You are on page 1of 18

J Neuroimmune Pharmacol (2012) 7:42–59

DOI 10.1007/s11481-011-9287-2

INVITED REVIEW

Tumor Necrosis Factor-alpha and the Roles it Plays


in Homeostatic and Degenerative Processes
Within the Central Nervous System
Sara L. Montgomery & William J. Bowers

Received: 1 June 2011 / Accepted: 20 June 2011 / Published online: 5 July 2011
# Springer Science+Business Media, LLC 2011

Abstract Tumor Necrosis Factor-alpha (TNF-α) is a nervous system (CNS). During states of chronic neuro-
prototypic pro-inflammatory cytokine involved in the inflammation, extensive experimental evidence implicates
innate immune response. TNF-α ligation and downstream TNF-α as a key mediator in disease progression, gliosis,
signaling with one of its cognate receptors, TNF-RI or demyelination, inflammation, blood–brain-barrier deterio-
TNF-RII, modulates fundamental processes in the brain ration, and cell death. This review explores the complex
including synapse formation and regulation, neurogenesis, roles of TNF-α in the CNS under normal physiologic
regeneration, and general maintenance of the central conditions and during neurodegeneration. We focus our
discussion on Multiple Sclerosis, Parkinson’s disease, and
Sources of Support: This work was supported by NIH F31-AG038063 Alzheimer’s disease, relaying the outcomes of preclinical
to SLM and NIH R01-AG023593 and NIH R01-AG026328 to WJB. and clinical testing of TNF-α directed therapeutic strate-
S. L. Montgomery gies, and arguing that despite the wealth of functions
Department of Pathology and Laboratory Medicine, attributed to this central cytokine, surprisingly little is
University of Rochester Medical Center,
Rochester, NY 14642, USA
known about the cell type- and stage-specific roles of TNF-
α in these debilitating disorders.
W. J. Bowers
Departments of Neurology, Keywords Neuroinflammation . Cytokine . Development .
University of Rochester Medical Center,
Rochester, NY 14642, USA
Alzheimer’s disease . Parkinson’s disease . Multiple
Sclerosis . Intracellular signaling
W. J. Bowers
Microbiology and Immunology,
University of Rochester Medical Center,
Rochester, NY 14642, USA
Introduction

W. J. Bowers Tumor necrosis factor-alpha (TNF-α) is a critical regulatory


Pharmacology and Physiology, cytokine exerting both homeostatic and pathophysiological
University of Rochester Medical Center,
Rochester, NY 14642, USA
roles in the central nervous system (CNS). This potent
cytokine drives acute and chronic inflammatory responses,
S. L. Montgomery : W. J. Bowers the former of which are normally self-limiting and leads to
Center for Neural Development and Disease, the removal of injurious stimuli and restoration of homeo-
University of Rochester Medical Center,
Rochester, NY 14642, USA
stasis. However, sustained inflammation for protracted
periods of time may result in cellular dysfunction and
W. J. Bowers (*) initiation of disease. Chronic inflammation, as evidenced by
Department of Neurology, the elaboration of proinflammatory marker expression,
Center for Neural Development and Disease,
University of Rochester Medical Center,
elevated cytokine and chemokines levels, and the prolifer-
601 Elmwood Ave., Box 645, Rochester, NY 14642, USA ation and/or clustering of glial cells within afflicted brain
e-mail: william_bowers@urmc.rochester.edu regions, are characteristic hallmarks of a number of
J Neuroimmune Pharmacol (2012) 7:42–59 43

neurodegenerative diseases. Although 36 years have passed sTNF and tmTNF are biologically active and exert their
since TNF-α was first discovered by Carswell and distinct signaling functions through engagement of two
colleagues (Carswell et al. 1975), current strategies for cognate single-pass transmembrane glycoprotein receptors:
TNF-α intervention in the human brain appear to lack the TNF-receptor 1 (TNF-RI) and TNF-receptor 2 (TNF-RII).
robustness and specificity necessary to ameliorate disease. At the post-translational level, adenine-uracil rich elements
This review will summarize the homeostatic and pathologic (ARE’s) located within the 3′ untranslated region (Fig. 1)
consequences imparted by TNF-α expression within the target TNF-α mRNA for translational degradation, thereby
CNS and will discuss the current views on the potential preventing excessive cytokine production in the absence of
therapeutic promise of anti-TNF-α therapeutics in preventing an inflammatory trigger (Kontoyiannis et al. 1999). Inflam-
and/or combating neurodegenerative diseases. matory mediators, such as prostaglandins and corticoste-
roids, downregulate TNF-α transcription and/or translation
to limit the duration of active TNF-α signaling (Huang and
TNF-α signaling Eden 1993; Schottelius et al. 1999; Yamane et al. 2000;
Shinomiya et al. 2001).
TNF-α belongs to the large TNF ligand superfamily and is The two cognate TNF-α receptors, TNF-RI and TNF-
generally produced as a type II integral membrane protein RII, are differentially expressed and controlled. TNF-RI is
(Wajant et al. 2003). Synthesis of this highly multifunc- constitutively expressed on most cell types, with the
tional cytokine occurs in a broad range of cell types, exception of erythrocytes. Conversely, TNF-RII expression
including immune regulatory macrophages, T cells, mast is induced upon inflammatory stimulation and is limited to
cells, granulocytes, natural killer cells and non-immune hematopoietic lineage cells, including microglia and endo-
endothelial cells, osteoblasts, smooth muscle cells, fibro- thelial cells. Due to its relatively restricted expression, it has
blasts, and keratinocytes. Within the central nervous been generally accepted that fewer TNF-α related signaling
system, microglia, astrocytes and neurons are major sources processes are driven by TNF-RII (Grell 1995; McCoy and
of TNF-α (Lieberman et al. 1989; Liu et al. 1994; Tansey 2008). TNF-RI and TNF-RII share limited sequence
Morganti-Kossman et al. 1997; Lau and Yu 2001; Hanisch homology (28%) overall, with much of this homology
2002; Klintworth et al. 2009). being shared within a cysteine-rich ectodomain (Aggarwal
TNF-α synthesis is stringently regulated and can be et al. 2000). TNF-RI and TNF-RII can bind either sTNF or
triggered by bacterial and viral pathogens, injury and tmTNF, but particular ligand-receptor combinations are
immunological stimuli including cytokines, complement preferred. Secreted sTNF binds with high affinity to TNF-
factors, and immune complexes. TNF-α is regulated at the RI and TNF-RII, but dissociates from TNF-RII much more
transcriptional, post-transcriptional, and translational level. readily, supporting the ligand-passing hypothesis in which
Transcriptionally, the TNF-α gene is controlled at its 5′ sTNF binds to TNF-RII then quickly dissociates and
flanking region that contains three κβ- like enhancers (four transfers to TNF-RI. This mechanism leads to increased
in mouse) and a MHC class II-like “Y box” (Fig. 1) (Drouet local concentrations of TNF-α and rapid signal transmis-
et al. 1991). Several transcription factors are capable of sion via TNF-RI (Tartaglia et al. 1993a). Similarly, pairing
binding and activating the TNF-α promoter, including NF- of TNF-RII with the tmTNF ligand is favorable for highly
κβ, CCAAT/enhancer binding protein β (C/EBPβ), SP-1, robust TNF-RII signaling activities (Grell et al. 1995).
Erg-1, and c-Jun (Pope et al. 1994; Wedel et al. 1996; Yao et Ectodomain shedding of TNF-RI and TNF-RII through
al. 1997; Udalova et al. 1998). TNF-α mRNA translation proteolytic cleavage can also occur, whereby these receptor
leads to the generation of a 26-kDa precursor fragments act as neutralizing agents that inhibit the ability
transmembrane-associated TNF-α (tmTNF) protein. This of TNF-α to efficiently engage with functional membrane-
pro-TNF-α is proteolytically processed by the TNF-α bound receptors (Wallach et al. 1991).
converting enzyme (TACE), a matrix metalloproteinase, to Cell type specificity of TNF-α receptor expression,
release a soluble 17-kDa TNF-α (sTNF) homotrimer. Both ligand affinity, and cellular composition within a given

1 2 3 4
5 3 UTR
5 UTR 3

Fig. 1 Schematic representation of the TNF-α gene structure located encode the signal peptide sequence (Nedwin et al. 1985). The 3′ UTR
on human chromosome 6. The TNF-α genomic locus is approximately harbors an AU-rich element (ARE), while the 5′ UTR comprises
3 kb in length and contains 4 coding exons interrupted by 3 introns. Solid regulatory transcriptional elements including κβ enhancers and a Y-box
squares represent exons and introns are depicted by the horizontal line. similar to that of the MHC class II gene (Idriss and Naismith 2000)
Over 80% of exon 4 encodes for the secreted protein, while exons 1 and 2
44 J Neuroimmune Pharmacol (2012) 7:42–59

tissue are factors that significantly influence the types of death domain, is able to directly trigger TNF-α exposure-
cellular responses elicited by TNF-α. Historically, TNF-RI related cell death (Heller et al. 1992), and Haridas and
engagement was believed to promote apoptosis and colleagues demonstrated TNF-RII overexpression leads to
cytotoxicity, while cell survival, proliferation and protective the induction of TNF-α-dependent apoptosis (Haridas et al.
cellular responses elicited by TNF-α were attributed 1998). More recently, it has been reported that TNF-RII can
primarily to TNF-RII binding. The complexity and pleio- activate apoptosis independently of TNF-RI in PVC60 cells
tropic activities of TNF-α in vivo have become better and cell death can be blocked via caspase 1 and 8 inhibition,
appreciated during the past decade. The signaling cascades implicating the upstream FADD adaptor protein in TNF-
underlying TNF-α function include activation of the highly RII-mediated cell death mechanisms (Depuydt et al. 2005).
characterized NFκβ, p38 MAP kinase, c-jun N-terminal TNF-RII signaling has been shown to be protective in a
kinase (JNK), and sphingosine-based ceramide pathways considerable number of disease processes, including heart
(Hsu et al. 1995; Rothe et al. 1995a; Natoli et al. 1997; disease, neurodegenerative disorders, ulcerative colitis and
Kronke 1999). Prior to TNF-α binding, the extracellular Crohn’s disease, type 1 diabetes, retinal ischemia, and
pre-ligand-binding assembly domain (PLAD) allows for the familial rheumatoid arthritis (Shen et al. 1997; Weiss et al.
trimerization of the TNF-receptors, and this event is critical 1998; Barton et al. 2001; Dieude et al. 2002; Sashio et al.
for TNF-α signaling and activation. Ligation of TNF-α 2002; Marchetti et al. 2004; Pierik et al. 2004; Monden et
occurs within the ligand-binding pockets of cysteine-rich al. 2007). In retinal ischemia, loss of TNF-RI resulted in
domains 2 (CDR2) and 3 (CDR3) of the extracellular reduced neurodegeneration, while depletion of TNF-RII
segments of the TNF receptors, resulting in signal transduction increased neuronal cell loss, suggesting possible opposing
at the cell surface and ligand-receptor clustering (Banner et al. roles for TNF-RI and TNF-RII in cytotoxicity and
1993; MacEwan 2002). Both TNF-RI and TNF-RII lack protection, respectively (Fontaine et al. 2002). TNF-RII is
enzymatic activity within their respective intracellular critical for maintaining the oligodendrocyte progenitor cell
domains, and therefore, signal via intracellular adaptor pool in a cuprizone model of demyelination (Arnett et al.
molecule aggregation (Magnusson and Vaux 1999). The 2001) and hippocampal neurons are sensitive to TNF-α
existence of a cytoplasmic protein-protein interacting death toxicity in the absence of TNF-RII, but not TNF-RI (Yang
domain within TNF-RI mediates the induction of apoptosis et al. 2002). These differential effects of TNF-RI and TNF-
and NFκβ signaling (Tartaglia et al. 1993b). Adaptor proteins RII are dependent upon cell type, environment, age and the
bind following the dissociation of the silencer of death activation status of those cells.
domain (SODD), a negative regulator that associates with the tmTNF can trigger TNF-α signaling by acting as both a
death domain. Removal of SODD leads to the aggregation ligand and as a receptor. Reverse signaling, where membrane-
and recruitment of the TNF receptor-associated death domain integrated TNF-α can mitigate intracellular signaling and
(TRADD) (Magnusson and Vaux 1999). TRADD serves as a function as a receptor, allows for bidirectional communication
signaling platform and recruits downstream signaling mole- between cells (Fig. 2). Receptor-mediated tmTNF binding
cules, including the Fas-associated death domain (FADD), with TNF-α antagonists or TNF-receptor-expressing cells
TNF-receptor associated factor 2 (TRAF2) and receptor can provoke NFκB activation or programmed cell death, as
interacting protein (RIP) to form the active TNF-RI signaling well as down-regulate sTNF-α, IL-1, IL-6, and IL-10
complex. Recruitment of cellular inhibitors of apoptosis cytokine secretion (Eissner et al. 2000; Harashima et al.
(cIAP) 1 and 2 to the TNF-RI signaling complex potentiates 2001; Zhang et al. 2008). Reverse signaling leads to an
signal activation through NFκβ, p38, JNK, and ceramide/ enhancement of intracellular calcium and activation of p38
sphingomyelinase pathways (Fig. 2) (Rothe et al. 1995a; mitogen protein kinase (Watts et al. 1999; Waetzig et al.
Winston et al. 1995; Wang et al. 1998; Lee et al. 2003). 2002). The function of reverse signaling is important during
These same pathways, including those leading to immune responses such as T cell activation, formation of
apoptosis, can be induced through TNF-RII signaling. germinal centers, and production of specific immunoglobulin
While TNF-RI indirectly recruits TRAF2, TNF-RII directly isotypes that are important in pathogen elimination and
associates with this adaptor protein along with TRAF1 to allergic reactions (van Essen et al. 1995; Boursalian and Fink
elicit intracellular crosstalk between the receptors (Rothe et 2003). The regulation of reverse signaling appears to be
al. 1994; Rothe et al. 1995b). Notably, when TNF-RI and mediated, at least in part, by post-translational modification
TNF-RII are co-stimulated an additive effect of NFκβ of tmTNF-α. Casein Kinase 1 (CK1) phosphorylates a
transduction is observed, where it is likely both TNF- conserved sequence in the cytoplasmic tail of TNF-related
receptor pathways share similar signaling proteins. Inter- family members, including TNF-α (Pocsik et al. 1995), and
estingly, cooperativity between the two receptor types has this phosphorylation event has been shown to alter tmTNF-
been shown to significantly enhance TNF-RI-induced α-mediated reverse signaling activity in monocytes (Watts et
cytotoxicity (Weiss et al. 1997). TNF-RII, which lacks a al. 1999; Eissner et al. 2004). Friedmann and colleagues
J Neuroimmune Pharmacol (2012) 7:42–59 45

Fig. 2 Soluble and TNF-producing cell


transmembrane TNF-α signaling
through TNF-RI and TNF-RII. Reverse Signaling
tmTNF-α forms a trimeric
structure and is proteolytically
cleaved by TACE to release the
soluble homotrimeric TNF-α
protein. Both tmTNF-α and
sTNF-α bind receptors of the tmTNF TACE sTNF
TNF-receptor superfamily, Trimer
TNF-RI and TNF-RII, although
sTNF is primarily restricted to
TNF-RI and tmTNF associates TNF-RII TNF-RI
more prominently with TNF-RII.
A cytoplasmic death domain of
TNF-RI allows for the binding of
TRADD and the recruitment of
FADD to induce apoptosis. The
sphinomyelinase/ceramide
pathway is triggered via binding T
R FAN
the TNF-RI adaptor protein, FAN. A TRAF2 cIAP
F T SMase/
TRAF2 of TNF-RI or TNF-RII 1 R
A Ceramide
leads to the downstream cIAP F TRADD
activation of NFκβ, JNK, and 2 FADD
p38 signaling pathways and
MEKK
elicits diverse TNF-mediated
biological responses
RIP Apoptosis

JNK MKK3 NEMO

p38

Iκβ
cJun/AP-1
ATFs NFκβ

TNF-responsive cell
Activation

reported that in activated dendritic cells two transmembrane development is the extensive apoptotic cell death that
aspartyl signal peptide peptidases (SPPLs) promote proteo- occurs when neurons do not receive sufficient neurotrophic
lytic cleavage of the intracellular domain of tmTNF leading support during the process of target innervation (Oppenheim
to the induction of the pro-inflammatory cytokine, IL-12 1996; Pettmann and Henderson 1998). TNF-α-mediated
(Weihofen et al. 2002; Friedmann et al. 2006). The apoptosis during this period was proposed by Zhao et al. In
identification and involvement of other kinases, phospha- their study, which employed septo-hippocampal neuron
tases, or proteases that may act to modulate tmTNF reverse cultures, TNF-α induced death of neurons by stimulating
signaling continues to be a work in progress (Watts et al. caspase-3, and ultimately led to the activation of apoptotic
1999; Eissner et al. 2004). machinery (Zhao et al. 2001). Sympathetic and nociceptive
sensory neurons require specific neurotrophic signaling from
nerve growth factor (NGF) to prevent programmed cell
TNF-α in the CNS death. Barker and colleagues demonstrated that TNF-α
exacerbates neuronal apoptosis when neurons are deprived
Development of NGF. In the same study, cell death was blocked with the
treatment of anti-TNF-α or TNF-RI antibodies in cultures of
TNF-α plays important roles during the development of the NGF-dependent embryonic neurons from the trigeminal
mammalian CNS. One important characteristic of CNS ganglia (Barker et al. 2001). Intrinsic and extrinsic factors
46 J Neuroimmune Pharmacol (2012) 7:42–59

have been reported to mediate cell death of motoneurons. In altered hippocampal development whereby dentate gyrus
rat explants, there appears to be a critical period for maturation is quicker and dendritic tree arborization of the
motoneuron commitment to cell death. Prior to this critical CA1 and CA3 sub-regions are significantly retarded, which
period, between E12 and E13, motoneurons transiently may be due to the ability of TNF-α to modulate growth
express TNF-RI, and when exposed to macrophage- factors that influence pyramidal cell migration of the
secreted TNF-α, motoneurons undergo programmed cell hippocampus (Golan et al. 2004). Interestingly, astrocytes
death, suggesting that TNF-α acts to initiate apoptosis in this may promote these morphological alterations given that
setting (Sedel et al. 2004). astrocytic GFAP-deficient mice harbor neurons comprised
Cell-fate determination during the differentiation of of dense and elongated neurites, possibly speaking to a role
neural stem cells into specific neuronal and glial cell for TNF-α in astrocyte differentiation as mentioned
lineages, as well as migration and proliferation, are highly previously (Menet et al. 2000; Keohane et al. 2010).
orchestrated processes that are central to normal develop- Nevertheless, data also support TNF-α as a pro-
ment. TNF-α is one of many soluble factors that influences neurogenic cytokine. At low concentrations, TNF-α pro-
neuronal maturation, survival, and function, although the motes neurogenesis and axonogenesis of subventricular
specific mechanisms by which TNF-α mediates these zone (SVZ) cultures and this response appears to be
processes are somewhat unresolved due to conflicting dependent upon TNF-RI engagement with its ligand
results in the published literature. Keohane et al. reported (Bernardino et al. 2008). Obregon and colleagues further
embryonic neural progenitor cell (NPC) proliferation is alluded to a pro-neurogenic effect of TNF-α such that when
unaffected by TNF-α, while cell lineage fate differentiation TNF-α is neutralized with antibodies, immature neuronal
is anti-neurogenically skewed resulting in a loss of cells lines are incapable of differentiating into mature
embryonic neurons and an elevation of newly born neurons (Fig. 3) (Obregon et al. 1999). TNF-α has also
astrocytes (Fig. 3) (Keohane et al. 2010). The authors been shown to alter the cellular morphology of hippocam-
posited that TNF-α differentially regulates cell proliferation pal neurons by increasing cell body size and reducing
and differentiation and these differences may be due to neurite outgrowth (Neumann et al. 2002). Taken together,
upregulated HES1 gene expression, an anti-neurogenic these data suggest that there is a subtle line between TNF-
factor, and increased TNF-receptor expression subsequent α-induced pro- and anti-neurogenic function, which may be
to the addition of TNF-α on differentiating NPCs. contingent on local TNF-α concentrations, timing, receptor
Similarly, Liu et al. reported TNF-α negatively affects cell expression, and cellular location and context.
fate determination of embryonic rat NPCs into neurons (Liu
et al. 2005b). Furthermore, TNF-α knockout mice exhibit Synaptic transmission

Synaptic plasticity augments activity-dependent modifica-


Neural Stem
Cell (NSC) tions of neuronal networks by rapidly adjusting individual
synapses in response to synaptic activity, as measured via
alterations in long-term potentiation (LTP) and long-term
Self-renewal depression (LTD) reviewed by (Bear and Malenka 1994).
TNF-α
Another process regulating synaptic plasticity is a homeo-
Progenitor Cells
static mechanism called “synaptic scaling”. Chronic or
prolonged activity can induce neurons to alter the strength
Glial Progenitor Neuronal Progenitor of all synapses thereby allowing for the maintenance of
stably functioning circuits (Turrigiano et al. 1998). Pub-
lished evidence supports a role for TNF-α in synaptic
Committed Cells plasticity. For instance, TNF-α increases α-amino-3-hydroxy-
TNF-α 5-methyl-4-isoxazolepropionic acid (AMPA) glutamate re-
ceptor expression leading to enhanced synapse efficacy
and rapid synaptic transmission. AMPA receptor insertion
is dramatically increased when hippocampal neurons in
Oligodendrocyte Astrocyte Neuron
culture are treated with TNF-α, and abrogation of TNF-α
signaling downregulates AMPA receptors (Beattie et al.
Fig. 3 Neural progenitor cell differentiation and maturation is TNF-α 2002). Furthermore, in response to a prolonged absence of
dependent. TNF-α has been shown to be anti-neurogenic during cell
lineage fate determination, resulting in an increased numbers of newly
neural activity, TNF-α induces synaptic scaling and relies
born astrocytes and TNF-α is required for neuron maturation in on glial cell-secreted TNF-α (Stellwagen and Malenka
specific contexts 2006). Other reports suggest that TNF-α is imperative for
J Neuroimmune Pharmacol (2012) 7:42–59 47

the maintenance of synaptic plasticity, and if TNF-α inflammation has been reported to significantly reduce
signaling is neutralized for more than 12 h, neuronal neurogenesis, and administration of an anti-inflammatory
synapses are refined, where scaffold proteins important for antibiotic can reverse these suppressive actions (Ekdahl et
the make-up of the postsynaptic density are re-organized al. 2003). Since human NSCs express both TNF-RI and
leading to suboptimal synaptic connectivity and a reduction in TNF-RII, these cells can be influenced by TNF-α through
sustained synaptic scaling (Steinmetz and Turrigiano 2010). autocrine and paracrine actions (Sheng et al. 2005). TNF-α
N-methyl-D-aspartate (NMDA) receptors are also regulated has an anti-neurogenic effect during adult neurogenesis
by TNF-α. Wheeler et al. demonstrated that through whereby in culture, microglia-secreted TNF-α is detrimen-
increased ceramide by spingomyelinase-2 regulation, tal to hippocampal progenitor cells (HPC) by abruptly
NMDA surface localization was enhanced (Wheeler et al. halting cell division and leading to progenitor cell death
2009). In addition to glutamate receptor expression, calcium (Cacci et al. 2005). In contrast, others have shown striatal
homeostasis is modulated by TNF-α, where type-1 inositol and hippocampal neurogenesis is compromised when an
1,4,5-trisphosphate receptors that are involved in calcium antibody to TNF-α is transiently infused into the lateral
homeostasis and long-term potentiation mechanisms are ventricle of a rat stroke model. This indicates that
influenced by TNF-α, particularly through JNK signaling subsequent to stroke, TNF-α can encourage the survival
(Fujii et al. 2000; Park et al. 2008). of neural progenitor cells and the authors propose a
The synapse represents a vital neuronal structure and possible role for TNF-RII due to the reported neuro-
progressive loss of the circuitry is a common theme in protective roles for TNFα engagement to this receptor
neurodegenerative diseases. Moreover, reduction in synapse (Marchetti et al. 2004; Heldmann et al. 2005). Iosif et al.
density has been shown to represent the best correlate to demonstrated that TNF-RI negatively regulates adult neuro-
disease progression, especially in the case of AD and brain genesis of the hippocampus where TNF-RI or RI/RII
injury (DeKosky and Scheff 1990; Scheff et al. 2006; Ding knockout mice exhibit an increased number of adult
et al. 2009). Hermann and colleagues determined that hippocampal neurons following status epilepticus (Iosif et
during injury, TNF-α in cooperation with glutamate, al. 2006). In addition, during ischemic stroke, binding of
promotes neuronal excitotoxicity due to the excessive and TNF-α to TNF-RI suppresses SVZ progenitor proliferation
rapid surface expression of AMPA receptors leading to (Iosif et al. 2008). These data demonstrate TNF-α has
increased intracellular calcium (Hermann et al. 2001). In positive and negative effects during adult neurogenesis,
addition, Ca2+ permeable AMPA/kainite channels of hippo- which are dependent upon induction context and receptor
campal CA1 and CA3 neurons are upregulated when sub-type engagement.
exposed to TNF-α, possibly indicating that during injury
or disease TNF-α may exacerbate damage (Ogoshi et al.
2005). In aggregate, these studies suggest that TNF-α is TNF-α in neurodegenerative disease
critical in the normal brain for proper synaptic strength, but
that alteration in the level of this molecule, such as during Multiple sclerosis
neurodegeneration and traumatic brain injury, results in
acute neuronal disruption and potential chronic dysfunction Multiple sclerosis (MS) is an inflammatory autoimmune
that may ultimately compromise the viability of neurons. disease of the CNS with the clinical manifestation of focal
demyelinating plaques, progressive axonal loss, and gliosis
Adult neurogenesis (Trapp et al. 1998; Lassmann et al. 2007). Formation of
white matter lesions trigger infiltration of systemic lym-
Neural stem cells (NSCs) are present in the adult human phocytes, such as Th1 and Th17 cells, macrophages and
brain, and new neurons are continuously generated brain-resident microglia resulting in a T cell mediated
throughout an individual’s lifetime (Stemple and Anderson autoimmune attack against myelin peptide that eventually
1992; Temple 2001). Two highly neurogenic regions of the leads to widespread demyelination. Pathological hallmarks
adult brain include the subgranular zone (SGZ) of the of MS are stage-dependent, where approximately 80% of
dentate gyrus that gives rise to intermediate hippocampal MS patients are diagnosed with relapsing/remitting MS and
progenitors and the subventricular zone (SVZ) encompass- 65% of these patients follow a progressive phase called
ing the lateral ventricles that give rise to olfactory bulb “secondary progressive”. Relapsing MS is accompanied by
neurons (Lie et al. 2004; Deng et al. 2010). Chemokines an inflammatory reaction and the formation of new
and cytokines exert critical functions in modulating NSCs demyelinating lesions. In contrast, progressive MS includes
whereby some of these factors may induce, while others gray and white matter atrophy with limited genesis of new
impair, neurogenesis. In general, inflammation can restrict inflammatory lesions (Sanfilipo et al. 2006; Lassmann et al.
the generation of new neurons. Specifically, LPS-induced 2007). Inflammatory responses are the primary mechanism
48 J Neuroimmune Pharmacol (2012) 7:42–59

leading to myelin destruction and olidgodendrocyte cell TNF-α antibody was infused into two human patients
death. Pro-inflammatory TNF-α is detected upon post- exhibiting rapidly progressing disease. As a result of
mortem examination of MS brain lesions, is abnormally transient pan-TNF-α inhibition, magnetic resonance imag-
elevated in the CSF of patients, and the levels of this ing (MRI) revealed increased gadolinium-enhancing lesions
cytokine correlate with disease severity and progression and lymphocyte numbers indicating exacerbation of disease
(Beck et al. 1988; Sharief and Hentges 1991) (Hofman et (van Oosten et al. 1996). Subsequently, in a double-blinded,
al. 1989). In MS, TNF-α is implicated in myelin and placebo controlled, multicentered phase II study, 168
oligodendrocyte deterioration, lymphocyte infiltration, as- relapse-remitting MS patients were administered lenercept,
trocyte activation, and the upregulation of major histocom- a sTNF-RI fusion protein that neutralizes TNF-α. Com-
patibility complex (MHC) I and II molecules on CNS- pared to placebo control patients, lenercept-treated individ-
resident cells, thereby triggering T cell responses (reviewed uals experienced higher occurrence of relapse and increased
in Constantinescu 2010). neurological deficit (1999). Moreover, anti-TNF-α agents
Several animal model studies of MS suggest TNF-α used to treat other inflammatory diseases such as psoriatic
signaling leads to demyelination, increased inflammatory arthritis, ankylosing spondylitis, and rheumatoid arthritis
status, and compromised oligodendrocyte health. In fact, have also been associated with demyelination that closely
when TNF-α is overexpressed in the mouse brain, chronic resembles that observed in multiple sclerosis (Mohan et al.
inflammation including reactive gliosis, T cell infiltration, 2001; Titelbaum et al. 2005; Winkelmann et al. 2008). The
and demyelination occurs leading to premature death. ineffectiveness of anti-TNF-α therapy in MS may be a
Remarkably, this phenotype is rescued with a TNF-α consequence of divergent roles for the TNF receptors,
neutralizing antibody and illustrates the deleterious effect considering that blocking TNF-RI in mouse models damp-
of TNF-α on myelin and oligodendrocyte integrity (Probert ens disease severity, while suppressing TNF-RII, the
et al. 1995). Furthermore, in experimental autoimmune receptor that induces remyelination and harbors immuno-
encephalomyelitis (EAE), a mouse model for brain inflam- suppressive properties, results in exacerbated disease
mation and MS, anti-TNF or sTNF receptor antibodies (Arnett et al. 2001; Kassiotis and Kollias 2001).
ameliorate disease (Selmaj et al. 1991; Baker et al. 1994). Recently, pharmacological agents selectively targeting
Studies have demonstrated that TNF-α is necessary for the TNF-RI have been investigated. Using phage display
early initiation of EAE, where mice deficient in TNF-α technology, a TNF-RI antagonist was developed, and
display delayed onset of disease compared to wild-type upon evaluation in EAE mice it was found that adminis-
counterparts. However in later phases of disease, TNF- tration of this selective antagonist improved clinical
deficient mice still develop severe EAE with extensive scores, lessened cerebral demyelination, and suppressed
inflammation and primary demyelination, indicating that the number of infiltrating inflammatory cells, as well as
TNF-α may be more important for initial leukocyte homing Th1 and Th17 immune responses (Nomura et al. 2011).
rather than progression of later stage disease (Korner et al. Although these experiments were performed in mice, and
1997). The immunosuppressive roles of TNF-α may be one have yet to be tested in humans, investigators are actively
possible explanation for disease progression in TNF- pursuing these more selective strategies to modulate TNF-α
deficient mice. TNF-α is partially involved in deactivating signaling in MS.
myelin basic protein or (MBP)-reactive autoimmune T cells
and TNF-deficient mice harbor a prolonged T cell response Parkinson’s disease
against myelin, but these reactive T cells accumulate and
subsequently contribute to chronic EAE (Kassiotis and Parkinson’s disease (PD) is the second most common age-
Kollias 2001). Failure to reverse these reactive T cell related neurodegenerative disorder causing progressive and
responses appears to be TNF-RII-mediated, since TNF-RI chronic mobility debilitation. Afflicted individuals develop
KO mice harbor reduced inflammatory cell infiltrates and resting tremor, bradykinesia, muscle rigidity, and postural
demyelination, while severe disease persists in TNF-RII irregularity. Hallmark pathologies include gradual loss of
KO mice (Suvannavejh et al. 2000). These data suggest that dopaminergic neurons in the substantia nigra (SN) pars
not only can TNF-α play a protective role in EAE by compacta and intraneuronal proteinaceous inclusions called
suppressing potential MBP-reactive T cells, but at elevated Lewy bodies, which contain aggregates of fibrillar α-
levels can accelerate disease progression and onset of white synuclein and ubiquitinated proteins (Spillantini et al.
matter lesions. 1997; Chung et al. 2001). Neuronal loss and degeneration
Therapeutic anti-TNF-α inhibition for MS patients has is most severe in the SN; however, non-motor, PD-
had limited success, despite a wealth of preclinical research associated deficits in cognition and autonomic nervous
findings indicating the beneficial effects of TNF-α sup- system function may be attributed to cholinergic, adrener-
pression. During an open-label phase I trial, a monoclonal gic, and serotoninergic cell death (Chaudhuri et al. 2006).
J Neuroimmune Pharmacol (2012) 7:42–59 49

Currently, there are no disease-modifying therapies for PD, trations contribute to DA neuron demise in PD. In a 6-
although symptomatic treatment is effective during earlier hydroxydopamine (6-OHDA)-based animal model that
stages of disease. Over time, with increasing disease exhibits slowly progressing degeneration that is more
severity, these drugs are no longer effective (Schapira reminiscent of human PD, TNF-α protein levels are
2009). While the etiology of PD remains to be fully significantly enhanced within the striatum and SN (Mogi
elucidated, a majority of idiopathic PD cases appear to arise et al. 1999). When low levels of TNF-α (17–19 pg/mg) are
from a complex interaction between genetic susceptibility expressed in the SN for a period of 2 weeks, DA
and environmental factors (Gorell et al. 2004). Increased neurodegeneration progressively ensues and leads to motor
risk for PD has been shown with rural residency, pesticide impairments, infiltration of leukocytes, astrogliosis, and
exposure, drinking well water and occupations including microglial activation via COX-2 and/or NOS activity,
mining, welding, and farming (Priyadarshi et al. 2001; suggesting that TNF-α is both necessary and sufficient to
Firestone et al. 2005; Jankovic 2005). Beside environmen- exacerbate DA neuron demise (De Lella Ezcurra et al.
tal stimuli, 15 distinct chromosomal loci have been linked 2010). In another neurotoxicant PD model, intrastriatal
to increased susceptibility to PD, and specific genes with administration of a soluble TNF-α dominant-negative
some of these loci have been identified and implicated in inhibitor to neutralize endogenous TNF-α resulted in a
pathogenesis (Lesage and Brice 2009). rescue of approximately 50% of the DA neurons that
Neuroinflammatory processes appear to play key roles in typically succumb to oxidative neurotoxin 6-OHDA or
DA neuron dysfunction and death in PD. Activated micro- endotoxin-induced death (McCoy et al. 2006).
glia and T-lymphocytes are present in the SN of postmortem In contrast, several investigators have shown beneficial
PD brain and positron emission tomography (PET) studies effects for TNF-α in the context of PD. For example, mice
using [11 C](R)-PK11195, a marker for activated microglia, deficient for both TNF-RI and RII exposed to 1-methyl-4-
show that more activated microglia selectively cluster in phenyl-1,2,3,4-tetrahydropyridine (MPTP), a DA neuronal
regions of the brain damaged by PD, including the basal toxin, exhibit DA neuron loss and perform worse on the
ganglia, striatum, the pons, and frontal and temporal cortex rotarod motor test, as compared to mice lacking only one of
(McGeer et al. 1988; Banati et al. 1998; Imamura et al. the TNF receptors or wild-type littermates (Rousselet et al.
2003; Gerhard et al. 2006). In fact, large numbers of TNF-α 2002). Similarly, wild-type, TNF-RI KO, and TNF-RII KO
immunoreactive glial cells are found in the SN of PD mice treated with MPTP for 8 days displayed no differences
patients compared to healthy controls and most DA neurons in striatal dopamine or dopamine transporter levels, arguing
are TNF receptor-positive (Boka et al. 1994). TNF-α is against a role for TNF-α neurotoxicity (Leng et al. 2005).
elevated in the striatum and cerebral spinal fluid (CSF) of In another study, mice lacking both high-affinity TNF
PD patients, while sTNF-RI levels are also increased in the receptors exhibit decreased microglial activation within the
CSF (Mogi et al. 1994; Mogi et al. 1995; Dobbs et al. 1999; striatum following exposure to MPTP (Sriram et al. 2002).
Mogi et al. 2000). Single-nucleotide polymorphisms local- Surprisingly, however, increased neuronal damage and
ized to the TNF-α promoter region that positively correlate microtubule-associated protein-2 reactivity were found in
with increased risk for developing sporadic PD have been their hippocampi (Sriram et al. 2002, 2006), demonstrating
reported (Nishimura et al. 2001). Moreover, variance in a clear region-specific difference in TNF-α signaling within
TNF-α promoter methylation patterns may favor selective the same brain, where TNF-α is protective in the
vulnerability to certain populations of neurons, particularly hippocampus but drives neurodegeneration in the SN
to SN-resident neurons. In both healthy and PD patients following MPTP neurotoxicant exposure. Chertoff and
post-translational methylation is reduced in the SN com- colleagues have also explored the concept of TNF-α
pared to the cortex. However, PD patients harbor a playing dual roles in PD pathophysiology. Young 2–
significant increase in unmethylated cytosine CpG dinucle- 3 month-old mice subjected to 6-OHDA with constitutive,
otide residues within the TNF-α promoter of SN neurons low-level expression of TNF-α are spared in respect to
that may modulate AP-2 and Sp1 transcription factor nigrostriatial degeneration, whereas overt neuronal loss and
binding, since promoter transcription factor binding is a inflammatory responses occurs in mice overexpressing
methylation-dependent process (Pieper et al. 2008). These TNF-α, indicating that the level and timing of TNF-α
data may have implications in the role of TNF-α gene plays a pivotal role in the progression of disease (Chertoff
transcriptional regulation and susceptibility of SN-resident et al. 2011). Collectively, these data demonstrate that the
neurons to PD-related degeneration. development of anti-TNF-α PD therapies requires a highly
TNF-α levels have been shown to correlate with the tuned and selective strategy based upon assiduous exami-
extent of DA neuronal death in PD-afflicted brain regions, nation of the dependence of disease stage, TNF-α levels,
suggesting that either the death of DA neurons induces cellular context, and individual receptor involvement in
local cytokine production or that increased TNF-α concen- multiple models of PD. It is clear with the latter that the
50 J Neuroimmune Pharmacol (2012) 7:42–59

generation of predictive preclinical models of PD is of the levels correlate with disease severity (Fillit et al. 1991;
utmost importance if such complex signaling effects of Dickson 1997; Paganelli et al. 2002). More recently, MCI
TNF-α during idiopathic PD are to ever be accurately patients who subsequently developed AD were found to
dissected. harbor increased levels of sTNF-RI and sTNF-RII that
correlate with β-site APP-cleaving enzyme 1 (BACE1)
Alzheimer’s disease cleavage activity, which is a key enzyme involved in
pathological Aβ metabolism (Buchhave et al. 2010). In
Alzheimer’s disease (AD) is the most prevalent age-related addition, genome-wide screens have attributed microsatel-
disorder, which is associated with progressive development lite TNF-α and gene promoter polymorphisms to increased
of severe memory loss, cognitive deficits, personality and production of TNF-α and increased risk for developing AD
behavior changes influencing mood, communication, judg- (Culpan et al. 2003; Ma et al. 2004; Collins et al. 2000;
ment, and reasoning. According to the Alzheimer’s Asso- Laws et al. 2005).
ciation, this neurodegenerative malady affects 5.4 million Transgenic mouse models have been instrumental in
Americans, and alarmingly, AD prevalence is ever increas- deciphering the role of TNF-α in AD, but like in the PD
ing. By 2050, it is projected that 11–16 million people in research field, the accuracy of presently available models in
the U.S. alone will be afflicted with AD (http://www.alz. faithfully mimicking the human disease and their utility in
org/). Albeit the vast majority of AD cases are sporadic assessing potential efficacy of new therapeutic interventions
with symptoms beginning after the age of 65, cases of is up for debate. Given these caveats, TNF-α is upregu-
early-onset familial AD have been documented, which are lated, co-localized with amyloid plaques, and neurotoxic in
caused by inherited autosomal-dominant mutations in the mouse models that recapitulate specific human AD-related
amyloid precursor protein (APP), presenilin-1 (PS1) and pathologies. For instance, the triple transgenic mouse model
presenilin-2 (PS2) genes. While the exact cause of sporadic of AD (3xTg-AD) that harbors three familial-linked AD
AD is not fully understood, age is the strongest predictor of mutations (APPswe, PS1M146V , and TauP301L) and progres-
disease. Nevertheless, due to the complexity and multifac- sively develops amyloid and tau pathologies exhibits
torial nature of AD, the development of effective disease- increased TNF-α and monocyte chemoattractant protein-1
modifying therapies has been especially challenging. (MCP-1) transcript levels, as well as increased numbers of
Classic neuropathological correlates of AD include activated microglia, in the entorhinal cortex at an age
extracellular amyloid plaques primarily composed of preceding appearance of plaque and tangle pathologies
accumulated Aβ peptides and intraneuronal filamentous (Janelsins et al. 2005). The amyloidogenic Tg2576 mouse
inclusions called neurofibrillary tangles caused by hyper- model develops Aβ plaques, increased TNF-α immuno-
phosphorylation of the tau cytoskeletal protein (Merz et al. reactivity, and glial cell clustering adjacent to fibrillar
1983; Braak and Braak 1988). In addition, brain region- plaques (Mehlhorn et al. 2000). Similarly, in an APPswe/
specific synapse loss, inflammation and neuronal death are PS1dE9 mouse model, TNF-α expression increases in a
other major pathophysiological markers of AD. The disease stage-specific manner, and TNF-α immunoreactive
amyloid cascade hypothesis is a predominate theory in the cells co-localize with ThioflavineS-positive Aβ plaques
field stating that overproduction of Aβ, or a failure to (Ruan et al. 2009).
efficiently clear the peptide is the earliest initiating event in Significant experimental evidence exists that identify
AD. Aβ is fibrillogenic in nature and can further aggregate relationships between Aβ and TNF-α signaling. Both
into oligomeric structures, which are highly neurotoxic and cognate TNF-α receptors have been shown to physically
synaptotoxic, and eventually these Aβ aggegates form bind soluble Aβ40 (Li et al. 2004), and overexpression of
higher order structures and deposit as plaques within the TNF-RI in neurons exposed to Aβ40 in vitro induces
brain parenchyma (Hardy and Higgins 1992; Pimplikar neuronal death (Li et al. 2004). TNF-α exposure triggers
2009). Amyloidogenic plaques are characterized by the APP cleavage to generate disease-related Aβ peptides by
presence of intimately associated activated microglia and regulating processing enzyme activity (Kuo et al. 2008;
reactive astrocytes. These cells, as stated above, are major Chen et al. 2011; Yamamoto et al. 2007). Elaboration of
sources of pro-inflammatory molecules such as chemo- Aβ42, the more hydrophobic plaque-associated form of Aβ,
kines, complement factors, cytokines, and reactive oxygen is sequentially cleaved from APP by orchestrated β-
species (Ruan et al. 2009). Therefore, it is not surprising (BACE1) and γ-secretase processing enzyme activities.
that the master regulating cytokine, TNF-α, has been TNF-RI-mediated TNF-α signaling upregulates BACE1
intensely studied and debated regarding its purported role activity via NFκB activation and APP mice lacking TNF-RI
in AD pathogenesis. Examination of post-mortem AD harbor reduced Aβ peptide concentrations and amyloid
brains reveals that increased TNF-α co-localizes with Aβ plaque load (He et al. 2007). Competition for APP occurs
plaques, is upregulated in both CSF and serum, and its between BACE1 and TACE, the TNF-α converting
J Neuroimmune Pharmacol (2012) 7:42–59 51

enzyme (also referred to as α-secretase), which cleaves degeneration, and reactive gliosis (Giuliani et al. 2009). In
both precursor TNF-α and the ectodomain of APP leading the absence of TNF-α, spatial memory as assessed by the
to decreased Aβ generation (Skovronsky et al. 2000). Since Morris Water Maze revealed no improvements in spatial
TNF-α is highly expressed in AD, TACE may be in high learning and memory at 6 and 15 months of age compared
demand for TNF-α maturation, and as a result, BACE1 to control mice, and amyloid burden was greatly enhanced
may be more available to initiate amyloidogenic cleavage at 20 months of age (Giuliani et al. 2009). Furthermore,
of APP, thereby releasing more Aβ peptide. APP metabo- long-term abrogation of TNF-α receptor expression leads to
lism by γ-secretase is also modulated by TNF-α and elevated extracellular amyloid plaque deposition and
depends upon JNK mitogen-activated protein (MAP) kinase accelerated paired helical filament formation of 15 month-
signaling (Liao et al. 2004). old 3xTg-AD mice. The enhancement of amyloid pathol-
Experimental manipulation of TNF-α levels in vivo has ogy is, in part, due to impaired phagocytosis activity by
provided valuable insight into the role of TNF-α during AD microglia derived from 3xTg-AD mice lacking TNF-RI and
pathogenesis and therapeutic potential of anti-TNF TNF-RII, suggesting that long-term pan TNF-α inhibition
approaches to prevent and/or treat AD. Employing a worsens pathology and suppresses glial activation that may
selective soluble TNF-α dominant-negative inhibitor, which be required for Aβ clearance (Montgomery et al., under
has a predilection for TNF-RI engagement, transiently in review). In further support for a possible protective role of
conjunction with LPS-induced inflammation, reduces intra- TNF-α in AD, mouse TNF-α was stereotaxically injected
neuronal Aβ accumulation and APP C-terminal fragment into the hippocampus of TgCNRD8 mice using recombi-
generation in the hippocampus, cortex, and amygdala of nant adeno-associated virus 1/2 and amyloid plaque
3xTg-AD mice (McAlpine et al. 2009). APP/PS1 transgen- pathology was dramatically decreased and a strong micro-
ic mice intracerebroventricularly injected with the mono- glia cell activation was induced (Chakrabarty et al. 2011).
clonal anti-TNF antibody, infliximab, for 3–14 days Although this study overexpressed TNF-α transiently, it
exhibited reduced amyloid plaque deposition and tau ascertains potential beneficial roles for TNF-α in activating
hyperphosphorylation. Interestingly, CD11c-positive microglia to mediate Aβ clearance and may address the
dendritic-like cells clustered in close proximity with importance for cell-specific restriction and/or facilitation of
amyloid plaques specifically in mice receiving infliximab TNF-signaling during particular stages of AD.
(Shi et al. 2011), suggesting that these cells participated in Activated microglia are capable of Aβ peptide clearance
clearing Aβ peptide in treated mice (Butovsky et al. 2007). by internalizing both soluble Aβ and fibrillar Aβ con-
Furthermore, by pharmacologically blocking TNF-α or formations. Specifically, microglia-mediated internalization
through use of TNF-RI knockout mice intrahippocampally of soluble Aβ transpires through fluidic macropinocytosis
injected with Aβ1-40, learning and memory deficits typical- that is distinct from phagocytosis and receptor mediated
ly evoked by the synergistic effects of TNF-α and Aβ were endocytosis. In addition, soluble Aβ uptake, of which
rescued (Medeiros et al. 2007). While these studies have localizes diffusely throughout the entire cell, seems to be
been very informative in understanding the effects of TNF- independent of clathrin-coated vesicle assembly and mem-
α on AD progression and support the use of anti-TNF-α brane cholesterol, whereas internalization of fibrillary Aβ is
therapy for human treatment, a few critical unknowns trafficked to primarily large cytoplasmic phagocytic
remain. The abovementioned studies were performed over a vesicles (Mandrekar et al. 2009). Phagocytosis of fibrillary
relatively short period of time in rodent models, whereas Aβ depends on cell surface marker expression, including
human patients would require longer-term administration of toll-like receptors, CD36, CD14, α6β1, CD47, and SIRP-
anti-TNF-α inhibitors. Hence, what is the effect of long- β1, and a subset of these proteins are regulated by TNF-α
term suppression of TNF-α signaling and are there (Koenigsknecht and Landreth 2004; Liu et al. 2005a;
distinct disease stage-specific positive roles that TNF-α Gaikwad et al. 2009; Reed-Geaghan et al. 2009). For
plays that would be inhibited by anti-TNF-α therapeu- example, CD14 expression positively correlates with TNF-
tics? In addition, the above studies do not take into α levels, whereas CD36 negatively correlates with TNF-α
account cell type-specific mechanisms of TNF-α within (Liu et al. 2005a; Hickman et al. 2008). Moreover, TNF-α
the CNS, where TNF-α signaling may need to be is a strong inducer of microglial activation and appears to
maintained in one or several cell types for “normal” be an important factor in regulating Aβ internalization.
physiologic processes. Pan-inhibition of TNF-α activity Thus, intact microglial TNF-α signaling may facilitate
in the setting of AD could undermine such processes and reduced Aβ plaque deposition via enhancement of Aβ
lead to increased dysfunction. peptide clearance (Fig. 4). Further studies are needed to
Recently, Giuliani and colleagues demonstrated the fully understand and elucidate when microglial TNF-α
effect of chronic TNF-α ablation in the PDAPP mouse signaling is required, since during later stages of disease,
model, which displays amyloid plaque pathology, neuro- microglial phagocytosis or internalization of Aβ peptide
52 J Neuroimmune Pharmacol (2012) 7:42–59

No TNF-α Modulation Pan TNF-α Modulation Cell-selective TNF-α Modulation

TNF-α Production TNF-α Production TNF-α Production

Microglial Neuronal Microglial Neuronal Microglial Neuronal


Signaling Signaling Signaling Signaling Signaling Signaling

Aβ Clearance Aβ Production Aβ Clearance Aβ Production Aβ Clearance Aβ Production

Transient
Suppression
Disease Disease Disease
Progression or Progression ? Progression
Chronic
Suppression

Fig. 4 Potential anti-TNF-α therapeutic strategies for AD. Intact mediated Aβ clearance. Pan TNF-α inhibition may quell TNF-α
microglial TNF-α signaling potentiates Aβ uptake and subsequent induced Aβ production by neurons, but also will suppress microglial
clearance of the peptide. Neuronal TNF-α signaling may lead to Aβ clearance. Selectively targeting neuronal TNF signaling and
increased Aβ generation and eventually result in the death of this cell allowing microglial signaling at certain disease stages may reduce
type. Without any therapeutic TNF-α modulation, disease progression neuronal Aβ genesis but permit microglia-driven Aβ clearance
ensues with heightened Aβ production and limited microglial-

pools become compromised resulting in the profound toxicity via NFκB-dependent signaling (Barger et al. 1995).
production of highly toxic pro-inflammatory molecules that Furthermore, TNF-α is protective to neurons against
contributes significantly to the chronic inflammatory milieu oxidative stress by stimulating antioxidant signaling and
and coincident neurotoxicity. superoxide accumulation through induced MnSOD activity
While microglial TNF-α signaling at certain stages of (Bruce et al. 1996; Bruce-Keller et al. 1999). Taken
AD may repress AD-associated pathology, other brain- together, more investigation is warranted to better discern
resident cells likely respond differently. Neurons have been the cell-specific roles of TNF-α in the context of AD and
well studied in the AD field because these cells are the the cell-cell interactions amongst CNS-resident cell pop-
primary sources of Aβ peptides and are the sites of ulations in order to develop therapeutics that may be able to
dysfunction. Existing data suggest TNF-α signaling in selectively modulate TNF-α signaling in a cell type-
neurons is neurotoxic, and considering the direct role TNF- specific manner.
α plays in APP generation, it is plausible that inhibiting
neuronal TNF-α signaling would be protective. During AD,
neurons produce appreciable levels of TNF-α, and when TNF-α ablation as a therapeutic strategy
TNF-α is chronically overexpressed specifically by neurons
in the 3xTg-AD model, amyloid and phospho-tau pathol- Historical and epidemiological perspective for TNF-α
ogies are exacerbated compared to non-transgenic control immune modulation in brain diseases
mice, eventually leading to neuronal cell death (Janelsins et
al. 2008). Similarly, primary neurons overexpressing TNF- A naturally occurring TNF-α inhibitor was discovered in
RI and treated with Aβ leads to neuronal cell death, while human urine by Seckinger and colleagues in 1988, which
neurons from TNF-RI knockout mice are spared in the was identified as a soluble form of the TNF-receptor that
presence of Aβ (Li et al. 2004). These latter data suggest acted by neutralizing the cytokine (Seckinger et al. 1990).
that selectively preventing neuronal TNF-α signaling, Subsequently, two TNF-binding proteins were purified that
through targeted blocking of receptor expression, may were capable of inhibiting the binding of TNF-α to cells
preserve neurons during the course of AD (Fig. 4). (Engelmann et al. 1990). The identification of soluble TNF-
Despite the ample data supporting detrimental effects for receptors paved the way for the development of soluble
intact neuronal TNF-α signaling, several reports have TNF-receptor antibodies currently used for the treatment of
alluded to neuroprotective roles for TNF-α. In the presence several systemic inflammatory diseases, including rheuma-
of Aβ peptide, dissociated neuronal cultures pretreated with toid arthritis (RA), juvenile polyarticular RA, inflammatory
TNF-α were saved from Aβ-induced neuronal death by bowel disease, psoriatic arthritis and ankylosing spondylitis
specifically protecting against iron and intracellular Ca2+ (Sfikakis 2010). There are three anti-TNF-α agents
J Neuroimmune Pharmacol (2012) 7:42–59 53

approved for clinical use: Etanercept (Enbrel®), infliximab mediated by pro-inflammatory cytokines, including IL-1β,
(Remicade®), and adalimumab (Humira®). The latter two IL-6, and pertinent to this discussion, TNF-α (Heneka and
are full-length bivalent IgG monoclonal antibodies specific O’Banion 2007). Cumulatively, these studies demonstrate
for sTNF and tmTNF, whereas Etanercept is a genetically that inflammation is a critical mechanism in neurodegener-
engineered Fc fusion protein generated from the extracel- ative disease progression and for a therapeutic benefit to be
lular domain of human TNF-RII and functions as a decoy realized, careful dissection of these processes in the setting
receptor to block sTNF, tmTNF, and distinct ligands of of disease-relevant models is crucial.
lymphotoxin, a TNF-related protein (Tracey et al. 2008). A
detailed review of these TNF-α antagonists and their Clinical trials for selective TNF-α inhibition
mechanisms of action have been published elsewhere
(Tracey et al. 2008). Clinical trials examining the effects of TNF-α inhibition
Epidemiological studies support the use of immunomo- have been conducted on patients with MS and AD. As
dulation strategies to suppress inflammatory processes in briefly discussed, strategies to inhibit TNF-α in MS seemed
neurodegenerative disease. Long-term use of non-steroidal promising in preclinical applications, but disappointingly,
anti-inflammatory drugs (NSAIDs) that inhibit cyclooxy- have widely failed in human clinical trials due to the lack of
genase, an enzyme involved in prostaglandin synthesis, has therapeutic selectivity. These studies employed either
been shown in retrospective studies to be protective for infliximab or lenercept and broadly neutralized both
chronic brain diseases such as in PD and AD. In 1997, soluble and transmembrane TNF-α in all cell types
Stewart et al. reported that prolonged use of NSAIDs for (1999; van Oosten et al. 1996). Infliximab was adminis-
2 years or more reduced AD onset in a longitudinal study of tered to two patients with rapidly progressing disease
1,686 subjects (Stewart et al. 1997). These results have during an open-label phase I clinical trial, while in a
been confirmed by several other studies using population- double-blinded and multicentered phase II study 168
based cohort studies involving 6,989 participants, com- relapse-remitting patients were given lenercept. In both
bined data from nine different studies of 14,654 people, and studies, disease status was intensified. TNF-α interven-
a large case–control study of 246,199 subjects (in t’ Veld et tion for AD has been evaluated in open-labeled phase I
al. 2001; Etminan et al. 2003; Vlad et al. 2008). In a pilot clinical trials where perispinal extrathecal administration
study, while administration of a non-selective NSAID, of Etanercept was administered weekly to a small
indomethacin, for 6 months appeared to slow cognitive number of patients ranging from mild to severe AD for
decline in mild to moderate impaired AD patients, a second a short duration of 6 month that claimed substantial
pilot study using diclofenac and misoprostal, a drug used to cognitive and behavioral improvements, including verbal
protect the gastrointestine, reported no differences between fluency and aphasia (Tobinick et al. 2006; Tobinick and
placebo and treated patients (Scharf et al. 1999). Large Gross 2008). Currently, a phase II study is recruiting to
multi-site trials were subsequently performed where people evaluate the safety and tolerability of Etanercept in AD
with established AD were administered nimesulide, cele- (www.clinicaltrials.gov). These results seem promising,
coxib, naproxen, or prednisone, which preferentially inhibit but conclusions regarding the promise of such a therapeu-
cyclooxygenase 2 activity. No significant effects on tic strategy should be reserved until after extensive
cognitive and behavioral function were observed (Aisen et chronic suppression of TNF-α activity is performed in
al. 2000; Aisen et al. 2002; Aisen et al. 2003). Possible preclinical models and double-blind human clinical trials
explanations for the failure of COX-2 inhibition could be have been conducted and results critically reviewed by the
attributed to advanced disease stage of the patients, duration research community.
of treatment, doses of drugs administered or targeting of the
wrong inflammatory pathway, since NSAIDs are broadly
acting, interact with many molecular targets, and are more Conclusion and future considerations
effective at inhibiting microglia-related inflammatory pro-
cesses (Mackenzie and Munoz 1998). In fact, Klegeris et al. There is no doubt inflammatory processes, especially those
reported that NSAIDs reduced pro-inflammatory cytokine mediated by the central pro-inflammatory cytokine, TNF-α,
production from microglia, increased microglial phagocy- play key roles in degenerative conditions afflicting the
tosis activity, and prevented microglial-induced neuronal CNS. The precise role(s) TNF-α plays, in particular, remain
cell killing (Klegeris and McGeer 2005). Besides targeting highly controversial due to the complexity and pleiotropic
cyclooxygenase activity, NSAIDs can activate the peroxi- nature of this cytokine and the activities attributed to TNF-
some proliferator-activated receptor gamma or PPARγ α during critical developmental and homeostatic cellular
(Jaradat et al. 2001). PPARγ is expressed by macrophages processes. Multiple factors determine whether TNF-α will
and receptor engagement inhibits inflammatory processes exert deleterious or beneficial effects for neuronal survival,
54 J Neuroimmune Pharmacol (2012) 7:42–59

and some of these differential actions relate to its duration involvement of a kappa B-binding factor and attenuation of
peroxide and Ca2+ accumulation. Proc Natl Acad Sci USA
of expression (chronic vs. transient), concentration, receptor 92:9328–9332
conformation, and the inflammatory milieu. However, Barker V, Middleton G, Davey F, Davies AM (2001) TNFalpha
despite the elaborate and promising data collected thus far contributes to the death of NGF-dependent neurons during
to assign function to TNF-α in neurodegeneration, surpris- development. Nat Neurosci 4:1194–1198
Barton A, John S, Ollier WE, Silman A, Worthington J (2001)
ingly little is still known about the cellular and stage- Association between rheumatoid arthritis and polymorphism of
specific roles of this cytokine. Given the limited under- tumor necrosis factor receptor II, but not tumor necrosis factor
standing of selective TNF-α signaling, caution is urged receptor I, in Caucasians. Arthritis Rheum 44:61–65
when clinically inhibiting the activity of TNF-α, and it is Bear MF, Malenka RC (1994) Synaptic plasticity: LTP and LTD. Curr
Opin Neurobiol 4:389–399
likely that prolonged global suppression of TNF-receptor Beattie EC, Stellwagen D, Morishita W, Bresnahan JC, Ha BK, Von
signaling may not be an effective approach to remedy Zastrow M, Beattie MS, Malenka RC (2002) Control of synaptic
neurodegeneration and may even intensify the disease state. strength by glial TNFalpha. Science 295:2282–2285
Therefore, further in vivo dissection of TNF-α signaling Beck J, Rondot P, Catinot L, Falcoff E, Kirchner H, Wietzerbin J
(1988) Increased production of interferon gamma and tumor
and its associated receptors in a cell and stage specific necrosis factor precedes clinical manifestation in multiple
context is justified for the successful development of sclerosis: do cytokines trigger off exacerbations? Acta Neurol
selective therapeutic interventions that are safe and effica- Scand 78:318–323
ciously ameliorate the deleterious actions of TNF-α in Bernardino L, Agasse F, Silva B, Ferreira R, Grade S, Malva JO
(2008) Tumor necrosis factor-alpha modulates survival, prolifer-
neurodegenerative disease. ation, and neuronal differentiation in neonatal subventricular
zone cell cultures. Stem Cells 26:2361–2371
Boka G, Anglade P, Wallach D, Javoy-Agid F, Agid Y, Hirsch EC
References (1994) Immunocytochemical analysis of tumor necrosis factor
and its receptors in Parkinson’s disease. Neurosci Lett
172:151–154
(1999) TNF neutralization in MS: results of a randomized, placebo- Boursalian TE, Fink PJ (2003) Mutation in fas ligand impairs
controlled multicenter study. The Lenercept Multiple Sclerosis maturation of thymocytes bearing moderate affinity T cell
Study Group and The University of British Columbia MS/MRI receptors. J Exp Med 198:349–360
Analysis Group. Neurology 53:457–465 Braak H, Braak E (1988) Neuropil threads occur in dendrites of
Aggarwal BB, Samanta A, Feldmann M. (2000) TNF receptors. In: tangle-bearing nerve cells. Neuropathol Appl Neurobiol
Oppenheim JJ, Editor, Cytokine reference, Academic Press, 14:39–44
London, pp. 1620–1632 Bruce AJ, Boling W, Kindy MS, Peschon J, Kraemer PJ, Carpenter
Aisen PS, Davis KL, Berg JD, Schafer K, Campbell K, Thomas RG, MK, Holtsberg FW, Mattson MP (1996) Altered neuronal and
Weiner MF, Farlow MR, Sano M, Grundman M, Thal LJ (2000) microglial responses to excitotoxic and ischemic brain injury in
A randomized controlled trial of prednisone in Alzheimer’s mice lacking TNF receptors. Nat Med 2:788–794
disease. Alzheimer’s disease cooperative study. Neurology Bruce-Keller AJ, Geddes JW, Knapp PE, McFall RW, Keller JN,
54:588–593 Holtsberg FW, Parthasarathy S, Steiner SM, Mattson MP (1999)
Aisen PS, Schmeidler J, Pasinetti GM (2002) Randomized pilot study Anti-death properties of TNF against metabolic poisoning:
of nimesulide treatment in Alzheimer’s disease. Neurology mitochondrial stabilization by MnSOD. J Neuroimmunol
58:1050–1054 93:53–71
Aisen PS, Schafer KA, Grundman M, Pfeiffer E, Sano M, Davis KL, Buchhave P, Zetterberg H, Blennow K, Minthon L, Janciauskiene
Farlow MR, Jin S, Thomas RG, Thal LJ (2003) Effects of S, Hansson O (2010) Soluble TNF receptors are associated
rofecoxib or naproxen vs placebo on Alzheimer disease progres- with Abeta metabolism and conversion to dementia in subjects
sion: a randomized controlled trial. JAMA 289:2819–2826 with mild cognitive impairment. Neurobiol Aging 31:1877–
Arnett HA, Mason J, Marino M, Suzuki K, Matsushima GK, Ting JP 1884
(2001) TNF alpha promotes proliferation of oligodendrocyte Butovsky O, Kunis G, Koronyo-Hamaoui M, Schwartz M (2007)
progenitors and remyelination. Nat Neurosci 4:1116–1122 Selective ablation of bone marrow-derived dendritic cells
Baker D, Butler D, Scallon BJ, O’Neill JK, Turk JL, Feldmann M increases amyloid plaques in a mouse Alzheimer’s disease
(1994) Control of established experimental allergic encephalo- model. Eur J Neurosci 26:413–416
myelitis by inhibition of tumor necrosis factor (TNF) activity Cacci E, Claasen JH, Kokaia Z (2005) Microglia-derived tumor
within the central nervous system using monoclonal antibodies necrosis factor-alpha exaggerates death of newborn hippocampal
and TNF receptor-immunoglobulin fusion proteins. Eur J progenitor cells in vitro. J Neurosci Res 80:789–797
Immunol 24:2040–2048 Carswell EA, Old LJ, Kassel RL, Green S, Fiore N, Williamson B
Banati RB, Daniel SE, Blunt SB (1998) Glial pathology but absence (1975) An endotoxin-induced serum factor that causes necrosis
of apoptotic nigral neurons in long-standing Parkinson’s disease. of tumors. Proc Natl Acad Sci USA 72:3666–3670
Mov Disord 13:221–227 Chakrabarty P, Herring A, Ceballos-Diaz C, Das P, Golde TE (2011)
Banner DW, D’Arcy A, Janes W, Gentz R, Schoenfeld HJ, Broger C, Hippocampal expression of murine TNFalpha results in attenu-
Loetscher H, Lesslauer W (1993) Crystal structure of the soluble ation of amyloid deposition in vivo. Mol Neurodegener 6:16
human 55 kd TNF receptor-human TNF beta complex: implica- Chaudhuri KR, Healy DG, Schapira AH (2006) Non-motor symptoms
tions for TNF receptor activation. Cell 73:431–445 of Parkinson’s disease: diagnosis and management. Lancet
Barger SW, Horster D, Furukawa K, Goodman Y, Krieglstein J, Neurol 5:235–245
Mattson MP (1995) Tumor necrosis factors alpha and beta protect Chen CH, Zhou W, Liu S, Deng Y, Cai F, Tone M, Tone Y, Tong Y, Song W
neurons against amyloid beta-peptide toxicity: evidence for (2011) Increased NF-kappaB signalling up-regulates BACE1
J Neuroimmune Pharmacol (2012) 7:42–59 55

expression and its therapeutic potential in Alzheimer’s disease. Int J enhance the plasticity of the immune system. Cytokine Growth
Neuropsychopharmacol doi:10.1017/S1461145711000149 Factor Rev 15:353–366
Chertoff M, Di Paolo N, Schoeneberg A, Depino A, Ferrari C, Wurst Ekdahl CT, Claasen JH, Bonde S, Kokaia Z, Lindvall O (2003)
W, Pfizenmaier K, Eisel U, Pitossi F (2011) Neuroprotective and Inflammation is detrimental for neurogenesis in adult brain. Proc
neurodegenerative effects of the chronic expression of tumor Natl Acad Sci USA 100:13632–13637
necrosis factor alpha in the nigrostriatal dopaminergic circuit of Engelmann H, Novick D, Wallach D (1990) Two tumor necrosis
adult mice. Exp Neurol 227:237–251 factor-binding proteins purified from human urine. Evidence for
Chung KK, Zhang Y, Lim KL, Tanaka Y, Huang H, Gao J, Ross immunological cross-reactivity with cell surface tumor necrosis
CA, Dawson VL, Dawson TM (2001) Parkin ubiquitinates the factor receptors. J Biol Chem 265:1531–1536
alpha-synuclein-interacting protein, synphilin-1: implications Etminan M, Gill S, Samii A (2003) Effect of non-steroidal anti-
for Lewy-body formation in Parkinson disease. Nat Med inflammatory drugs on risk of Alzheimer’s disease: systematic
7:1144–1150 review and meta-analysis of observational studies. BMJ 327:128
Collins JS, Perry RT, Watson B Jr, Harrell LE, Acton RT, Blacker Fillit H, Ding WH, Buee L, Kalman J, Altstiel L, Lawlor B, Wolf-
D, Albert MS, Tanzi RE, Bassett SS, McInnis MG, Campbell Klein G (1991) Elevated circulating tumor necrosis factor levels
RD, Go RC (2000) Association of a haplotype for tumor in Alzheimer’s disease. Neurosci Lett 129:318–320
necrosis factor in siblings with late-onset Alzheimer disease: Firestone JA, Smith-Weller T, Franklin G, Swanson P, Longstreth WT
the NIMH Alzheimer Disease Genetics Initiative. Am J Med Jr, Checkoway H (2005) Pesticides and risk of Parkinson disease:
Genet 96:823–830 a population-based case-control study. Arch Neurol 62:91–95
Constantinescu S-YLaCS (2010) TNF-alpha: A Paradigm of Paradox Fontaine V, Mohand-Said S, Hanoteau N, Fuchs C, Pfizenmaier K,
and Complexity in Multiple Sclerosis and its Animal Models. Eisel U (2002) Neurodegenerative and neuroprotective effects of
The Open Autoimmunity Journal 2:160–170 tumor Necrosis factor (TNF) in retinal ischemia: opposite roles of
Culpan D, MacGowan SH, Ford JM, Nicoll JA, Griffin WS, Dewar D, TNF receptor 1 and TNF receptor 2. J Neurosci 22:RC216
Cairns NJ, Hughes A, Kehoe PG, Wilcock GK (2003) Tumour Friedmann E, Hauben E, Maylandt K, Schleeger S, Vreugde S,
necrosis factor-alpha gene polymorphisms and Alzheimer’s Lichtenthaler SF, Kuhn PH, Stauffer D, Rovelli G, Martoglio B
disease. Neurosci Lett 350:61–65 (2006) SPPL2a and SPPL2b promote intramembrane proteolysis
De Lella Ezcurra AL, Chertoff M, Ferrari C, Graciarena M, Pitossi F of TNFalpha in activated dendritic cells to trigger IL-12
(2010) Chronic expression of low levels of tumor necrosis factor- production. Nat Cell Biol 8:843–848
alpha in the substantia nigra elicits progressive neurodegeneration, Fujii S, Matsumoto M, Igarashi K, Kato H, Mikoshiba K (2000)
delayed motor symptoms and microglia/macrophage activation. Synaptic plasticity in hippocampal CA1 neurons of mice
Neurobiol Dis 37:630–640 lacking type 1 inositol-1,4,5-trisphosphate receptors. Learn
DeKosky ST, Scheff SW (1990) Synapse loss in frontal cortex Mem 7:312–320
biopsies in Alzheimer’s disease: correlation with cognitive Gaikwad S, Larionov S, Wang Y, Dannenberg H, Matozaki T,
severity. Ann Neurol 27:457–464 Monsonego A, Thal DR, Neumann H (2009) Signal regulatory
Deng W, Aimone JB, Gage FH (2010) New neurons and new protein-beta1: a microglial modulator of phagocytosis in
memories: how does adult hippocampal neurogenesis affect Alzheimer’s disease. Am J Pathol 175:2528–2539
learning and memory? Nat Rev Neurosci 11:339–350 Gerhard A, Pavese N, Hotton G, Turkheimer F, Es M, Hammers A,
Depuydt B, van Loo G, Vandenabeele P, Declercq W (2005) Induction Eggert K, Oertel W, Banati RB, Brooks DJ (2006) In vivo
of apoptosis by TNF receptor 2 in a T-cell hybridoma is FADD imaging of microglial activation with [11C](R)-PK11195 PET in
dependent and blocked by caspase-8 inhibitors. J Cell Sci idiopathic Parkinson’s disease. Neurobiol Dis 21:404–412
118:497–504 Giuliani F, Vernay A, Leuba G, Schenk F (2009) Decreased behavioral
Dickson DW (1997) The pathogenesis of senile plaques. J Neuro- impairments in an Alzheimer mice model by interfering with
pathol Exp Neurol 56:321–339 TNF-alpha metabolism. Brain Res Bull 80:302–308
Dieude P, Petit E, Cailleau-Moindrault S, Osorio J, Pierlot C, Martinez Golan H, Levav T, Mendelsohn A, Huleihel M (2004) Involvement of
M, Faure S, Alibert O, Lasbleiz S, De Toma C, Bardin T, Prum B, tumor necrosis factor alpha in hippocampal development and
Cornelis F (2002) Association between tumor necrosis factor function. Cereb Cortex 14:97–105
receptor II and familial, but not sporadic, rheumatoid arthritis: Gorell JM, Peterson EL, Rybicki BA, Johnson CC (2004) Multiple
evidence for genetic heterogeneity. Arthritis Rheum 46:2039– risk factors for Parkinson’s disease. J Neurol Sci 217:169–174
2044 Grell M (1995) Tumor necrosis factor (TNF) receptors in cellular
Ding JY, Kreipke CW, Schafer P, Schafer S, Speirs SL, Rafols JA signaling of soluble and membrane-expressed TNF. J Inflamm
(2009) Synapse loss regulated by matrix metalloproteinases in 47:8–17
traumatic brain injury is associated with hypoxia inducible factor- Grell M, Douni E, Wajant H, Lohden M, Clauss M, Maxeiner B,
1alpha expression. Brain Res 1268:125–134 Georgopoulos S, Lesslauer W, Kollias G, Pfizenmaier K,
Dobbs RJ, Charlett A, Purkiss AG, Dobbs SM, Weller C, Peterson Scheurich P (1995) The transmembrane form of tumor necrosis
DW (1999) Association of circulating TNF-alpha and IL-6 with factor is the prime activating ligand of the 80 kDa tumor necrosis
ageing and parkinsonism. Acta Neurol Scand 100:34–41 factor receptor. Cell 83:793–802
Drouet C, Shakhov AN, Jongeneel CV (1991) Enhancers and Hanisch UK (2002) Microglia as a source and target of cytokines. Glia
transcription factors controlling the inducibility of the tumor 40:140–155
necrosis factor-alpha promoter in primary macrophages. J Harashima S, Horiuchi T, Hatta N, Morita C, Higuchi M, Sawabe T,
Immunol 147:1694–1700 Tsukamoto H, Tahira T, Hayashi K, Fujita S, Niho Y (2001)
Eissner G, Kirchner S, Lindner H, Kolch W, Janosch P, Grell M, Outside-to-inside signal through the membrane TNF-alpha
Scheurich P, Andreesen R, Holler E (2000) Reverse signaling induces E-selectin (CD62E) expression on activated human
through transmembrane TNF confers resistance to lipopolysac- CD4+ T cells. J Immunol 166:130–136
charide in human monocytes and macrophages. J Immunol Hardy JA, Higgins GA (1992) Alzheimer’s disease: the amyloid
164:6193–6198 cascade hypothesis. Science 256:184–185
Eissner G, Kolch W, Scheurich P (2004) Ligands working as Haridas V, Darnay BG, Natarajan K, Heller R, Aggarwal BB (1998)
receptors: reverse signaling by members of the TNF superfamily Overexpression of the p80 TNF receptor leads to TNF-dependent
56 J Neuroimmune Pharmacol (2012) 7:42–59

apoptosis, nuclear factor-kappa B activation, and c-Jun kinase Activation of peroxisome proliferator-activated receptor isoforms
activation. J Immunol 160:3152–3162 and inhibition of prostaglandin H(2) synthases by ibuprofen,
He P, Zhong Z, Lindholm K, Berning L, Lee W, Lemere C, naproxen, and indomethacin. Biochem Pharmacol 62:1587–1595
Staufenbiel M, Li R, Shen Y (2007) Deletion of tumor Kassiotis G, Kollias G (2001) Uncoupling the proinflammatory
necrosis factor death receptor inhibits amyloid beta generation from the immunosuppressive properties of tumor necrosis
and prevents learning and memory deficits in Alzheimer’s factor (TNF) at the p55 TNF receptor level: implications for
mice. J Cell Biol 178:829–841 pathogenesis and therapy of autoimmune demyelination. J Exp
Heldmann U, Thored P, Claasen JH, Arvidsson A, Kokaia Z, Lindvall Med 193:427–434
O (2005) TNF-alpha antibody infusion impairs survival of Keohane A, Ryan S, Maloney E, Sullivan AM, Nolan YM (2010)
stroke-generated neuroblasts in adult rat brain. Exp Neurol Tumour necrosis factor-alpha impairs neuronal differentiation but
196:204–208 not proliferation of hippocampal neural precursor cells: Role of
Heller RA, Song K, Fan N, Chang DJ (1992) The p70 tumor necrosis Hes1. Mol Cell Neurosci 43:127–135
factor receptor mediates cytotoxicity. Cell 70:47–56 Klegeris A, McGeer PL (2005) Non-steroidal anti-inflammatory drugs
Heneka MT, O’Banion MK (2007) Inflammatory processes in (NSAIDs) and other anti-inflammatory agents in the treatment of
Alzheimer’s disease. J Neuroimmunol 184:69–91 neurodegenerative disease. Curr Alzheimer Res 2:355–365
Hermann GE, Rogers RC, Bresnahan JC, Beattie MS (2001) Tumor Klintworth H, Garden G, Xia Z (2009) Rotenone and paraquat do not
necrosis factor-alpha induces cFOS and strongly potentiates directly activate microglia or induce inflammatory cytokine
glutamate-mediated cell death in the rat spinal cord. Neurobiol release. Neurosci Lett 462:1–5
Dis 8:590–599 Koenigsknecht J, Landreth G (2004) Microglial phagocytosis of
Hickman SE, Allison EK, El Khoury J (2008) Microglial dysfunction fibrillar beta-amyloid through a beta1 integrin-dependent mech-
and defective beta-amyloid clearance pathways in aging anism. J Neurosci 24:9838–9846
Alzheimer’s disease mice. J Neurosci 28:8354–8360 Kontoyiannis D, Pasparakis M, Pizarro TT, Cominelli F, Kollias G
Hofman FM, Hinton DR, Johnson K, Merrill JE (1989) Tumor (1999) Impaired on/off regulation of TNF biosynthesis in mice
necrosis factor identified in multiple sclerosis brain. J Exp Med lacking TNF AU-rich elements: implications for joint and gut-
170:607–612 associated immunopathologies. Immunity 10:387–398
Hsu H, Xiong J, Goeddel DV (1995) The TNF receptor 1-associated Korner H, Riminton DS, Strickland DH, Lemckert FA, Pollard JD,
protein TRADD signals cell death and NF-kappa B activation. Sedgwick JD (1997) Critical points of tumor necrosis factor
Cell 81:495–504 action in central nervous system autoimmune inflammation
Huang ZB, Eden E (1993) Effect of corticosteroids on IL1 beta and defined by gene targeting. J Exp Med 186:1585–1590
TNF alpha release by alveolar macrophages from patients with Kronke M (1999) Involvement of sphingomyelinases in TNF
AIDS and Pneumocystis carinii pneumonia. Chest 104:751–755 signaling pathways. Chem Phys Lipids 102:157–166
Idriss HT, Naismith JH (2000) TNF alpha and the TNF receptor Kuo LH, Hu MK, Hsu WM, Tung YT, Wang BJ, Tsai WW, Yen CT,
superfamily: structure-function relationship(s). Microsc Res Tech Liao YF (2008) Tumor necrosis factor-alpha-elicited stimulation
50:184–195 of gamma-secretase is mediated by c-Jun N-terminal kinase-
Imamura K, Hishikawa N, Sawada M, Nagatsu T, Yoshida M, dependent phosphorylation of presenilin and nicastrin. Mol Biol
Hashizume Y (2003) Distribution of major histocompatibility Cell 19:4201–4212
complex class II-positive microglia and cytokine profile of Lassmann H, Bruck W, Lucchinetti CF (2007) The immunopathology
Parkinson’s disease brains. Acta Neuropathol 106:518–526 of multiple sclerosis: an overview. Brain Pathol 17:210–218
in t’ Veld BA, Ruitenberg A, Hofman A, Launer LJ, van Duijn CM, Lau LT, Yu AC (2001) Astrocytes produce and release interleukin-1,
Stijnen T, Breteler MM, Stricker BH (2001) Nonsteroidal interleukin-6, tumor necrosis factor alpha and interferon-gamma
antiinflammatory drugs and the risk of Alzheimer’s disease. N following traumatic and metabolic injury. J Neurotrauma 18:351–359
Engl J Med 345:1515–1521 Laws SM, Perneczky R, Wagenpfeil S, Muller U, Forstl H, Martins
Iosif RE, Ekdahl CT, Ahlenius H, Pronk CJ, Bonde S, Kokaia Z, RN, Kurz A, Riemenschneider M (2005) TNF polymorphisms in
Jacobsen SE, Lindvall O (2006) Tumor necrosis factor receptor 1 Alzheimer disease and functional implications on CSF beta-
is a negative regulator of progenitor proliferation in adult amyloid levels. Hum Mutat 26:29–35
hippocampal neurogenesis. J Neurosci 26:9703–9712 Lee TH, Huang Q, Oikemus S, Shank J, Ventura JJ, Cusson N,
Iosif RE, Ahlenius H, Ekdahl CT, Darsalia V, Thored P, Jovinge S, Vaillancourt RR, Su B, Davis RJ, Kelliher MA (2003) The death
Kokaia Z, Lindvall O (2008) Suppression of stroke-induced domain kinase RIP1 is essential for tumor necrosis factor alpha
progenitor proliferation in adult subventricular zone by tumor signaling to p38 mitogen-activated protein kinase. Mol Cell Biol
necrosis factor receptor 1. J Cereb Blood Flow Metab 23:8377–8385
28:1574–1587 Leng A, Mura A, Feldon J, Ferger B (2005) Tumor necrosis factor-
Janelsins MC, Mastrangelo MA, Oddo S, LaFerla FM, Federoff HJ, alpha receptor ablation in a chronic MPTP mouse model of
Bowers WJ (2005) Early correlation of microglial activation with Parkinson’s disease. Neurosci Lett 375:107–111
enhanced tumor necrosis factor-alpha and monocyte chemo- Lesage S, Brice A (2009) Parkinson’s disease: from monogenic forms
attractant protein-1 expression specifically within the entorhinal to genetic susceptibility factors. Hum Mol Genet 18:R48–59
cortex of triple transgenic Alzheimer’s disease mice. J Neuro- Li R, Yang L, Lindholm K, Konishi Y, Yue X, Hampel H, Zhang D,
inflammation 2:23 Shen Y (2004) Tumor necrosis factor death receptor signaling
Janelsins MC, Mastrangelo MA, Park KM, Sudol KL, Narrow WC, Oddo cascade is required for amyloid-beta protein-induced neuron
S, LaFerla FM, Callahan LM, Federoff HJ, Bowers WJ (2008) death. J Neurosci 24:1760–1771
Chronic neuron-specific tumor necrosis factor-alpha expression Liao YF, Wang BJ, Cheng HT, Kuo LH, Wolfe MS (2004) Tumor
enhances the local inflammatory environment ultimately leading to necrosis factor-alpha, interleukin-1beta, and interferon-gamma
neuronal death in 3xTg-AD mice. Am J Pathol 173:1768–1782 stimulate gamma-secretase-mediated cleavage of amyloid precur-
Jankovic J (2005) Searching for a relationship between manganese sor protein through a JNK-dependent MAPK pathway. J Biol
and welding and Parkinson’s disease. Neurology 64:2021–2028 Chem 279:49523–49532
Jaradat MS, Wongsud B, Phornchirasilp S, Rangwala SM, Shams G, Lie DC, Song H, Colamarino SA, Ming GL, Gage FH (2004)
Sutton M, Romstedt KJ, Noonan DJ, Feller DR (2001) Neurogenesis in the adult brain: new strategies for central
J Neuroimmune Pharmacol (2012) 7:42–59 57

nervous system diseases. Annu Rev Pharmacol Toxicol Merz PA, Wisniewski HM, Somerville RA, Bobin SA, Masters
44:399–421 CL, Iqbal K (1983) Ultrastructural morphology of amyloid
Lieberman AP, Pitha PM, Shin HS, Shin ML (1989) Production of fibrils from neuritic and amyloid plaques. Acta Neuropathol
tumor necrosis factor and other cytokines by astrocytes stimulated 60:113–124
with lipopolysaccharide or a neurotropic virus. Proc Natl Acad Sci Mogi M, Harada M, Riederer P, Narabayashi H, Fujita K, Nagatsu T
USA 86:6348–6352 (1994) Tumor necrosis factor-alpha (TNF-alpha) increases both
Liu T, Clark RK, McDonnell PC, Young PR, White RF, Barone FC, in the brain and in the cerebrospinal fluid from parkinsonian
Feuerstein GZ (1994) Tumor necrosis factor-alpha expression in patients. Neurosci Lett 165:208–210
ischemic neurons. Stroke 25:1481–1488 Mogi M, Harada M, Kondo T, Riederer P, Nagatsu T (1995) Brain beta
Liu Y, Walter S, Stagi M, Cherny D, Letiembre M, Schulz-Schaeffer 2-microglobulin levels are elevated in the striatum in Parkinson’s
W, Heine H, Penke B, Neumann H, Fassbender K (2005a) LPS disease. J Neural Transm Park Dis Dement Sect 9:87–92
receptor (CD14): a receptor for phagocytosis of Alzheimer’s Mogi M, Togari A, Tanaka K, Ogawa N, Ichinose H, Nagatsu T
amyloid peptide. Brain 128:1778–1789 (1999) Increase in level of tumor necrosis factor (TNF)-alpha in
Liu YP, Lin HI, Tzeng SF (2005b) Tumor necrosis factor-alpha and 6-hydroxydopamine-lesioned striatum in rats without influence
interleukin-18 modulate neuronal cell fate in embryonic neural of systemic L-DOPA on the TNF-alpha induction. Neurosci Lett
progenitor culture. Brain Res 1054:152–158 268:101–104
Ma SL, Tang NL, Lam LC, Chiu HF (2004) Association between Mogi M, Togari A, Kondo T, Mizuno Y, Komure O, Kuno S, Ichinose
tumor necrosis factor-alpha promoter polymorphism and H, Nagatsu T (2000) Caspase activities and tumor necrosis factor
Alzheimer’s disease. Neurology 62:307–309 receptor R1 (p55) level are elevated in the substantia nigra from
MacEwan DJ (2002) TNF ligands and receptors–a matter of life and parkinsonian brain. J Neural Transm 107:335–341
death. Br J Pharmacol 135:855–875 Mohan N, Edwards ET, Cupps TR, Oliverio PJ, Sandberg G, Crayton
Mackenzie IR, Munoz DG (1998) Nonsteroidal anti-inflammatory H, Richert JR, Siegel JN (2001) Demyelination occurring during
drug use and Alzheimer-type pathology in aging. Neurology anti-tumor necrosis factor alpha therapy for inflammatory
50:986–990 arthritides. Arthritis Rheum 44:2862–2869
Magnusson C, Vaux DL (1999) Signalling by CD95 and TNF Monden Y, Kubota T, Inoue T, Tsutsumi T, Kawano S, Ide T, Tsutsui H,
receptors: not only life and death. Immunol Cell Biol 77:41–46 Sunagawa K (2007) Tumor necrosis factor-alpha is toxic via
Mandrekar S, Jiang Q, Lee CY, Koenigsknecht-Talboo J, Holtzman receptor 1 and protective via receptor 2 in a murine model of
DM, Landreth GE (2009) Microglia mediate the clearance of myocardial infarction. Am J Physiol Heart Circ Physiol 293:
soluble Abeta through fluid phase macropinocytosis. J Neurosci H743–753
29:4252–4262 Morganti-Kossman MC, Lenzlinger PM, Hans V, Stahel P, Csuka E,
Marchetti L, Klein M, Schlett K, Pfizenmaier K, Eisel UL (2004) Ammann E, Stocker R, Trentz O, Kossmann T (1997) Production
Tumor necrosis factor (TNF)-mediated neuroprotection against of cytokines following brain injury: beneficial and deleterious for
glutamate-induced excitotoxicity is enhanced by N-methyl-D- the damaged tissue. Mol Psychiatry 2:133–136
aspartate receptor activation. Essential role of a TNF receptor 2- Natoli G, Costanzo A, Ianni A, Templeton DJ, Woodgett JR, Balsano
mediated phosphatidylinositol 3-kinase-dependent NF-kappa B C, Levrero M (1997) Activation of SAPK/JNK by TNF receptor
pathway. J Biol Chem 279:32869–32881 1 through a noncytotoxic TRAF2-dependent pathway. Science
McAlpine FE, Lee JK, Harms AS, Ruhn KA, Blurton-Jones M, Hong 275:200–203
J, Das P, Golde TE, LaFerla FM, Oddo S, Blesch A, Tansey MG Nedwin GE, Naylor SL, Sakaguchi AY, Smith D, Jarrett-Nedwin J,
(2009) Inhibition of soluble TNF signaling in a mouse model of Pennica D, Goeddel DV, Gray PW (1985) Human lymphotoxin
Alzheimer’s disease prevents pre-plaque amyloid-associated and tumor necrosis factor genes: structure, homology and
neuropathology. Neurobiol Dis 34:163–177 chromosomal localization. Nucleic Acids Res 13:6361–6373
McCoy MK, Tansey MG (2008) TNF signaling inhibition in the CNS: Neumann H, Schweigreiter R, Yamashita T, Rosenkranz K, Wekerle H,
implications for normal brain function and neurodegenerative Barde YA (2002) Tumor necrosis factor inhibits neurite out-
disease. J Neuroinflammation 5:45 growth and branching of hippocampal neurons by a rho-
McCoy MK, Martinez TN, Ruhn KA, Szymkowski DE, Smith CG, dependent mechanism. J Neurosci 22:854–862
Botterman BR, Tansey KE, Tansey MG (2006) Blocking soluble Nishimura M, Mizuta I, Mizuta E, Yamasaki S, Ohta M, Kaji R, Kuno
tumor necrosis factor signaling with dominant-negative tumor S (2001) Tumor necrosis factor gene polymorphisms in patients
necrosis factor inhibitor attenuates loss of dopaminergic neurons with sporadic Parkinson’s disease. Neurosci Lett 311:1–4
in models of Parkinson’s disease. J Neurosci 26:9365–9375 Nomura T et al (2011) Therapeutic effect of PEGylated TNFR1-
McGeer PL, Itagaki S, Boyes BE, McGeer EG (1988) Reactive microglia selective antagonistic mutant TNF in experimental autoimmune
are positive for HLA-DR in the substantia nigra of Parkinson’s and encephalomyelitis mice. J Control Release 149:8–14
Alzheimer’s disease brains. Neurology 38:1285–1291 Obregon E, Punzon C, Fernandez-Cruz E, Fresno M, Munoz-
Medeiros R, Prediger RD, Passos GF, Pandolfo P, Duarte FS, Fernandez MA (1999) HIV-1 infection induces differentiation of
Franco JL, Dafre AL, Di Giunta G, Figueiredo CP, Takahashi immature neural cells through autocrine tumor necrosis factor
RN, Campos MM, Calixto JB (2007) Connecting TNF-alpha and nitric oxide production. Virology 261:193–204
signaling pathways to iNOS expression in a mouse model of Ogoshi F, Yin HZ, Kuppumbatti Y, Song B, Amindari S, Weiss JH
Alzheimer’s disease: relevance for the behavioral and synaptic (2005) Tumor necrosis-factor-alpha (TNF-alpha) induces rapid
deficits induced by amyloid beta protein. J Neurosci 27:5394– insertion of Ca2+ −permeable alpha-amino-3-hydroxyl-5-
5404 methyl-4-isoxazole-propionate (AMPA)/kainate (Ca-A/K)
Mehlhorn G, Hollborn M, Schliebs R (2000) Induction of cytokines in channels in a subset of hippocampal pyramidal neurons. Exp
glial cells surrounding cortical beta-amyloid plaques in transgen- Neurol 193:384–393
ic Tg2576 mice with Alzheimer pathology. Int J Dev Neurosci Oppenheim RW (1996) Neurotrophic survival molecules for moto-
18:423–431 neurons: an embarrassment of riches. Neuron 17:195–197
Menet V, Gimenez YRM, Sandillon F, Privat A (2000) GFAP null Paganelli R, Di Iorio A, Patricelli L, Ripani F, Sparvieri E, Faricelli
astrocytes are a favorable substrate for neuronal survival and R, Iarlori C, Porreca E, Di Gioacchino M, Abate G (2002)
neurite growth. Glia 31:267–272 Proinflammatory cytokines in sera of elderly patients with
58 J Neuroimmune Pharmacol (2012) 7:42–59

dementia: levels in vascular injury are higher than those of Scharf S, Mander A, Ugoni A, Vajda F, Christophidis N (1999) A
mild-moderate Alzheimer’s disease patients. Exp Gerontol double-blind, placebo-controlled trial of diclofenac/misoprostol
37:257–263 in Alzheimer’s disease. Neurology 53:197–201
Park KM, Yule DI, Bowers WJ (2008) Tumor necrosis factor-alpha Scheff SW, Price DA, Schmitt FA, Mufson EJ (2006) Hippocampal
potentiates intraneuronal Ca2+ signaling via regulation of the synaptic loss in early Alzheimer’s disease and mild cognitive
inositol 1,4,5-trisphosphate receptor. J Biol Chem 283:33069– impairment. Neurobiol Aging 27:1372–1384
33079 Schottelius AJ, Mayo MW, Sartor RB, Baldwin AS Jr (1999)
Pettmann B, Henderson CE (1998) Neuronal cell death. Neuron Interleukin-10 signaling blocks inhibitor of kappaB kinase
20:633–647 activity and nuclear factor kappaB DNA binding. J Biol Chem
Pieper HC, Evert BO, Kaut O, Riederer PF, Waha A, Wullner U (2008) 274:31868–31874
Different methylation of the TNF-alpha promoter in cortex and Seckinger P, Zhang JH, Hauptmann B, Dayer JM (1990) Character-
substantia nigra: Implications for selective neuronal vulnerability. ization of a tumor necrosis factor alpha (TNF-alpha) inhibitor:
Neurobiol Dis 32:521–527 evidence of immunological cross-reactivity with the TNF
Pierik M, Vermeire S, Steen KV, Joossens S, Claessens G, Vlietinck R, receptor. Proc Natl Acad Sci USA 87:5188–5192
Rutgeerts P (2004) Tumour necrosis factor-alpha receptor 1 and 2 Sedel F, Bechade C, Vyas S, Triller A (2004) Macrophage-derived
polymorphisms in inflammatory bowel disease and their associ- tumor necrosis factor alpha, an early developmental signal for
ation with response to infliximab. Aliment Pharmacol Ther motoneuron death. J Neurosci 24:2236–2246
20:303–310 Selmaj K, Raine CS, Cross AH (1991) Anti-tumor necrosis factor
Pimplikar SW (2009) Reassessing the amyloid cascade hypothesis of therapy abrogates autoimmune demyelination. Ann Neurol
Alzheimer’s disease. Int J Biochem Cell Biol 41:1261–1268 30:694–700
Pocsik E, Duda E, Wallach D (1995) Phosphorylation of the 26 kDa Sfikakis PP (2010) The first decade of biologic TNF antagonists in
TNF precursor in monocytic cells and in transfected HeLa cells. J clinical practice: lessons learned, unresolved issues and future
Inflamm 45:152–160 directions. Curr Dir Autoimmun 11:180–210
Pope RM, Leutz A, Ness SA (1994) C/EBP beta regulation of the Sharief MK, Hentges R (1991) Association between tumor necrosis
tumor necrosis factor alpha gene. J Clin Invest 94:1449–1455 factor-alpha and disease progression in patients with multiple
Priyadarshi A, Khuder SA, Schaub EA, Priyadarshi SS (2001) sclerosis. N Engl J Med 325:467–472
Environmental risk factors and Parkinson’s disease: a metaanal- Shen Y, Li R, Shiosaki K (1997) Inhibition of p75 tumor necrosis
ysis. Environ Res 86:122–127 factor receptor by antisense oligonucleotides increases hypoxic
Probert L, Akassoglou K, Pasparakis M, Kontogeorgos G, Kollias G injury and beta-amyloid toxicity in human neuronal cell line. J
(1995) Spontaneous inflammatory demyelinating disease in Biol Chem 272:3550–3553
transgenic mice showing central nervous system-specific expres- Sheng WS, Hu S, Ni HT, Rowen TN, Lokensgard JR, Peterson PK
sion of tumor necrosis factor alpha. Proc Natl Acad Sci USA (2005) TNF-alpha-induced chemokine production and apoptosis
92:11294–11298 in human neural precursor cells. J Leukoc Biol 78:1233–1241
Reed-Geaghan EG, Savage JC, Hise AG, Landreth GE (2009) CD14 Shi JQ, Shen W, Chen J, Wang BR, Zhong LL, Zhu YW, Zhu HQ,
and toll-like receptors 2 and 4 are required for fibrillar A{beta}- Zhang QQ, Zhang YD, Xu J (2011) Anti-TNF-alpha reduces
stimulated microglial activation. J Neurosci 29:11982–11992 amyloid plaques and tau phosphorylation and induces CD11c-
Rothe M, Wong SC, Henzel WJ, Goeddel DV (1994) A novel family positive dendritic-like cell in the APP/PS1 transgenic mouse
of putative signal transducers associated with the cytoplasmic brains. Brain Res 1368:239–247
domain of the 75 kDa tumor necrosis factor receptor. Cell Shinomiya S, Naraba H, Ueno A, Utsunomiya I, Maruyama T,
78:681–692 Ohuchida S, Ushikubi F, Yuki K, Narumiya S, Sugimoto Y,
Rothe M, Sarma V, Dixit VM, Goeddel DV (1995a) TRAF2-mediated Ichikawa A, Oh-ishi S (2001) Regulation of TNFalpha and
activation of NF-kappa B by TNF receptor 2 and CD40. Science interleukin-10 production by prostaglandins I(2) and E(2):
269:1424–1427 studies with prostaglandin receptor-deficient mice and prosta-
Rothe M, Pan MG, Henzel WJ, Ayres TM, Goeddel DV (1995b) The glandin E-receptor subtype-selective synthetic agonists. Biochem
TNFR2-TRAF signaling complex contains two novel proteins Pharmacol 61:1153–1160
related to baculoviral inhibitor of apoptosis proteins. Cell Skovronsky DM, Moore DB, Milla ME, Doms RW, Lee VM (2000)
83:1243–1252 Protein kinase C-dependent alpha-secretase competes with beta-
Rousselet E, Callebert J, Parain K, Joubert C, Hunot S, Hartmann A, secretase for cleavage of amyloid-beta precursor protein in the
Jacque C, Perez-Diaz F, Cohen-Salmon C, Launay JM, Hirsch trans-golgi network. J Biol Chem 275:2568–2575
EC (2002) Role of TNF-alpha receptors in mice intoxicated with Spillantini MG, Schmidt ML, Lee VM, Trojanowski JQ, Jakes R,
the parkinsonian toxin MPTP. Exp Neurol 177:183–192 Goedert M (1997) Alpha-synuclein in Lewy bodies. Nature
Ruan L, Kang Z, Pei G, Le Y (2009) Amyloid deposition and 388:839–840
inflammation in APPswe/PS1dE9 mouse model of Alzheimer’s Sriram K, Matheson JM, Benkovic SA, Miller DB, Luster MI,
disease. Curr Alzheimer Res 6:531–540 O’Callaghan JP (2002) Mice deficient in TNF receptors are
Sanfilipo MP, Benedict RH, Weinstock-Guttman B, Bakshi R protected against dopaminergic neurotoxicity: implications for
(2006) Gray and white matter brain atrophy and neuropsy- Parkinson’s disease. FASEB J 16:1474–1476
chological impairment in multiple sclerosis. Neurology Sriram K, Matheson JM, Benkovic SA, Miller DB, Luster MI,
66:685–692 O’Callaghan JP (2006) Deficiency of TNF receptors suppresses
Sashio H, Tamura K, Ito R, Yamamoto Y, Bamba H, Kosaka T, Fukui microglial activation and alters the susceptibility of brain regions
S, Sawada K, Fukuda Y, Satomi M, Shimoyama T, Furuyama J to MPTP-induced neurotoxicity: role of TNF-alpha. FASEB J
(2002) Polymorphisms of the TNF gene and the TNF receptor 20:670–682
superfamily member 1B gene are associated with susceptibility to Steinmetz CC, Turrigiano GG (2010) Tumor necrosis factor-alpha
ulcerative colitis and Crohn’s disease, respectively. Immunoge- signaling maintains the ability of cortical synapses to express
netics 53:1020–1027 synaptic scaling. J Neurosci 30:14685–14690
Schapira AH (2009) Neurobiology and treatment of Parkinson’s Stellwagen D, Malenka RC (2006) Synaptic scaling mediated by glial
disease. Trends Pharmacol Sci 30:41–47 TNF-alpha. Nature 440:1054–1059
J Neuroimmune Pharmacol (2012) 7:42–59 59

Stemple DL, Anderson DJ (1992) Isolation of a stem cell for neurons TRAF2 and c-IAP1 and c-IAP2 to suppress caspase-8 activation.
and glia from the mammalian neural crest. Cell 71:973–985 Science 281:1680–1683
Stewart WF, Kawas C, Corrada M, Metter EJ (1997) Risk of Watts AD, Hunt NH, Wanigasekara Y, Bloomfield G, Wallach D,
Alzheimer’s disease and duration of NSAID use. Neurology Roufogalis BD, Chaudhri G (1999) A casein kinase I motif
48:626–632 present in the cytoplasmic domain of members of the tumour
Suvannavejh GC, Lee HO, Padilla J, Dal Canto MC, Barrett TA, necrosis factor ligand family is implicated in ‘reverse signalling’.
Miller SD (2000) Divergent roles for p55 and p75 tumor necrosis EMBO J 18:2119–2126
factor receptors in the pathogenesis of MOG(35–55)-induced Wedel A, Sulski G, Ziegler-Heitbrock HW (1996) CCAAT/enhancer
experimental autoimmune encephalomyelitis. Cell Immunol binding protein is involved in the expression of the tumour
205:24–33 necrosis factor gene in human monocytes. Cytokine 8:335–341
Tartaglia LA, Pennica D, Goeddel DV (1993a) Ligand passing: the Weihofen A, Binns K, Lemberg MK, Ashman K, Martoglio B (2002)
75-kDa tumor necrosis factor (TNF) receptor recruits TNF for Identification of signal peptide peptidase, a presenilin-type
signaling by the 55-kDa TNF receptor. J Biol Chem aspartic protease. Science 296:2215–2218
268:18542–18548 Weiss T, Grell M, Hessabi B, Bourteele S, Muller G, Scheurich P, Wajant
Tartaglia LA, Ayres TM, Wong GH, Goeddel DV (1993b) A novel H (1997) Enhancement of TNF receptor p60-mediated cytotoxicity
domain within the 55 kd TNF receptor signals cell death. Cell by TNF receptor p80: requirement of the TNF receptor-associated
74:845–853 factor-2 binding site. J Immunol 158:2398–2404
Temple S (2001) The development of neural stem cells. Nature Weiss T, Grell M, Siemienski K, Muhlenbeck F, Durkop H,
414:112–117 Pfizenmaier K, Scheurich P, Wajant H (1998) TNFR80-
Titelbaum DS, Degenhardt A, Kinkel RP (2005) Anti-tumor necrosis dependent enhancement of TNFR60-induced cell death is
factor alpha-associated multiple sclerosis. AJNR Am J Neuro- mediated by TNFR-associated factor 2 and is specific for
radiol 26:1548–1550 TNFR60. J Immunol 161:3136–3142
Tobinick EL, Gross H (2008) Rapid improvement in verbal fluency Wheeler D, Knapp E, Bandaru VV, Wang Y, Knorr D, Poirier C,
and aphasia following perispinal etanercept in Alzheimer’s Mattson MP, Geiger JD, Haughey NJ (2009) Tumor necrosis
disease. BMC Neurol 8:27 factor-alpha-induced neutral sphingomyelinase-2 modulates syn-
Tobinick E, Gross H, Weinberger A, Cohen H (2006) TNF-alpha aptic plasticity by controlling the membrane insertion of NMDA
modulation for treatment of Alzheimer’s disease: a 6-month pilot receptors. J Neurochem 109:1237–1249
study. MedGenMed 8:25 Winkelmann A, Patejdl R, Wagner S, Benecke R, Zettl UK (2008)
Tracey D, Klareskog L, Sasso EH, Salfeld JG, Tak PP (2008) Tumor Cerebral MRI lesions and anti-tumor necrosis factor-alpha
necrosis factor antagonist mechanisms of action: a comprehen- therapy. J Neurol 255(Suppl 6):109–114
sive review. Pharmacol Ther 117:244–279 Winston BW, Lange-Carter CA, Gardner AM, Johnson GL, Riches
Trapp BD, Peterson J, Ransohoff RM, Rudick R, Mork S, Bo L (1998) DW (1995) Tumor necrosis factor alpha rapidly activates the
Axonal transection in the lesions of multiple sclerosis. N Engl J mitogen-activated protein kinase (MAPK) cascade in a MAPK
Med 338:278–285 kinase kinase-dependent, c-Raf-1-independent fashion in mouse
Turrigiano GG, Leslie KR, Desai NS, Rutherford LC, Nelson SB macrophages. Proc Natl Acad Sci USA 92:1614–1618
(1998) Activity-dependent scaling of quantal amplitude in Yamamoto M, Kiyota T, Horiba M, Buescher JL, Walsh SM,
neocortical neurons. Nature 391:892–896 Gendelman HE, Ikezu T (2007) Interferon-gamma and tumor
Udalova IA, Knight JC, Vidal V, Nedospasov SA, Kwiatkowski D necrosis factor-alpha regulate amyloid-beta plaque deposition and
(1998) Complex NF-kappaB interactions at the distal tumor beta-secretase expression in Swedish mutant APP transgenic
necrosis factor promoter region in human monocytes. J Biol mice. Am J Pathol 170:680–692
Chem 273:21178–21186 Yamane H, Sugimoto Y, Tanaka S, Ichikawa A (2000) Prostaglandin E
van Essen D, Kikutani H, Gray D (1995) CD40 ligand-transduced co- (2) receptors, EP2 and EP4, differentially modulate TNF-alpha
stimulation of T cells in the development of helper function. and IL-6 production induced by lipopolysaccharide in mouse
Nature 378:620–623 peritoneal neutrophils. Biochem Biophys Res Commun 278:224–
van Oosten BW, Barkhof F, Truyen L, Boringa JB, Bertelsmann FW, 228
von Blomberg BM, Woody JN, Hartung HP, Polman CH (1996) Yang L, Lindholm K, Konishi Y, Li R, Shen Y (2002) Target depletion
Increased MRI activity and immune activation in two multiple of distinct tumor necrosis factor receptor subtypes reveals
sclerosis patients treated with the monoclonal anti-tumor necrosis hippocampal neuron death and survival through different signal
factor antibody cA2. Neurology 47:1531–1534 transduction pathways. J Neurosci 22:3025–3032
Vlad SC, Miller DR, Kowall NW, Felson DT (2008) Protective effects Yao J, Mackman N, Edgington TS, Fan ST (1997) Lipopolysaccharide
of NSAIDs on the development of Alzheimer disease. Neurology induction of the tumor necrosis factor-alpha promoter in human
70:1672–1677 monocytic cells. Regulation by Egr-1, c-Jun, and NF-kappaB
Waetzig GH, Seegert D, Rosenstiel P, Nikolaus S, Schreiber S (2002) transcription factors. J Biol Chem 272:17795–17801
p38 mitogen-activated protein kinase is activated and linked to Zhang H, Yan D, Shi X, Liang H, Pang Y, Qin N, Chen H, Wang J,
TNF-alpha signaling in inflammatory bowel disease. J Immunol Yin B, Jiang X, Feng W, Zhang W, Zhou M, Li Z (2008)
168:5342–5351 Transmembrane TNF-alpha mediates “forward” and “reverse”
Wajant H, Pfizenmaier K, Scheurich P (2003) Tumor necrosis factor signaling, inducing cell death or survival via the NF-kappaB
signaling. Cell Death Differ 10:45–65 pathway in Raji Burkitt lymphoma cells. J Leukoc Biol
Wallach D, Engelmann H, Nophar Y, Aderka D, Kemper O, Hornik 84:789–797
V, Holtmann H, Brakebusch C (1991) Soluble and cell surface Zhao X, Bausano B, Pike BR, Newcomb-Fernandez JK, Wang KK,
receptors for tumor necrosis factor. Agents Actions Suppl Shohami E, Ringger NC, DeFord SM, Anderson DK, Hayes RL
35:51–57 (2001) TNF-alpha stimulates caspase-3 activation and apoptotic
Wang CY, Mayo MW, Korneluk RG, Goeddel DV, Baldwin AS Jr cell death in primary septo-hippocampal cultures. J Neurosci Res
(1998) NF-kappaB antiapoptosis: induction of TRAF1 and 64:121–131

You might also like