ
(1996). Fas-induced activation of the cell death-related
protease CPP32 is inhibited by Bcl-2 and by ICE family
protease inhibitors. J Biol Chem 271, 1685016855.
Barger SW, Horster D, Furukawa K, Goodman Y, Krieglstein J
& Mattson MP (1995). Tumor necrosis factors alpha and
beta protect neurones against amyloid beta-peptide toxicity:
evidence for involvement of a kappa B-binding factor and
attenuation of peroxide and Ca2+ accumulation. Proc Natl
Acad Sci U S A 92, 93289332.

668

M. Pickering and others

Beattie EC, Stellwagen D, Morishita W, Bresnahan JC, Ha BK,


Von Zastrow M, Beattie MS & Malenka RC (2002). Control
of synaptic strength by glial TNFalpha. Science 295,
22822285.
Benveniste EN & Benos DJ (1995). TNF-alpha- and
IFN-gamma-mediated signal transduction pathways: effects
on glial cell gene expression and function. FASEB J 9,
15771584.
Benzing T, Kottgen M, Johnson M, Schermer B, Zentgraf H,
Walz G & Kim E (2002). Interaction of 1433 protein with
regulator of G protein signaling 7 is dynamically regulated by
tumor necrosis factor-alpha. J Biol Chem 277,
3295432962.
Beutler B & Van Huffel C (1994a). An evolutionary and
functional approach to the TNF receptor/ligand family.
Ann NY Acad Sci 730, 118133.
Beutler B & Van Huffel C (1994b). Unraveling function in the
TNF ligand and receptor families. Science 264, 667668.
Beyaert R & Fiers W (1994). Molecular mechanisms of tumor
necrosis factor-induced cytotoxicity. What we do understand
and what we do not. FEBS Lett 340, 916.
Bezzi P, Domercq M, Brambilla L, Galli R, Schols D, De Clercq
E, Vescovi A, Bagetta G, Kollias G, Meldolesi J & Volterra A
(2001). CXCR4-activated astrocyte glutamate release via
TNF: amplification by microglia triggers neurotoxicity.
Nat Neurosci 4, 702707.
Bliss TV & Collingridge GL (1993). A synaptic model of
memory: long-term potentiation in the hippocampus.
Nature 361, 3139.
Boka G, Anglade P, Wallach D, Javoy-Agid F, Agid Y & Hirsch
EC (1994). Immunocytochemical analysis of tumor necrosis
factor and its receptors in Parkinsons disease. Neurosci Lett
172, 151154.
Boulanger LM, Huh GS & Shatz CJ (2001). Neuronal plasticity
and cellular immunity: shared molecular mechanisms.
Curr Opin Neurobiol 11, 568578.
Bruce AJ, Boling W, Kindy MS, Peschon J, Kraemer PJ,
Carpenter MK, Holtsberg FW & Mattson MP (1996). Altered
neuronal and microglial responses to excitotoxic and
ischemic brain injury in mice lacking TNF receptors.
Nat Med 2, 788794.
Butler MP, Moynagh PN & OConnor JJ (2002). Methods of
detection of the transcription factor NF-kappa B in rat
hippocampal slices. J Neurosci Methods 119, 185190.
Butler MP, OConnor JJ & Moynagh PN (2004). Dissection of
tumour-necrosis factor-alpha inhibition of long-term
potentiation (LTP) reveals a p38 mitogen-activated protein
kinase-dependent mechanism which maps to early- but not
late-phase LTP. Neuroscience 124, 319326.
Carroll RC, Beattie EC, von Zastrow M & Malenka RC (2001).
Role of AMPA receptor endocytosis in synaptic plasticity.
Nat Rev Neurosci 2, 315324.
Cheng B, Christakos S & Mattson MP (1994). Tumor necrosis
factors protect neurons against excitotoxic/metabolic insults
and promote maintenance of calcium homeostasis. Neuron
12, 139153.
Choi DW (1988). Glutamate neurotoxicity and diseases of the
nervous system. Neuron 1, 623634.
Chun J (2001). Selected comparison of immune and nervous
system development. Adv Immunol 77, 297322.

Exp Physiol 90.5 pp 663670

Chung IY & Benveniste EN (1990). Tumor necrosis factor-alpha


production by astrocytes. Induction by lipopolysaccharide,
IFN-gamma, and IL-1 beta. J Immunol 144, 29993007.
Coussens CM & Teyler TJ (1996). Protein kinase and
phosphatase activity regulate the form of synaptic plasticity
expressed. Synapse 24, 97103.
Cumiskey D, Butler MP, Moynagh PN & OConnor JJ (2004).
The inhibitory effect of tumour necrosis factor- on
long-term potentiation is attenuated by type 1 metabotropic
glutamate receptor blockade. J Physiol 560.P, C23.
Cunningham AJ, Murray CA, ONeill LA, Lynch MA &
OConnor JJ (1996). Interleukin-1 beta (IL-1 beta) and
tumour necrosis factor (TNF) inhibit long-term potentiation
in the rat dentate gyrus in vitro. Neurosci Lett 203, 1720.
Curran BP, Murray HJ & OConnor JJ (2003). A role for c-jun
n-terminal kinase in the inhibition of long-term potentiation
by interleukin-1beta and long-term depression in the rat
dentate gyrus in vitro. Neuroscience 118, 347357.
Curran B & OConnor JJ (2001). The pro-inflammatory
cytokine interleukin-18 impairs long-term potentiation and
NMDA receptor-mediated transmission in the rat
hippocampus in vitro. Neuroscience 108, 8390.
Emch GS, Hermann GE & Rogers RC (2000). TNF-alpha
activates solitary nucleus neurons responsive to gastric
distension. Am J Physiol Gastrointest Liver Physiol 279,
G582G586.
Emch GS, Hermann GE & Rogers RC (2001). TNF-alphainduced c-fos generation in the nucleus of the solitary tract is
blocked by NBQX and MK-801. Am J Physiol Regul Integr
Comp Physiol 281, R1394R1400.
Floden AM, Li S & Combs CK (2005). Beta-amyloid-stimulated
microglia induce neuron death via synergistic stimulation of
tumor necrosis factor alpha and NMDA receptors. J Neurosci
25, 25662575.
Frautschy SA, Yng F, Irrizarry M, Hyman B, Saido TC, Hsiao K
& Cole GM (1998). Microglial response to amyloid plaques
in APPsw transgenic mice. Am J Pathol 152, 307317.
Frey U, Huang YY & Kandel ER (1993). Effects of cAMP
simulate a late stage of LTP in hippocampal CA1 neurons.
Science 260, 16611664.
Furukawa K & Mattson MP (1998). The transcription factor
NF-kappaB mediates increases in calcium currents and
decreases in NMDA- and AMPA/kainate-induced currents
induced by tumor necrosis factor-alpha in hippocampal
neurons. J Neurochem 70, 18761886.
Garcia JE, Nonner D, Ross D & Barrett JN (1992). Neurotoxic
components in normal serum. Exp Neurol 118, 309316.
Gary DS, Bruce-Keller AJ, Kindy MS & Mattson MP (1998).
Ischemic and excitotoxic brain injury is enhanced in mice
lacking the p55 tumor necrosis factor receptor. J Cereb Blood
Flow Metab 18, 12831287.
Ghosh S & Karin M (2002). Missing pieces in the NF-kappaB
puzzle. Cell 109 S81S96.
Golan H, Levav T, Mendelsohn A & Huleihel M (2004).
Involvement of tumor necrosis factor alpha in hippocampal
development and function. Cerebral Cortex 14, 97105.
Goodman Y & Mattson MP (1996). Ceramide protects
hippocampal neurons against excitotoxic and oxidative
insults, and amyloid beta-peptide toxicity. J Neurochem 66,
869872.

C The Physiological Society 2005

Exp Physiol 90.5 pp 663670

TNF- and glutamate in the CNS

Goodman JC, Robertson CS, Grossman RG & Narayan RK


(1990). Elevation of tumor necrosis factor in head injury.
J Neuroimmunol 30, 213217.
Griffin WST, Sheng JG, Roberts GW & Mrak RE (1995).
Interleukin-1 expression in different plaque types in
Alzheimers disease, significance in plaque evolution,
J Neuropathol Exp Neurol 54, 276281.
Griffin WSTLC, Stanley C, Ling V, MacLeod LJ, Perrot,
White III et al. (1989). Brain interleukin-1 and S100
immunoreactivity are elevated in Down syndrome and
Alzheimer disease. Proc Natl Acad Sci U S A 86,
76117615.
Haydon PG (2001). Glia: listening and talking to the synapse.
Nat Rev Neurosci 2, 185193.
Hermann GE, Rogers RC, Bresnahan JC & Beattie MS (2001).
Tumor necrosis factor-alpha induces cFOS and strongly
potentiates glutamate-mediated cell death in the rat spinal
cord. Neurobiol Dis 8, 590599.
Huang YY & Kandel ER (1994). Recruitment of long-lasting
and protein kinase A-dependent long-term potentiation in
the CA1 region of hippocampus requires repeated
tetanization. Learn Mem 1, 7482.
Israel A (2000). The IKK complex: an integrator of all
signals that activate NF-kappaB? Trends Cell Biol 10,
129133.
Kinouchi K, Brown G, Pasternak G & Donner DB (1991).
Identification and characterization of receptors for tumor
necrosis factor-alpha in the brain. Biochem Biophys Res
Commun 181, 15321538.
Kolesnick R & Golde DW (1994). The sphingomyelin pathway
in tumor necrosis factor and interleukin-1 signaling. Cell 77,
325328.
Kronke M, Schutze S, Scheurich P, Meichle A, Hensel G,
Thoma B, Kruppa G & Pfizenmaier K (1990). Tumour
necrosis factor signal transduction. Cell Signal 2,
18.
Li Q & Verma IM (2002). NF-kappaB regulation in the
immune system. Nat Rev Immunol 2, 725734.
Lieberman AP, Pitha PM, Shin HS & Shin ML (1989).
Production of tumor necrosis factor and other cytokines by
astrocytes stimulated with lipopolysaccharide or a
neurotropic virus. Proc Natl Acad Sci U S A 86,
63486352.
Liu T, Clark RK, McDonnell PC, Young PR, White RF, Barone
FC & Feuerstein GZ (1994). Tumor necrosis factor-alpha
expression in ischemic neurons. Stroke 25, 14811488.
Mattson MP, Barger SW, Furukawa K, Bruce AJ, Wyss-Coray
T, Mark RJ & Mucke L (1997a). Cellular signaling roles of
TGF beta, TNF alpha and beta APP in brain injury responses
and Alzheimers disease. Brain Res Brain Res Rev 23, 4761.
Mattson MP, Goodman Y, Luo H, Fu W & Furukawa K
(1997b). Activation of NF-kappaB protects hippocampal
neurons against oxidative stress-induced apoptosis: evidence
for induction of Mn-SOD and suppression of peroxynitrite
production and protein tyrosine nitration. J Neurosci Res 49,
681697.
Mayford M, Wang J, Kandel ER & ODell TJ (1995). CaMKII
regulates the frequencyresponse function of hippocampal
synapses for the production of both LTD and LTP. Cell 81,
891904.

C The Physiological Society 2005

669

Morganti-Kossman MC, Lenzlinger PM, Hans V, Stahel P,


Csuka E, Ammann E, Stocker R, Trentz O & Kossmann T
(1997). Production of cytokines following brain injury:
beneficial and deleterious for the damaged tissue.
Mol Psychiatry 2, 133136.
Nath R, Robert A, McGinnis KM & Wang KKW (1998).
Evidence for activation of caspase-3-like protease in
excitotoxins and hypoxia/hypoglycemia-injured
cerebrocortical neurons. J Neurochem 71,
186195.
Nguyen PV & Kandel ER (1996). A macromolecular
synthesis-dependent late phase of long-term potentiation
requiring cAMP in the medial perforant pathway
of rat hippocampal slices. J Neurosci 16,
31893198.
ONeill LA & Kaltschmidt C (1997). NF-kappa B: a crucial
transcription factor for glial and neuronal cell function.
Trends Neurosci 20, 252258.
Rieckmann P, Albrecht M, Kitze B, Weber T, Tumani H,
Broocks A, Luer W, Helwig A & Poser S (1995). Tumor
necrosis factor RNA expression in patients with
relapsing-remitting multiple sclerosis is associated with
disease activity. Ann Neurol 37, 8288.
Rothe J, Gehr G, Loetscher H & Lesslauer W (1992). Tumor
necrosis factor receptors structure and function. Immunol
Res 11, 8190.
Selmaj KW & Raine CS (1998). Tumor necrosis factor mediates
myelin and oligodendrocyte damage in vitro. Ann Neurol 23,
339346.
Silva AJ, Smith AM & Giese KP (1997). Gene targeting and the
biology of learning and memory. Annu Rev Genet 31,
527546.
Silverman N & Maniatis T (2001). NF-kappaB signaling
pathways in mammalian and insect innate immunity. Genes
Dev 15, 23212342.
Sitcheran R, Gupta P, Fisher PB & Baldwin AS (2005). Positive
and negative regulation of EAAT2 by NF-kappaB: a role for
N-myc in TNFalpha-controlled repression. EMBO J 24,
510520.
Stellwagen D, Beattie EC, Seo JY & Malenka RC (2005).
Differential regulation of AMPA receptor and GABA
receptor trafficking by tumor necrosis factor-alpha.
J Neurosci 25, 32193228.
Tanaka K, Watase K, Manabe T, Yamada K, Watanabe M,
Takahashi K et al. (1997). Epilepsy and exacerbation of brain
injury in mice lacking the glutamate transporter GLT-1.
Science 276, 16991702.
Tancredi V, DArcangelo G, Grassi F, Tarroni P, Palmieri G,
Santoni A & Eusebi F (1992). Tumor necrosis factor alters
synaptic transmission in rat hippocampal slices. Neurosci
Lett 146, 176178.
Tartaglia LA, Weber RF, Figari IS, Reynolds C, Palladino MA Jr
& Goeddel DV (1991). The two different receptors for tumor
necrosis factor mediate distinct cellular responses. Proc Natl
Acad Sci U S A 88, 92929296.
Taylor DL, Jones F, Kubota ES & Pocock JM (2005).
Stimulation of microglial metabotropic glutamate receptor
mGlu2 triggers tumor necrosis factor alpha-induced
neurotoxicity in concert with microglial-derived Fas ligand.
J Neurosci 25, 29522964.

670

M. Pickering and others

Volterra A & Steinhauser C (2004). Glial modulation of


synaptic transmission in the hippocampus. Glia 47, 249257.
Wajant H & Scheurich P (2001). Tumor necrosis factor receptor
associated factor (TRAF) 2 and its role in TNF signaling.
Int J Biochem Cell Biol 33, 1932.
Wang KKW, Nath R, Raser KJ & Hajimohammadreza I (1996).
Maitotoxin induces calpain activation in SH-SY5Y
neuroblastoma cells and cerebrocortical cultures. Arch
Biochem Biophys 331, 208214.
Wang X, Yue TL, Barone FC, White RF, Gagnon RC &
Feuerstein GZ (1994). Concomitant cortical expression of
TNF-alpha and IL-1 beta mRNAs follows early response gene
expression in transient focal ischemia. Mol Chem
Neuropathol 23, 103114.

Exp Physiol 90.5 pp 663670

Yang L, Lindholm K, Konishi Y, Li R & Shen Y (2002). Target


depletion of distinct tumor necrosis factor receptor subtype
reveals hippocampal neuron death and survival through
different signal transduction pathways. J Neurosci 22,
30253032.
Zhao X; B, Bausano BR, Pike JK, Newcomb-Fernandez KK,
Wang E, Shohami NC, Ringger SM, DeFord DK, Anderson &
Hayes RL (2001). TNF-alpha stimulates caspase-3 activation
and apoptotic cell death in primary septo-hippocampal
cultures. J Neurosci Res 64, 121131.
Zou JY & Crews FT (2005). TNF alpha potentiates glutamate
neurotoxicity by inhibiting glutamate uptake in organotypic
brain slice cultures: neuroprotection by NF kappa B
inhibition. Brain Res 1034, 1124.


C The Physiological Society 2005

You might also like