AJA Asian Journal of Anesthesiology

Advancing, Capability, Improving lives

Review Article
Volume 50, Issue 4, Pages 178-182
Ching-Hui Shen 1.2 , Ru-Ying Tsai 3 , Chih-Shung Wong 3
2460 Views


Abstract

Opioids have been used as potent analgesics in clinics for decades; however, their long-term administration leads to tolerance. Two possible mechanisms for drug tolerance are postulated as within-system and between-systems adaptation. The within-system tolerance is involved in the signal transduction of opioid receptors, including downregulation of opioid receptors, uncoupling of G-protein from opioid receptors, and β-arrestin recruitment to opioid receptors, which causes receptor desensitization and internalization/endocytosis. The between-systems tolerance comprehends the glutamatergic receptor system and glial activation with the release of proinflammatory cytokines, and thus the analgesic effect of morphine is reduced. Tumor necrosis factor-α (TNF-α) is a vital proinflammatory cytokine and exerts either a neurotoxic or neuroprotective effect on different diseases of the central nervous system. TNF-α has also been demonstrated to correlate with neuronal plasticity via activation of spinal glial cells and enhancement of glutamatergic transmission. Previous studies had revealed an increased expression of TNF-α in morphine tolerance. This review article focuses on the role of TNF-α in neuroinflammation and the glutamatergic receptor system in morphine tolerance. It may provide another adjuvant therapy for morphine tolerance, which extends the effectiveness of opioids in clinical pain management.

Keywords

drug tolerance; inflammation: neural; morphine; synaptic transmission: glutamatergic; tumor necrosis factor-alpha;


1. Introduction

Opioids have a potent analgesic effect and have widely been used for decades. Long-term opioid administration develops tolerance, which limits its clinical efficacy. Koob and Bloom1 described two possible mechanisms for drug tolerance: within-system and between-systems adaptation. The within-system tolerance occurs by eliciting an opposite action within the same system; the between-systems tolerance is characterized by alterations in the primary drug-sensitive system adaptation not directly involved in the drug's primary action system. Investigations of the mechanisms of within-system included modulation of intracellular adenylyl cyclase (AC) and cAMP-dependent protein kinase A (PKA),23 uncoupling of G-protein signaling,45 increased binding of β-arrestin to opioid receptors,67 and μ-opioid receptor oligomerization.89 As to the mechanisms of the between-systems, N-methyl-d-aspartate (NMDA) receptors,1011 glutamate transporters (GTs),12 and glial activation with the release of proinflammatory cytokines stand out.1314

Tumor necrosis factor-α (TNF-α) was actively bound to TNF-α receptors, which are constitutively expressed on both neurons and glial cells in the central nervous system (CNS).15 The complexity of TNF-α responses is related to the receptor context, cell type, and functional status.16 Two different TNF-α receptors (p55/TNF-R1 and p75/TNF-R2) have been recognized17; they act in distinct pathways to mediate different cellular responses.18 Morphine tolerance shares similar neuronal plasticity mechanisms associated with hyperalgesia and allodynia in neuropathic pain, which are modulated by proinflammatory cytokines including TNF-α.11 Song and Zhao13 demonstrated that glial cells react to chronic morphine treatment by astroglial hypertrophy and increase glial fibrillary acidic protein expression in the spinal cord, posterior cingulated cortex, and hippocampus. Glutamate homeostasis is important in controlling the synaptic activity and further cellular dialogue between neurons, astrocytes, and microglia; the involvement of TNF-α was observed, via NF-κB activation, in the inhibition of GT expression and potentiation of glutamate neurotoxicity in organotypic brain slice culture.19 TNF-α enhances synaptic strength by increasing surface expression of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors, which is prevented by the blockade of the TNF-α signaling pathway in cultured hippocampal neurons.20 These findings prove that TNF-α plays a significant role in the modulation of glutamatergic transmission. This review addresses the role of the proinflammatory cytokine TNF-α in the development of morphine tolerance and the interactions between neuroinflammation and the glutamatergic receptor system.

2. TNF-α and neuroinflammation in the development of morphine tolerance

2.1. Role of neuroinflammation in morphine tolerance

Clinical evidence has showed that heroin addicts are prone to infection.21 It had been demonstrated that morphine compromises immune function by promoting macrophage apoptosis in opiate addicts22 and inducing Fas expression in lymphocytes.23 This immune compromise by morphine administration was postulated to result from a direct interaction between opioid receptors and immune cells, or indirectly through the activation of opioid receptors in the CNS, which modulate the hypothalamic pituitary adrenal axis and the autonomic function.24 Drug abusers, including those of cocaine and morphine, have been shown to have significant T-cell-dependent antibody responses with increasing IL-4 and shifting of the T-cell function to Th2-type cells via an opioid receptor-mediated action.2526 An acceleration of morphine tolerance development in animals with neuropathic pain suggests similar cellular mechanisms in tolerance and pain hypersensitivity.1127 A previous study demonstrated that glia-derived proinflammatory cytokines inhibit the antinociceptive effect of morphine by sensitizing pain-transmission neurons in animals with morphine tolerance and neuropathic pain.28 Both central and peripheral administration of proinflammatory cytokines TNF-α, IL-1β, and IL-6 facilitate pain transmission,2930 and this reduction of the antinociceptive effect of morphine can be reversed by inhibition of glial metabolism, antagonism of IL-1 receptors, and induction of anti-inflammatory cytokine IL-10 expression.1331 In our previous study, chronic morphine infusion induced a significant increase in TNF-α, IL-1β, and IL-6 mRNA expressions in the spinal cord dorsal horn of tolerant rats; inhibition of the proinflammatory cytokine expression attenuated the morphine tolerance; and administration of the TNF-α inhibitor etanercept reduced proinflammatory cytokines production and microglial activation, thus preserving the antinociceptive effect of morphine.32 Taken together, blockade of TNF-α signaling seems to be an effective biological target in the management of morphine tolerance.

2.2. Glial activation and morphine tolerance

Microglia have been shown to play a main role in cytokine release upon activation in the CNS.33 Compelling evidence has suggested that non-neuronal cells, instead of neurons, are crucial for the development of morphine tolerance.31 Repeated morphine administration leads to activation of calcitonin gene-related peptide (CGRP) receptors, which differentially control the synthesis and release of proinflammatory cytokines TNF-α, IL-1β, and IL-6 from microglia and astrocytes as well by extracellular signal-regulated kinase and p38MAPK.34 The blockade of CGRP receptor signaling in microglia and astrocytes prevents proinflammatory cytokine release and subsequent tolerance development. Based on previous studies, pharmacological modulation of spinal glial activity using amitriptyline and etanercept attenuated morphine tolerance.3235 Investigation of other potent anti-inflammatory drugs for morphine tolerance management is thus warranted to further explicate the features of morphine tolerance.

2.3. Effect of the TNF-α inhibitor etanercept in morphine tolerance

The first clinical use of TNF inhibitors for the treatment of rheumatoid arthritis was in 1998.36 Etanercept is a recombinant soluble p75 receptor (p75), which is linked to the Fc portion of human IgG. Nonsoluble but membraneous TNF-α receptors can elicit biological activity upon TNF-α binding. Etanercept binds two circulating TNF-α, preventing it from interacting with its membrane receptors, and restrains the following signaling.37 Application of etanercept in the treatment of other inflammatory disorders such as ankylosing spondilytis,38 Crohn disease,39 and Alzheimer disease40 has expanded in recent years. Perispinal administration of etanercept attenuated TNF-α effect on synaptic dysfunction, which rapidly improved cognitive function for Alzheimer patients.40 In our study, intrathecal pretreatment with etanercept 50 μg suppressed effectively microglia activation and mRNA expression of proinflammatory cytokines TNF-α, IL-1β, and IL-6 in the spinal cords of morphine-tolerant rats.32 It restored the antinociceptive effect of morphine with a maximum of 60% recovery in morphine-tolerant rats.

3. Glutamatergic receptor system and morphine tolerance

3.1. Excitatory amino acids and GTs

The acidic amino acid glutamate is a major excitatory neurotransmitter in the mammalian CNS including spinal cord. Excess of glutamate in the synaptic cleft may result in massive Ca2+ influx, causing neuronal plasticity and even excitotoxicity.4142 Hence, maintenance of glutamate homeostasis is crucial for highly efficient synaptic communication and prevention of neurotoxicity.43 In physiological conditions, glutamate is rapidly removed from the synaptic cleft by diffusion and reuptake.4445 The latter is accomplished by a family of high-affinity Na+-dependent GTs localized in the cytoplasmic membrane of astrocytes and neurons.46 Five GTs have been cloned and characterized. EAAC1 (excitatory amino acid carrier/transporter; EAAT3) and EAAT4 are predominantly localized in neurons, and EAAT5 is enriched in retinal tissue, whereas glutamate–aspartate transporter (GLAST; EAAT1) and GLT-1 (EAAT2) are generally expressed in astrocytes.47

3.2. Morphine tolerance and NMDA receptors

Glutamatergic receptors, in particular NMDA receptors, are critically involved in chronic opioid-induced neuronal adaptations, such as opioid tolerance, dependence, and withdrawal,104849 and in chronic pain-associated hyperalgesia.505152 Emerging evidence suggests that opioid tolerance and abnormal pain sensitivity, the opioid-induced hyperalgesia, may share common cellular mechanisms and mediate, at least in part, through NMDA receptors1253 and GT system.54 Our previous studies have demonstrated that coinfusion of the NMDA antagonist MK-801 with morphine shifted the AD50 (analgesic dose) of morphine to 11.2 μg compared to 83.8 μg in morphine tolerant-rats.55 In addition, dexamethasone retained the antinociceptive effect of morphine in chronic morphine-infused rats via inhibition of downregulation of GTs and the proceeding increased of EAAs.56 We therefore assume that GT regulation plays an important role in morphine tolerance, by inhibiting the accumulation of excitatory amino acid in the synaptic cleft and influencing the neurons, microglia, and astrocyte activity of surrounding cells.

3.3. TNF-α regulates glutamatergic receptor activity

As known, NF-κB and AP-1 are important transcriptional factors related to the TNF-α signaling cascade.57 Central glucocorticoid receptors modulate downregulation of spinal EAAC1 after peripheral nerve injury through diminished NF-κB expression.58 Intriguingly, we found that amitriptyline upregulates GT expressions, including GLAST, GLT-1, and EAAC1, through NF-κB-dependent activation.59 At present, the knowledge of EAAC1 regulation by NF-κB pathway is still limited and controversial. Chronic morphine exposure induced E3 ligase activation via cAMP/PKA signaling and resulted in a ubiquitin–proteasome system that mediated degradation of EAAC1.60 This means that more mechanisms are involved in the EAAC1 regulation after chronic morphine exposure; further studies are required to elucidate these underlying signaling mechanisms. Moreover, Sitcheran et al61 have demonstrated that TNF-α induces I kappa B degradation, which triggers NF-κB nuclear translocation and suppresses GLT-1 expression in H4 astroglioma cells. Nevertheless, differential effect of TNF-α on GLT-1 expression is suggested; it evokes GLT-1 expression in spinal microglia of rats by lipopolysaccharide treatment62 and also inhibits glutamate uptake by primary human fetal astrocyte.46 The reason for this apparent discrepancy is uncertain. Similarly, Korn et al demonstrated that increases in TNF-α might reduce GLAST protein expression, in a dose-dependent manner, which was prevented by preincubation with a neutralizing TNF-α antibody. It was assumed that downregulation of GLAST resulted in an excess of extracellular glutamate and triggered excitotoxicity through NMDA receptors in neurons and AMPA/kainite receptors in oligodendrocytes, and they concluded that TNF-α played an important role in CNS inflammatory disorders via reduction of glutamate uptake.63

In addition, TNF-α was demonstrated to enhance synaptic strength by increasing the surface expression of AMPA receptors in cultured hippocampal neurons; it was prevented by the blockade of the TNF-α signaling pathway.20 Stellwagen et al64 further demonstrated that TNF-α preferentially increased the synaptic expression of GluR2-lacking AMPA receptors that are more permeable to Ca2+ and simultaneously decreased the surface expression of GABAA receptors, which resulted in a reduction of the inhibitory synaptic transmission. It seems that the net effect of TNF-α is to alter the balance of excitation and inhibition. In contrast, Beattie et al20 found that TNF-α enhanced synaptic strength by increasing the expression of AMPA receptor, but not the NMDA receptor; they suggested that mobility of NMDA receptors was less than that of AMPA receptors due to differential attachment.65 Induction of GluR1 expression of AMPA receptors by TNF-α increases the neuron vulnerability to excitotoxicity, which is mediated through acid sphingomyelinase and NF-κB pathway.66 As already known, AMPA GluR2 expression affects calcium permeability through AMPA receptors and GluR2 subunit-lacking AMPA receptors will alter its synaptic function.67 NMDA receptors are highly permeable to Ca2+, and the NR1 subunit of the NMDA receptor is an essential functional unit of the NMDA receptor.68 As already mentioned, NMDA receptors are involved in the mechanism of morphine tolerance and NMDA receptor antagonists were demonstrated to inhibit morphine tolerance.101269 Lim et al70 observed an increase in the expression of the NMDA receptor NR1 subunit in the spinal cord dorsal horns of chronic morphine-infused rats, which was mediated by glucocorticoid receptor activation in a time-dependent manner. Moreover, Shimoyama et al71 also found that morphine tolerance was attenuated by the deletion of NMDA receptor NR1 subunit via intrathecal administration of antisense oligonucleotide. We therefore suggested that the increase in glutamate availability by TNF-α was due to the downregulation of membrane GTs, in accordance with the upregulation of surface AMPA and NMDA receptors in chronic morphine-infused rats, which contributed to the excessive activation of glutamatergic receptors and then to the reduction in the antinociceptive effect of morphine.72 Conceivably, activation of glutamatergic receptor system will enhance the intracellular mechanisms of morphine tolerance, such as interaction with μ-opioid receptors through Ca2+ influx, which activates PKC, NO synthase, and relevant gene regulation.11

4. Conclusion

In summary, the effect of neuroinflammation in the development of morphine tolerance and the possible role of etanercept in the attenuation of morphine tolerance are shown in Fig. 1. Long-term morphine administration induces microglial activation and alters glutamatergic transmission in the spinal cord. Morphine enhances the expression of proinflammatory cytokines TNF-α, IL-1β, and IL-6 in spinal cord microglia (see ① in Fig. 1) and upregulates expressions of AMPA- and NMDA-receptor subunits on the postsynaptic neuronal plasma membrane (② in Fig. 1). It therefore inhibits the membraneous GT expression in the spinal astrocytes or postsynaptic neurons, including GLAST, GLT-1, and EACC1 (③ in Fig. 1), thus facilitating the TNF-α-related signaling. According to the results of our studies and others, we suggest that the TNF-α inhibitor etanercept may restore the membraneous GTs on the spinal astrocytes in particular (④ in Fig. 1) and reduce the accumulation of synaptic excitatory amino acids, subsequently inhibiting the upregulation of AMPA- and NMDA-receptor subunits on the postsynaptic neuron plasma membrane (⑤ in Fig. 1). Recent works have focused on research on morphine tolerance and the interaction between inflammation and glutamatergic receptor system function; studies have demonstrated that repeated morphine administration leads to an increase in the TNF-α expression in spinal cord microglia.32 Watkins et al73 have suggested that chronic administration of opioids activates glia as facilitators of pain. The activated glia release neuroexcitatory substances including proinflammatory cytokines, and modulation of glial activity can be the target of treating morphine tolerance. The therapeutic potential of blockade of TNF-α activity is widely used in a variety of disorders of excessive inflammation and has been approved by the US Food and Drug Administration for clinical use. Modulation of glutamatergic transmission by TNF-α suggests that inhibition of TNF-α signaling provides another potential therapy of clinical pain management, particularly in patients who need long-term morphine administration or suffer from neuropathic pain. Etanercept reduced mechanical allodynia and attenuated membraneous expression of Ca2+-permeable AMPA receptors by reducing GluR1 membraneous insertion.74 However, more information is needed about the intracellular TNF-α signaling pathway and the subtypes of TNF-α receptors that are involved. This review emphasizes the clinical role of a selective TNF-α inhibitor in pain management, in particular, in the attenuation of morphine tolerance.

Fig. 1
Download full-size image
Fig. 1. Effect of neuroinflammation on and the possible role of etanercept in the development of morphine tolerance. EAAC = excitatory amino acid carrier; EAAT = excitatory amino acid transporter; IL = interleukin; GLAST = glutamate–aspartate transporter; NMDA = N-methyl-d-aspartate; TNF = tumor necrosis factor.

References

1
G.F. Koob, F.E. Bloom
Cellular and molecular mechanisms of drug dependence
Science, 242 (1988), pp. 715-723
2
G.D. Dalton, F.L. Smith, P.A. Smith, W.L. Dewey
Alterations in brain protein kinase A activity and reversal of morphine tolerance by two fragments of native protein kinase A inhibitor peptide (PKI)
Neuropharmacology, 48 (2005), pp. 648-657
3
Z. Wang, W. Sadee
Tolerance to morphine at the mu-opioid receptor differentially induced by cAMP-dependent protein kinase activation and morphine
Eur J Pharmacol, 389 (2000), pp. 165-171
4
S. Chakrabarti, M. Oppermann, A.R. Gintzler
Chronic morphine induces the concomitant phosphorylation and altered association of multiple signaling proteins: a novel mechanism for modulating cell signaling
Proc Natl Acad Sci U S A, 98 (2001), pp. 4209-4214
5
L.E. Otterbein, F.H. Bach, J. Alam, M. Soares, H. Tao Lu, M. Wysk, et al.
Carbon monoxide has anti-inflammatory effects involving the mitogen-activated protein kinase pathway
Nat Med, 6 (4) (Apr 2000), pp. 422-428
6
L.M. Bohn, R.J. Lefkowitz, R.R. Gainetdinov, K. Peppel, M.G. Caron, F.T. Lin
Enhanced morphine analgesia in mice lacking beta-arrestin 2
Science, 286 (1999), pp. 2495-2498
7
L.M. Bohn, R.R. Gainetdinov, F.T. Lin, R.J. Lefkowitz, M.G. Caron
Mu-opioid receptor desensitization by beta-arrestin-2 determines morphine tolerance but not dependence
Nature, 408 (2000), pp. 720-723
8
L. He, J. Fong, M. von Zastrow, J.L. Whistler
Regulation of opioid receptor trafficking and morphine tolerance by receptor oligomerization
Cell, 108 (2002), pp. 271-282
9
B.A. Jordan, L.A. Devi
G-protein-coupled receptor heterodimerization modulates receptor function
Nature, 399 (1999), pp. 697-700
10
K.A. Trujillo, H. Akil
Inhibition of morphine tolerance and dependence by the NMDA receptor antagonist MK-801
Science, 251 (1991), pp. 85-87
Article  
11
J. Mao, D.D. Price, D.J. Mayer
Mechanisms of hyperalgesia and morphine tolerance: a current view of their possible interactions
Pain, 62 (1995), pp. 259-274
12
J. Mao, B. Sung, R.R. Ji, G. Lim
Chronic morphine induces downregulation of spinal glutamate transporters: implications in morphine tolerance and abnormal pain sensitivity
J Neurosci, 22 (2002), pp. 8312-8323
13
P. Song, Z.Q. Zhao
The involvement of glial cells in the development of morphine tolerance
Neurosci Res, 39 (2001), pp. 281-286
14
V. Raghavendra, M.D. Rutkowski, J.A. DeLeo
The role of spinal neuroimmune activation in morphine tolerance/hyperalgesia in neuropathic and sham-operated rats
J Neurosci, 22 (2002), pp. 9980-9989
15
E.N. Benveniste, D.J. Benos
TNF-alpha- and IFN-gamma-mediated signal transduction pathways: effects on glial cell gene expression and function
FASEB J, 9 (1995), pp. 1577-1584
16
F.X. Pimentel-Muinos, B. Seed
Regulated commitment of TNF receptor signaling: a molecular switch for death or activation
Immunity, 11 (1999), pp. 783-793
17
B. Beutler, C. van Huffel
Unraveling function in the TNF ligand and receptor families
Science, 264 (1994), pp. 667-668
18
L.A. Tartaglia, R.F. Weber, I.S. Figari, C. Reynolds, M.A. Palladino Jr., D.V. Goeddel
The two different receptors for tumor necrosis factor mediate distinct cellular responses
Proc Natl Acad Sci U S A, 88 (1991), pp. 9292-9296
19
J.Y. Zou, F.T. Crews
TNF alpha potentiates glutamate neurotoxicity by inhibiting glutamate uptake in organotypic brain slice cultures: neuroprotection by NF kappa B inhibition
Brain Res, 1034 (2005), pp. 11-24
20
E.C. Beattie, D. Stellwagen, W. Morishita, J.C. Bresnahan, B.K. Ha, M. Von Zastrow, et al.
Control of synaptic strength by glial TNFalpha
Science, 295 (2002), pp. 2282-2285
21
R.A. Olsson, M.J. Romansky
Staphylococcal tricuspid endocarditis in heroin addicts
Ann Intern Med, 57 (1962), pp. 755-762
22
P.C. Singhal, P. Sharma, A.A. Kapasi, K. Reddy, N. Franki, N. Gibbons
Morphine enhances macrophage apoptosis
J Immunol, 160 (1998), pp. 1886-1893
23
D. Yin, R.A. Mufson, R. Wang, Y. Shi
Fas-mediated cell death promoted by opioids
Nature, 397 (1999), p. 218
24
A.L. Siren, R. McCarron, L. Wang, P. Garcia-Pinto, C. Ruetzler, D. Martin, et al.
Proinflammatory cytokine expression contributes to brain injury provoked by chronic monocyte activation
Mol Med, 7 (2001), pp. 219-229
25
E.D. Stanulis, S.D. Jordan, J.A. Rosecrans, M.P. Holsapple
Disruption of Th1/Th2 cytokine balance by cocaine is mediated by corticosterone
Immunopharmacology, 37 (1997), pp. 25-33
26
F. Tamion, V. Richard, G. Bonmarchand, J. Leroy, J.P. Lebreton, C. Thuillez
Induction of heme-oxygenase-1 prevents the systemic responses to hemorrhagic shock
Am J Respir Crit Care Med, 164 (10 Pt 1) (2001), pp. 1933-1938
27
D.J. Mayer, J. Mao, J. Holt, D.D. Price
Cellular mechanisms of neuropathic pain, morphine tolerance, and their interactions
Proc Natl Acad Sci U S A, 96 (1999), pp. 7731-7736
28
G.B. Stefano
Autoimmunovascular regulation: morphine and anandamide and ancondamide stimulated nitric oxide release
J Neuroimmunol, 83 (1998), pp. 70-76
29
A.J. Reeve, S. Patel, A. Fox, K. Walker, L. Urban
Intrathecally administered endotoxin or cytokines produce allodynia, hyperalgesia and changes in spinal cord neuronal responses to nociceptive stimuli in the rat
Eur J Pain, 4 (2000), pp. 247-257
30
J.A. DeLeo, R.W. Colburn, M. Nichols, A. Malhotra
Interleukin-6-mediated hyperalgesia/allodynia and increased spinal IL-6 expression in a rat mononeuropathy model
J Interferon Cytokine Res, 16 (1996), pp. 695-700
31
I.N. Johnston, E.D. Milligan, J. Wieseler-Frank, M.G. Frank, V. Zapata, J. Campisi, et al.
A role for proinflammatory cytokines and fractalkine in analgesia, tolerance, and subsequent pain facilitation induced by chronic intrathecal morphine
J Neurosci, 24 (2004), pp. 7353-7365
32
C.H. Shen, R.Y. Tsai, M.S. Shih, S.L. Lin, Y.H. Tai, C.C. Chien, et al.
Etanercept restores the antinociceptive effect of morphine and suppresses spinal neuroinflammation in morphine-tolerant rats
Anesth Analg, 112 (2011), pp. 454-459
33
F. Aloisi
Immune function of microglia
Glia, 36 (2001), pp. 165-179
34
Z. Wang, W. Ma, J.G. Chabot, R. Quirion
Cell-type specific activation of p38 and ERK mediates calcitonin gene-related peptide involvement in tolerance to morphine-induced analgesia
Faseb J, 23 (2009), pp. 2576-2586
35
Y.H. Tai, Y.H. Wang, J.J. Wang, P.L. Tao, C.S. Tung, C.S. Wong
Amitriptyline suppresses neuroinflammation and up-regulates glutamate transporters in morphine-tolerant rats
Pain, 124 (2006), pp. 77-86
36
R.N. Maini, F.C. Breedveld, J.R. Kalden, J.S. Smolen, D. Davis, J.D. Macfarlane, et al.
Therapeutic efficacy of multiple intravenous infusions of anti-tumor necrosis factor alpha monoclonal antibody combined with low-dose weekly methotrexate in rheumatoid arthritis
Arthritis Rheum, 41 (1998), pp. 1552-1563
37
D.L. Scott, G.H. Kingsley
Tumor necrosis factor inhibitors for rheumatoid arthritis
N Engl J Med, 355 (2006), pp. 704-712
38
H. Marzo-Ortega, D. McGonagle, P. O'Connor, P. Emery
Efficacy of etanercept in the treatment of the entheseal pathology in resistant spondylarthropathy: a clinical and magnetic resonance imaging study
Arthritis Rheum, 44 (2001), pp. 2112-2117
39
J.A. Wilson
Tumor necrosis factor alpha and colitis-associated colon cancer
N Engl J Med, 358 (2008), pp. 2733-2734
40
E.L. Tobinick, H. Gross
Rapid cognitive improvement in Alzheimer's disease following perispinal etanercept administration
J Neuroinflammation, 5 (2008), p. 2
Article   CrossRef  
41
M.F. Beal
Mechanisms of excitotoxicity in neurologic diseases
FASEB J, 6 (1992), pp. 3338-3344
42
S.A. Lipton, P.A. Rosenberg
Excitatory amino acids as a final common pathway for neurologic disorders
N Engl J Med, 330 (1994), pp. 613-622
Article  
43
G.J. Bennett
Update on the neurophysiology of pain transmission and modulation: focus on the NMDA-receptor
J Pain Symptom Manage, 19 (1 Suppl.) (2000), pp. S2-S6
Article  
44
S. Minucci, P.G. Pelicci
Histone deacetylase inhibitors and the promise of epigenetic (and more) treatments for cancer
Nat Rev Cancer, 6 (2006), pp. 38-51
45
S.H. Oliet, R. Piet, D.A. Poulain
Control of glutamate clearance and synaptic efficacy by glial coverage of neurons
Science, 292 (2001), pp. 923-926
46
S.M. Fine, R.A. Angel, S.W. Perry, L.G. Epstein, J.D. Rothstein, S. Dewhurst, et al.
Tumor necrosis factor alpha inhibits glutamate uptake by primary human astrocytes. Implications for pathogenesis of HIV-1 dementia
J Biol Chem, 271 (1996), pp. 15303-15306
47
G. Gegelashvili, A. Schousboe
High affinity glutamate transporters: regulation of expression and activity
Mol Pharmacol, 52 (1997), pp. 6-15
48
H.J. Kim, M. Rowe, M. Ren, J.S. Hong, P.S. Chen, D.M. Chuang
Histone deacetylase inhibitors exhibit anti-inflammatory and neuroprotective effects in a rat permanent ischemic model of stroke: multiple mechanisms of action
J Pharmacol Exp Ther, 321 (2007), pp. 892-901
Article   CrossRef  
49
K.A. Trujillo, H. Akil
Excitatory amino acids and drugs of abuse: a role for N-methyl-D-aspartate receptors in drug tolerance, sensitization and physical dependence
Drug Alcohol Depend, 38 (1995), pp. 139-154
50
T.J. Coderre, J. Katz, A.L. Vaccarino, R. Melzack
Contribution of central neuroplasticity to pathological pain: review of clinical and experimental evidence
Pain, 52 (1993), pp. 259-285
51
M.O. Urban, G.F. Gebhart
The glutamate synapse: a target in the pharmacological management of hyperalgesic pain states
Prog Brain Res, 116 (1998), pp. 407-420
52
F. Porreca, M.H. Ossipov, G.F. Gebhart
Chronic pain and medullary descending facilitation
Trends Neurosci, 25 (2002), pp. 319-325
53
B.H. Manning, J. Mao, H. Frenk, D.D. Price, D.J. Mayer
Continuous co-administration of dextromethorphan or MK-801 with morphine: attenuation of morphine dependence and naloxone-reversible attenuation of morphine tolerance
Pain, 67 (1996), pp. 79-88
54
G. Lim, S. Wang, J. Mao
cAMP and protein kinase A contribute to the downregulation of spinal glutamate transporters after chronic morphine
Neurosci Lett, 376 (2005), pp. 9-13
55
Z.H. Wen, Y.C. Chang, C.H. Cherng, J.J. Wang, P.L. Tao, C.S. Wong
Increasing of intrathecal CSF excitatory amino acids concentration following morphine challenge in morphine-tolerant rats
Brain Res, 995 (2004), pp. 253-259
56
H.Y. Lin, C.S. Chen, S.P. Lin, J.R. Weng, C.S. Chen
Targeting histone deacetylase in cancer therapy
Med Res Rev, 26 (2006), pp. 397-413
57
V. Baud, M. Karin
Signal transduction by tumor necrosis factor and its relatives
Trends Cell Biol, 11 (2001), pp. 372-377
58
S. Wang, G. Lim, L. Yang, B. Sung, J. Mao
Downregulation of spinal glutamate transporter EAAC1 following nerve injury is regulated by central glucocorticoid receptors in rats
Pain, 120 (2006), pp. 78-85
59
Y.H. Tai, R.Y. Tsai, Y.H. Wang, C.H. Cherng, P.L. Tao, T.M. Liu, et al.
Amitriptyline induces nuclear transcription factor-kappaB-dependent glutamate transporter upregulation in chronic morphine-infused rats
Neuroscience, 153 (2008), pp. 823-831
60
L. Yang, S. Wang, B. Sung, G. Lim, J. Mao
Morphine induces ubiquitin-proteasome activity and glutamate transporter degradation
J Biol Chem, 283 (2008), pp. 21703-21713
61
R. Sitcheran, P. Gupta, P.B. Fisher, A.S. Baldwin
Positive and negative regulation of EAAT2 by NF-kappaB: a role for N-myc in TNFalpha-controlled repression
EMBO J, 24 (2005), pp. 510-520
62
M. Persson, M. Brantefjord, E. Hansson, L. Ronnback
Lipopolysaccharide increases microglial GLT-1 expression and glutamate uptake capacity in vitro by a mechanism dependent on TNF-alpha
Glia, 51 (2005), pp. 111-120
Article   CrossRef  
63
T. Korn, T. Magnus, S. Jung
Autoantigen specific T cells inhibit glutamate uptake in astrocytes by decreasing expression of astrocytic glutamate transporter GLAST: a mechanism mediated by tumor necrosis factor-alpha
Faseb J, 19 (2005), pp. 1878-1880
64
D. Stellwagen, E.C. Beattie, J.Y. Seo, R.C. Malenka
Differential regulation of AMPA receptor and GABA receptor trafficking by tumor necrosis factor-alpha
J Neurosci, 25 (2005), pp. 3219-3228
65
D.W. Allison, V.I. Gelfand, I. Spector, A.M. Craig
Role of actin in anchoring postsynaptic receptors in cultured hippocampal neurons: differential attachment of NMDA versus AMPA receptors
J Neurosci, 18 (1998), pp. 2423-2436
66
Z. Yu, G. Cheng, X. Wen, G.D. Wu, W.T. Lee, D. Pleasure
Tumor necrosis factor alpha increases neuronal vulnerability to excitotoxic necrosis by inducing expression of the AMPA-glutamate receptor subunit GluR1 via an acid sphingomyelinase- and NF-kappaB-dependent mechanism
Neurobiol Dis, 11 (2002), pp. 199-213
67
M.S. Washburn, M. Numberger, S. Zhang, R. Dingledine
Differential dependence on GluR2 expression of three characteristic features of AMPA receptors
J Neurosci, 17 (1997), pp. 9393-9406
68
D. Forrest, M. Yuzaki, H.D. Soares, L. Ng, D.C. Luk, M. Sheng, et al.
Targeted disruption of NMDA receptor 1 gene abolishes NMDA response and results in neonatal death
Neuron, 13 (1994), pp. 325-338
69
N.J. Xu, L. Bao, H.P. Fan, G.B. Bao, L. Pu, Y.J. Lu, et al.
Morphine withdrawal increases glutamate uptake and surface expression of glutamate transporter GLT1 at hippocampal synapses
J Neurosci, 23 (2003), pp. 4775-4784
70
G. Lim, S. Wang, Q. Zeng, B. Sung, L. Yang, J. Mao
Expression of spinal NMDA receptor and PKCgamma after chronic morphine is regulated by spinal glucocorticoid receptor
J Neurosci, 25 (48) (Nov 30 2005), pp. 11145-11154
71
N. Shimoyama, M. Shimoyama, A.M. Davis, D.T. Monaghan, C.E. Inturrisi
An antisense oligonucleotide to the N-methyl-D-aspartate (NMDA) subunit NMDAR1 attenuates NMDA-induced nociception, hyperalgesia, and morphine tolerance
J Pharmacol Exp Ther, 312 (2005), pp. 834-840
72
C.H. Shen, R.Y. Tsai, Y.H. Tai, S.L. Lin, C.C. Chien, C.S. Wong
Intrathecal etanercept partially restores morphine's antinociception in morphine-tolerant rats via attenuation of the glutamatergic transmission
Anesth Analg, 113 (2011), pp. 184-190
73
L.R. Watkins, M.R. Hutchinson, I.N. Johnston, S.F. Maier
Glia: novel counter-regulators of opioid analgesia
Trends Neurosci, 28 (2005), pp. 661-669
74
J.I. Choi, C.I. Svensson, F.J. Koehrn, A. Bhuskute, L.S. Sorkin
Peripheral inflammation induces tumor necrosis factor dependent AMPA receptor trafficking and Akt phosphorylation in spinal cord in addition to pain behavior
Pain, 149 (2010), pp. 243-253

References

Close