Você está na página 1de 9

Inflammation & Cell Signaling 2014;1: e111.

http://www.smartscitech.com/index.php/ics

REVIEW

Novel Therapeutic Targets in Neuroinflammation and


Neuropathic Pain
Geeta Ramesh

Division of Bacteriology and Parasitology, Tulane National Primate Research Center, Tulane University, 18703 Three Rivers
Road, Covington, LA, USA

Correspondence: Geeta Ramesh


E-mail: gramesh@tulane.edu
Received: March 06, 2014
Published online: June 16, 2014

There is abounding evidence that neuroinflammation plays a major role in the pathogenesis of neurodegeneration
and neuropathic pain. Chemokine-induced recruitment of peripheral immune cells is a central feature in inflammatory
neurodegenerative disorders. Immune cells, glial cells and neurons constitute an integral network that coordinates the
immune response by releasing inflammatory mediators that in turn modulate inflammation, neurodegeneration and
the signal transduction of pain, via interaction with neurotransmitters and their receptors. The chemokine monocyte
chemoattractant protein-1/ chemokine (C-C motif) ligand (MCP-1/CCL2) and its receptor C-C chemokine receptor
(CCR2) play a major role in mediating neuroinflammation and targeting CCL2/CCR2 represents a promising strategy
to limit neuroinflammation-induced neuropathy. In addition, the CCL2/CCR2 axis is also involved in mediating the
pain response. Key cellular signaling events such as phosphorylation and subsequent activation of mitogen activated
protein kinase (MAPK) p38 and its substrate MAPK-activated protein MAPKAP Kinase (MK) MK-2, regulate
neuroinflammation, neuronal survival and synaptic activity. Further, MAPKs such as extracellular signal-regulated
kinases (ERK), c-jun N-terminal kinase (JNK) and p38 play vital roles in mediating the pain signaling cascade and
contribute to the maintenance of peripheral and central neuronal sensitization associated with chronic pain. This
review outlines the rationale for developing therapeutic strategies against CCL2/CCR2 and MAPK signaling networks,
identifying them as novel therapeutic targets for limiting neuroinflammation and neuropathic pain.
Keywords: neuroinflammation; pain signaling; MCP-1/CCL2; CCR2; MAPK p38, MK-2; MAPK activation

Inflammation & Cell Signaling 2014; 1: e111. doi: 10.14800/ics.111; 2014 by Geeta Ramesh.

Monocyte chemoattractant protein-1 (MCP-1/CCL2) crucial for normal neurological functioning. Chemokines
and its receptor CCR2 in neuroinflammation and may induce neuronal death directly through the activation
neuropathic pain of neuronal chemokine receptors or indirectly through the
activation of microglial killing mechanisms. Evidence of
Chemokines are implicated in many diseases of the
major roles for chemokines and their receptors in
nervous system. Evidence is emerging that chemokines
diseases of the brain is accumulating and this system is a
play a role in the physiology of the nervous system,
potential target for treatment of neurodegenerative
including neuronal migration, cell proliferation and
diseases [1].
synaptic activity, besides mediating neuroinflammation
through the recruitment of leukocytes by their chemo- The chemokine MCP-1/CCL2 that attracts monocytes
tactic activity. Chemokines and their receptors are among and T cells [2] plays a major role in the nervous system.
the key players responsible for communication between CCL2 has been suggested to trigger firm adhesion of
neurons and inflammatory cells, and this cross talk is leukocytes to activated endothelial cells by upregulating

1
Geeta Ramesh
Neuroinflammation and Pain signaling

integrins on monocytes, as well as influence their regulation of brain inflammation after peripheral
subsequent diapedesis [3]. Besides modulating the ex- endotoxemia [23].
pression of endothelial adhesion molecules, leukocyte
CCL2 also participates in pain regulation by directly
integrins and cytokine production during central nervous
interacting with sensory neurons and indirectly via
system (CNS) inflammation, CCL2 also regulates the
peripheral leukocyte activation in the PNS [24, 25]. More-
permeability of the blood-brain barrier by opening or
over, neuropathic pain induced by nerve injury is not
altering tight junctions [4]. It is one of the main effectors
elicited in CCR2 gene-deficient mice [24]. It is significant
driving post ischemic infiltration of monocytes into the
to note that CCL2 has been seen to be elevated in primary
brain parenchyma [5]. Expression of CCL2 by reactive
sensory neurons after nerve injury, and CCR2 expression
astrocytes has been documented in demyelinating
has been observed in both dorsal root ganglia (DRG)
multiple sclerosis lesions (MS) [6]. CCL2 expression by
sensory neurons and Schwann cells, in injured peripheral
glial cells is involved in directing leukocytes to sites of
nerves [26]. The addition of CCL2 to cultured DRG
axonal injury in the CNS [7]. Brain micro vessels from
neurons triggered the release of calcitonin gene-related
Alzheimers disease (AD) patients show elevated levels
peptide (CGRP), a nociceptor neurotransmitter, from
of CCL2 [8]. The absence of CCL2 protein in mice leads
these cells presumably as a result of increased neuronal
to decreased local macrophage recruitment in ex-
excitation [26]. The role of CGRP in different models of
perimental autoimmune encephalitis (EAE) [9]. CCL2
pain has been well documented [27].
plays a major role in the recruitment of leukocytes into
the subarachnoid space in various types of infectious The upregulation of CCL2 in the sensory neurons,
meningitis [10]. satellite glial cells and Schwann cells of DRG from
rhesus monkeys when incubated with live B. burgdorferi
The chemokine CCL2 is an important mediator in
spirochetes that cause Lyme neuroborreliosis (in which
many neuroinflammatory and neurodegenerative brain
patients present with excruciating pain along the spine
diseases characterized by neuronal degeneration [11, 12, 13].
and limbs due to radiculitis or inflammation in spinal
CCL2 has been found to be upregulated in actively
nerve roots) has been recently reported [28]. Besides
demyelinating MS plaques [14], and its expression is
mediating inflammation in the nerve roots, CCL2
increased in EAE [15]. CCL2 modulates microglial
induced in the cells of the DRG may also be involved in
activation and proliferation, thereby contributing to the
the signaling of the pain response in Lyme neuro-
inflammatory response mounted in the CNS [16]. CCL2 is
borreliosis. A recent study indicates that activation of
known to play a role in mediating nerve damage and
paracrine CCL2/CCR2 signaling between DRG neurons
demyelination of axons by causing an influx of mono-
plays a critical role in the development of peripheral
cytes and T cells in Wallerian degeneration [17]. CCL2
neuropathy associated with Taxol treatment for cancer.
levels are elevated in the cerebrospinal fluid (CSF) of
Blocking CCL2/CCR2 signaling by anti-CCL2 antibody
patients with Lyme neuroborreliosis [18] a disease that
resulted in the prevention of Taxol-induced peripheral
shows similar clinical signs as that shown by MS [19].
neuropathy including mechanical hypersensitivity and
High levels of CCL2 have been found in the CSF and
loss of intra epidermal nerve fibers, suggesting that
localized in the microglia of the spinal cord of rhesus
targeting CCL2/CCR2 signaling could be a novel
monkeys infected intrathecally with Borrelia burgdorferi,
therapeutic approach [29]. CCL2 is derived from several
the spirochete that causes Lyme neuroborreliosis [20], and
types of cells in the peripheral and central nervous
CCL2 may also possibly contribute to the axonal damage
systems following nerve injury, and is largely involved in
that affects patients with Lyme neuroborreliosis of the
the pathogenesis of neuropathic pain [30]. Further studies
peripheral nervous system (PNS) [21].
focussing on the functions of the chemokine-cytokine
The induction of neuronal CCL2 during mild network-mediated regulation of neuroinflammation may
impairment of oxidative metabolism caused by microglial lead to novel therapeutic strate-gies against neuropathic
recruitment/activation exacerbated neurodegeneration in pain.
Thiamine deficiency (TD)-induced neuronal death.
Conversely CCL2-knockout (KO) mice were resistant to
TD-induced neuronal death, suggesting that the The role of MAPK p38 in neuroinflammation,
chemokine CCL2 mediates microglial recruitment and apoptosis and glutamate mediated excitotoxicity
neurodegeneration in this model [22]. Interestingly, CCL2-
The MAPKs are a specific class of serine/threonine
deficient mice showed reduced neuroinflammatory
kinases that respond to extracellular signals such as
responses and increased peripheral inflammatory
growth factors, mitogens and cellular stress and mediate
responses to peripheral endotoxin insult. Collectively,
proliferation, differentiation and cell survival in
these data demonstrate a significant role for CCL2 in

2
Geeta Ramesh
Neuroinflammation and Pain signaling

mammalian cells. There are distinct groups of MAPKs damaging rather than a protective effect [45]. Prolonged
within mammalian cells including p38 MAPK, ERKs and and sustained activation of glial cells can result in an
JNKs [31]. exaggerated inflammatory response that causes neuronal
cell death through the elevated release of proin-
In the CNS, activation of the p38 MAPK pathway
flammatory cytokines, which have a potential neurotoxic
constitutes a key step in the development of neuroin-
effect leading to neurodegeneration [46, 47]. A role for p38
flammation. Inflammatory stimuli bind to receptors of the
MAPK activation in mediating astrogliosis has also been
cell surface triggering intracellular signal transduction
documented in Lyme neuroborreliosis [48, 49].
pathways such as the nuclear factor (NFkB) pathway and
the MAPK pathways [32, 33]. Intracellular p38 MAP kinase
gets activated and profoundly modulates somatic
The role of p38 MAPK and its substrate MAPK
inflammatory responses. MAPK p38 signaling controls
activated protein MAPKAP Kinase (MK) MK-2 in
the expression of adhesion molecules, cytokines and
neuroinflammation
chemokines, and a variety of other factors that mediate
and control the inflammatory process. Many in- Several p38 MAPK targets are kinases and
flammatory response proteins such as tumor necrosis transcription factors, known to play a role in
factor (TNF-, interleukin (IL-1, interleukin (IL-6), inflammation via the production and activation of
interleukin (IL-8) and CCL2 depend on p38 signaling for inflammatory mediators. It is known that p38 MAPK
their production [34, 35]. The chemokine CCL2 activates becomes activated by many cellular stresses besides
the p38 MAPK pathway in cultured rat hippocampal cells inflammatory cytokines [50, 51]. The identification of
[36]
. IL-1 induction of neuron apoptosis depends on p38
[52]
MAPK activity after spinal cord injury [37]. residues and the discovery of MK2 as being a direct
Apoptosis or cell death following cytokine-mediated
of the possible molecular mechanisms that could be
glutamate-induced excitotoxicity in the brain is also
involved in the activation of p38 MAPK cascades [53, 54].
mediated by p38 MAPK [38]. Pro-inflammatory cytokine
MAPKAP Kinases (MKs), MAPKAPK-2 (MK-2) and
expression and p38 MAPK signaling have been
MAPKAPK-3 (MK-3) are serine/threonine kinases are
implicated in glutamate-induced neuronal death of
part of the subfamily that bind to and are specifically
neonatal rats, and their inhibition may have an important
activated by the p38 MAPK isoforms [55, 56]. MK-2 is
neuroprotective role as part of an anti-inflammatory
recognized as the most important kinase to be activated
therapeutic strategy [38]. Excitotoxic neuronal death
by p38 MAPK since it has a vital role in mediating both
occurs through the activation of N-methyl-D-aspartate
cellular stress and inflammatory responses. The p38
(NMDA) and non-NMDA glutamatergic receptors in the
MAPK/MK-2 complex has been documented to
CNS [39]. Glutamate also induces strong activation of p38
contribute to inflammation in vivo, since MK-2 knockout
and indeed, cell death can be prevented by inhibitors of
mice are resistant to endotoxic shock as a result of
the p38 MAPK pathway. Furthermore, intracellular
lipopolysaccharide (LPS)-stimulation [57]. MK-2 is in-
signals generated by-amino-3-hydroxy-5-methyl-4-
volved in regulating the production of TNF-, IL-6, IL-8,
isoxazolepropionic acid (AMPA) receptors activate the
and several other cytokines that play a role in
stress sensitive MAP kinases implicated in apoptotic
inflammation [58, 59]. MK-2 expression and activation is
neuronal death such as JNK and p38. NMDA and AMPA
increased in microglia that have been stimulated with
receptor expression and cortical neuronal death are
LPS- and interferon-, and microglial cells cultured from
associated with p38 MAPK in glutamate-induced
MK-2 knockout mice showed a reduction in the levels of
excitotoxicity in vivo [39]. Neuronal cell death due to
inflammatory cytokines [60]. This signaling is of particular
glutamate excitotoxicity is mediated by p38 MAPK
interest as the p38 MAPK/MK-2 pathway and the
activation in the rat cerebral cortex [40]. Inhibition of
consequent production of inflammatory cytokines have a
delayed induction of p38 MAPK attenuates kainic
significant role in neurodegenerative disease processes
acid-induced neuronal loss in the hippocampus [41, 42, 43].
where oxidative stress and persistent neuroin- flammation
In addition, MAPK p38 participates in the neuronal MAP
are the primary causes of disease. MAPKAP kinase-2,
kinase cascade of signaling mechanisms that integrate
one of p38 MAPKs more prevalent substrates has also
synaptic plasticity and memory [44].
been implicated in Parkisons disease (PD), where it has
Irregularities in p38 MAPK signaling in neuronal cells been shown that MK2-deficient mice show decreased
have been associated with diseases linked with levels of neuroinflammation and loss of dopaminergic
neuroinflammatory processes where persistent inflamm- neurons within the substantia nigra after treatment with
atory stimuli such as chronic microglial activation have a the Parkinsons inducing neurotoxin 1-methy-4-phenyl-1,

3
Geeta Ramesh
Neuroinflammation and Pain signaling

2, 3, 6-tetrahydropyridine (MPTP) compared to MK-2 Experimental pain models have shown sustained
wild-type mice [61]. MAPK activation in nociceptive primary sensory neurons
present in the DRG, second order neurons located in the
MAPK p38 and its substrates have been shown to
dorsal horn of the spinal cord, neurons located in the
modulate neuronal plasticity and have been implicated in
brainstem and thalamic-cortical regions that are important
several neurodegenerative diseases. The release of
for nociceptive processing and interpretation of pain
pro-inflammatory cytokines as a result of activation of
related signaling information [72, 73]. Importantly, inhibi-
the p38 MAPK cascade, has been reported to be involved
tion of MAPK phosphorylation, both of ERKs and p38,
in AD, PD, MS, cerebral ischemia, depression and
resulted in the production of antinociceptive activity in
neuropathic pain [62]. The activation of p38 MAPK
experimental pain models [69]. MAPK activation is con-
signaling mediates changes in morphology and density of
sidered to be an essential component of nociceptive
dendritic spines seen during the development of
peripheral and central sensitization and proposed to
neurodegenerative disease and is associated with memory
function as a master switch in the initiation and
impairment after epileptic seizures [62]. The understanding
maintenance of pathological pain. Increased MAPK
of the functional role of the p38 MAPK signaling cascade
phosphorylation (activation) has been observed in
in affecting synaptic plasticity in the hippocampus and its
neurons and glia in peripheral nerve fibers, DRG cells,
potential role in neurodegenerative diseases such as AD
and the spinal cord dorsal horn and supraspinal areas
has made significant progress. Several formulations of
relevant to pain-processing [74]. Studies have indicated
p38 MAPK inhibitors are being actively screened in
that topical application of capsaicin to the hind paw
phase II clinical trials for depression. Evaluation of
results in the increased phospho-p38 (activated) immune-
MAPK p38 inhibitors as therapeutic agents in neural
reactivity in the nerve fibers of the dermis [75]. Further,
diseases is an active area of research and they are being
intraplantar injection of capsaicin induced phosp-
rigorously explored in both preclinical experimental
horylated ERK in intra-epidermal nerve fibers and nerve
models in addition to clinical trials for various
bundles, which was subsequently prevented when
inflammatory diseases [63].
pretreated with MAPK kinase enzyme (MEK) inhibitor
[76]
. Surgical models of neuropathic pain revealed
differential activation of p38, ERK and JNK in neurons
The role of Mitogen activated Protein Kinase (MAPK)
and glia of DRG that were in turn attenuated by using
signaling in the transduction of the pain response
inhibitors that block MAPK phosphorylation [72, 77, 78, 79,
80, 81, 82]
Pain is a feeling of distress caused by activation of .
specific nerve endings in response to various stimuli
MAPK p38 has been the subject of extensive efforts in
capable of causing tissue injury. This specialized sensory
basic research and drug discovery [83]. Inhibition of p38
processing, also defined as nociception leads to a reflex
MAPK and ERK differentially modulates cytokine
or withdrawal response that helps to reduce or prevent
expression [41]. MAPK p38 is a key signaling molecule
tissue injury [64]. However, over time, nociceptive
and a therapeutic target for inflammatory diseases such as
sensitization may take on a detrimental role where
arthritis [32]. A specific inhibitor of p38 MAPK,
increased sensitivity can lead to prolonged activation of
SB203580 (Calbiochem) was effective in affording brain
nociceptors, and the subsequent recruitment of peripheral
protection with a wide therapeutic window against focal
and central mechanisms may result in persistent pain
ischemic insult in rats by suppressing the expression of
perception [65]. There is growing evidence that MAPKs
pro-inflammatory cytokines [84]. Considering the central
such as p38, ERK1/2 and JNK play a critical role in the
role that MAPK signaling, particularly p38 MAPK plays
signal transduction cascade associated with chronic
in orchestrating the immune response, glutamate-
nociception [66, 67]. The activation of MAPKs mediated by
mediated excitotoxic neuronal and glial damage,
inflammatory and stress stimuli, neurotransmitters,
modulation of synaptic function and the signal
growth factors and hormones, mediate diverse intracell-
transduction of the pain response, MAPK signaling
ular responses operative both at the transcriptional and
cascades serve as novel targets for the development of
post-transcriptional levels [68]. Protein kinase p38 MAPK
therapeutic strategies aimed at treating neuroin-
is also a potential target for the treatment of pathological
flammatory diseases that result in neurodegeneration,
pain [69]. Activation of p38 MAPK in spinal microglia is a
demyelination as well as neuropathic pain.
critical link in inflammation-induced spinal pain
processing [70], and its activation in uninjured primary
afferent neurons and in spinal microglia contribute to the
Concluding remarks
development of neuropathic pain induced by selective
motor fiber injury [71].

4
Geeta Ramesh
Neuroinflammation and Pain signaling

Accumulating evidence suggests that there are major transendothelial migration of monocytes in vitro. J. Immunol
roles for the chemokine MCP-1/CCL2 and its receptor 1995; 155:3610-3618.
CCR2 in inflammatory neurodegenerative disease, in 4. Eugenin EA, Dyer G, Calderon TM, Berman JW. HIV-1 tat
addition to the signal transduction of the pain response. protein induces a migratory phenotype in human fetal
microglia by a CCL2 (MCP-1)-dependent mechanism:
The MAPK p38/MK-2 signaling cascade has funda-
possible role in NeuroAIDS. Glia 2005; 49:501-510.
mental roles in mediating neuroinflammation, modulating http://dx.doi.org/10.1002/glia.20137
neuronal and glial survival and apoptosis, particularly via PMid:15578658
glutamate mediated excitotoxicity, besides regulating 5. Dimitrijevic OB, Stamatovic SM, Keep RF, Andjelkovic AV.
neuronal plasticity. Although the pathological processes Effects of the chemokine CCL2 on blood-brain barrier
responsible are not fully understood, there is substantial permeability during ischemia-reperfusion injury. J Cereb
evidence that activation of MAPKs is critical to Blood Flow Metab 2006; 26:797-810.
peripheral and central sensitization associated with http://dx.doi.org/10.1038/sj.jcbfm.9600229
PMid:16192992
conditions of nociceptive inflammatory and neuropathic
pain. Crosstalk between neurons and glia is an essential 6. Van der Voorn P, Tekstra J, Beelen R H J, Tensen CP, Van
der Valk P, De Groot CJA. Expression of MCP-1 by reactive
component of pathogenic pain development and
astrocytes in demyelinating multiple sclerosis lesions. Am J
increased MAPK phosphorylation in neurons and glia of Pathol 1999; 154:45-51.
the DRG, spinal cord and cortex is considered to be a http://dx.doi.org/10.1016/S0002-9440(10)65249-2
biochemical marker of pain signaling. Nociceptive- 7. Babcock AA, Kuziel WA, Rivest S, Owens T. Chemokine
induced cellular signaling in experimental pain models, expression by glial cells directs leukocytes to sites of axonal
specifically MAPK phosphorylation, can be utilized to injury in the CNS. J Neurosci 2003; 23:7922-7930.
provide mechanistic insights into drug-target interaction PMid:12944523
along the nociceptive pathways. Together, CCL2/CCR2 8. Grammas P, Ovase R. Inflammatory factors are elevated in
and MAPK signaling networks represent novel brain microvessels in Alzheimer's disease. Neurobiol Aging
therapeutic targets for the development of therapeutic 2001; 22:837-42.
http://dx.doi.org/10.1016/S0197-4580(01)00276-7
strategies aimed at limiting neuroinflammation and
neuropathic pain associated with neurological disease 9. Huang DR, Wang J, Kivisakk P, Rollins BJ, Ransohoff RM.
states. Absence of monocyte chemoattractant protein 1 in mice leads
to decreased local macrophage recruitment and
antigen-specific T helper cell type 1 immune response in
experimental autoimmune encephalomyelitis. J Exp Med
Acknowledgement 2001; 193:713-26.
http://dx.doi.org/10.1084/jem.193.6.713
The author was supported by grant 51OD011104/ PMid:11257138 PMCid:PMC2193420
P51RR000164 from the Office of Research Infrastructure
10. Lahrtz F, Piali L, Spanausk S, Seebach J, Fontana A.
Programs (ORIP) and the National Center for Research Chemokines and chemotaxis of leukocytes in infectious
Resources of the National Institutes of Health to the meningitis. J Neuroimmunol 1998; 85:33-43.
Tulane National Primate Research Center and by grant http://dx.doi.org/10.1016/S0165-5728(97)00267-1
NS048952 from the National Institute of Neurologic 11. Conductier G, Blondeau N, Guyon A, Nahon JL, Rovr C.
Disorders and Stroke to Mario T. Philipp. The role of monocyte chemo-attractant protein MCP1/CCL2
in neuroinflammatory diseases. J Neuroimmunol 2010;
224:93-100.
http://dx.doi.org/10.1016/j.jneuroim.2010.05.010
References PMid:20681057
1. Catier L, Hartley O, Dubois-Dauphin M, Krause KH. 12. Gerard C, Rollins BJ. Chemokines and disease. Nat Immunol
Chemokine receptors in the central nervous system: role in 2001; 2:108-115.
brain inflammation and neurodegenerative diseases. Brain Res http://dx.doi.org/10.1038/84209
Brain Res Rev 2005; 48:16-42. PMid:11175802
http://dx.doi.org/10.1016/j.brainresrev.2004.07.021 13. Rollins BJ. Monocyte chemo-attractant protein-1: a potential
PMid:15708626 regulator of monocyte recruitment in inflammatory disease.
2. Carr MW, Roth SJ, Luther E, Rose SS, Springer TA. Mol Med Today 2001; 2:198-204.
Monocyte chemoattractant protein-1 acts as a T-lymphocyte http://dx.doi.org/10.1016/1357-4310(96)88772-7
chemoattractant. Proc. Natl. Acad. Sci. USA 1994; 14. Simpson JE, Newcombe J, Cuzner ML, Woodroofe MN.
91:3652-3656. Expression of monocyte chemo-attractant protein-1 and other
http://dx.doi.org/10.1073/pnas.91.9.3652 beta-chemokines by resident glia and inflammatory cells in
3. Randolf GJ, Furie MB. A soluble gradient of endogenous multiple sclerosis lesions. J Neuroimmunol 1998; 84:238-49.
monocyte chemoattractant protein-1 promotes the http://dx.doi.org/10.1016/S0165-5728(97)00208-7

5
Geeta Ramesh
Neuroinflammation and Pain signaling

15. Ransohoff RM, Hamilton TA, Tani M, Stoler MH, Shick HE, 26. F. A. White, P. Feldman, R. J. Miller, Chemokine signaling
Major JA, et al. Astrocyte expression of mRNA encoding and the management of neuropathic pain. Mol Interv 2009;
cytokines IP-10 and JE/MCP-1 in experimental autoimmune 9:188-195.
encephalomyelitis. FASEB 1993; 7:592-600 http://dx.doi.org/10.1124/mi.9.4.7
16. Hinojosa AE, Garcia-Bueno B, Leza JC, Madrigal JL. PMid:19720751 PMCid:PMC2861804
CCL2/MCP- 1 modulation of microglial activation and 27. Greco R, Tassorelli C, Sandrini G, Di Bella P, Buscone S,
proliferation. J Neuroinflammation 2011; 8:77-86. Nappi G. Role of calcitonin gene-related peptide and
http://dx.doi.org/10.1186/1742-2094-8-77 substance P in different models of pain. Cephalalgia 2008;
PMid:21729288 PMCid:PMC3146846 28:114-126.
17. Perrin FE, Lacroix S, Avils-Trigueros M, David S. PMid:18197882
Involvement of monocyte chemo-attractant protein-1, 28. Ramesh G, Santana-Gould L, Inglis FM, England JD, Philipp
macrophage inflammatory protein-1 alpha and interleukin-1 MT. The Lyme disease spirochete Borrelia burgdorferi
beta in Wallerian degeneration. Brain 2005; 128:854-866. induces inflammation and apoptosis in cells from dorsal root
http://dx.doi.org/10.1093/brain/awh407 ganglia. J Neuroinflammation 2013; 10:88-101.
PMid:15689362 http://dx.doi.org/10.1186/1742-2094-10-88
18. Grygorczuk S, Zajkowska J, Swierzbiska R, Pancewicz S, PMid:23866773 PMCid:PMC3721987
Kondrusik M, Hermanowska-Szpakowicz T. Concentration of 29. Zhang H, Boyette-Davis JA, Kosturakis AK, Li Y, Yoon SY,
interferon-inducible T cell chemoattactant and monocyte Walters ET, et al. Induction of monocyte chemoattractant
chemo-attractant protein-1 in serum and cerebrospinal fluid of protein-1 (MCP-1) and its receptor CCR2 in primary sensory
patients with Lyme borreliosis. Rocz Akad Med Bialymst neurons contributes to paclitaxel-induced peripheral
2005; 50:173-178. neuropathy. J Pain 2013; 10:1031-1044.
PMid:16358960 http://dx.doi.org/10.1016/j.jpain.2013.03.012
19. Stadelmann C, Wegner C, Brck W. Inflammation, PMid:23726937
demyelination and degeneration-recent insights from MS 30. Ramesh G, MacLean AG, Philipp MT. Cytokines and
pathology. Biochem Biophys Acta 2011; 1812:275-282. Chemokines at the Crossroads of Neuroinflammation,
20. Ramesh G, Borda JT, Gill A, Ribka EP, Morici LA, Mottram Neurodegeneration, and Neuropathic Pain. Review. Mediators
P, et al. Possible role of glial cells in the onset and progression of Inflammation, Special issue, Mediators of
of Lyme neuroborreliosis. J Neuroinflammation 2009; Neuroinflammation 07/2013; Volume 2013, Article ID
6:23-38. 480739. DOI: http://dx.doi.org/10.1155/2013/480739.
http://dx.doi.org/10.1186/1742-2094-6-23 http://dx.doi.org/10.1155/2013/480739
PMid:19706181 PMCid:PMC2748066 31. Cuadrado A, Nebreda A. Mechanisms and functions of p38
21. Kinstrand E, Nilsson BY, Hovmark A, Nennesmo I, Pirskanen MAPK signaling. Biochem J 2010; 429:403-417.
R, Solders G, et al. Polyneuropathy in late Lyme borreliosis - http://dx.doi.org/10.1042/BJ20100323
a clinical, neurophysiological and morphological description. PMid:20626350
Acta Neurol Scand 2000; 101:47-52. 32. Ono K, Han J. The p38 signal transduction pathway:
22. Yang G, Meng Y, Li W, Yong Y, Fan Z, Ding H, et al. activation and function. Cell Signal 2000; 12:1-13.
Neuronal MCP-1 mediates microglia recruitment and http://dx.doi.org/10.1016/S0898-6568(99)00071-6
neurodegeneration induced by the mild impairment of 33. Kumar S, Boehm1 J, John C, Lee JC. p38 MAP kinases: key
oxidative metabolism. Brain Pathol 2011; 21:279-297. signaling molecules as therapeutic targets for inflammatory
http://dx.doi.org/10.1111/j.1750-3639.2010.00445.x diseases. Nat Rev Drug Discov 2003; 2:717-726.
PMid:21029241 PMCid:PMC3046243 http://dx.doi.org/10.1038/nrd1177
23. Thompson WL, Karpus WJ, Van Eldik LJ. MCP-1-deficient PMid:12951578
mice show reduced neuroinflammatory responses and 34. Brook M, Sully G, Clark AR, Saklatvala J. Regulation of
increased peripheral inflammatory responses to peripheral TNF- mRNA stability by the MAPK p38 signaling cascade.
endotoxin insult. J Neuroinflammation 2008; 5:35. FEBS lett 2000; 483:57-61.
http://dx.doi.org/10.1186/1742-2094-5-35 http://dx.doi.org/10.1016/S0014-5793(00)02084-6
PMid:18706086 PMCid:PMC2527558 35. Goebeler M, Kilian K, Gliltzer R, Kunz M, Yoshimura T,
24. Abbadie C, Lindia JA, Cumiskey AM, Peterson LB, Mudgett Brocker EB, et al. The MKK6/p38 stress kinase cascade is
JS, Bayne EK, et al. Impaired neuropathic pain responses in critical for TNF--induced expression of MCP-1 in
mice lacking the chemokine receptor CCR2. Proc Natl Acad endothelial cells. Blood 1999; 93:857-865.
Sci 2003; 100: 7947-7952. PMid:9920834
http://dx.doi.org/10.1073/pnas.1331358100 36. Cho J, Gruol DL. The chemokine CCL2 activates p38
PMid:12808141 PMCid:PMC164693 mitogen-activated protein kinae pathway in cultured rat
25. White FA, Bhangoo SK, Miller RJ. Chemokines: integrators hippocampal cells. J Neuroimmunol 2008; 199:94-103.
of pain and inflammation. Nat Rev Drug Discov 2005; http://dx.doi.org/10.1016/j.jneuroim.2008.05.011
4:834-844. PMid:18584881 PMCid:PMC2583399
http://dx.doi.org/10.1038/nrd1852 37. Wang XJ, Kong KM, Qi WL, Ye WL, Song PS. Interleukin-1
PMid:16224455 PMCid:PMC2792904 beta induction of neuron apoptosis depends on p38
mitogen-activated protein kinase activity after spinal cord

6
Geeta Ramesh
Neuroinflammation and Pain signaling

injury. Acta Pharmacol Sin 2005; 8:934-942. http://dx.doi.org/10.1196/annals.1403.002


http://dx.doi.org/10.1111/j.1745-7254.2005.00152.x PMid:18077562
PMid:16038625 48. Ramesh G, Alvarez AL, Roberts ED, Dennis VA, Lasater BA,
38. Chaparro-Huerta V, Rivera-Cervantes MC, Flores-Soto ME, Alvarez X, et al. Pathogenesis of Lyme neuroborreliosis:
Gomez-Pinedo U, Beas-Zarate C. Proinflammatory cytokines Borrelia burgdorferi lipoproteins induce both proliferation and
and apoptosis following glutamate-induced excitotoxicity apoptosis in rhesus monkey astrocytes Eur. J. Immunol 2003;
mediated by p38 MAPK in the hippocampus of neonatal rats. 33:2539-2550.
J Neuroimmunol 2005; 165:53-62. http://dx.doi.org/10.1002/eji.200323872
http://dx.doi.org/10.1016/j.jneuroim.2005.04.025 PMid:12938230
PMid:15972237 49. Ramesh G, Philipp MT. Pathogenesis of Lyme
39. Rivera-Cervantes MC, Torres JS, Feria-Velasco A, neuroborreliosis: Mitogen-activated protein kinases Erk1,
Armendariz-Borunda J, Beas-Zrate C. NMDA and AMPA Erk2, and p38 in response of astrocytes to Borrelia
receptor expression and cortical neuronal death are associated burgdorferi lipoproteins. Neurosci Lett 2005; 384:112-116.
with p38 in glutamate-induced excitotoxicity in vivo. J http://dx.doi.org/10.1016/j.neulet.2005.04.069
Neurosci Res 2004; 76:678-687. PMid:15893422
http://dx.doi.org/10.1002/jnr.20103 50. Harper SJ, Lograsso P. Signalling for survival and death in
PMid:15139026 neurons: the role of stress-activated kinases, JNKand p38.
40. Segura Torres JE, Chaparro-Huerta V, Rivera Cervantres MC, Cell Signal 2001; 13:299-310.
Montes-Gonzlez R, Flores-Soto ME, Beas-Zrate C. http://dx.doi.org/10.1016/S0898-6568(01)00148-6
Neuronal cell death due to glutamate excitotocity is mediated 51. Obata T, Brown GE, Yaffe MB. Map kinase pathways
by p38 activation in the rat cerebral cortex. Neurosci Lett activated by stress: the p38 MAPK pathway. Crit Care Med
2006; 403:233-238. 2000; 28:67-77.
http://dx.doi.org/10.1016/j.neulet.2006.04.063 http://dx.doi.org/10.1097/00003246-200004001-00008
PMid:16782274
52. Han J, Lee JD, Bibbs L, Ulevitch RJ. A MAP kinase targeted
41. Kim SH, Kim J, Sharma RP. Inhibition of p38 and ERK MAP by endotoxin and hyperosmolarity in mammalian cells.
kinases blocks endotoxin-induced nitric oxide production and Science 1994; 265:808-811.
differentially modulates cytokine expression. Pharmacol Res http://dx.doi.org/10.1126/science.7914033
2004; 49:433-439. PMid:7914033
http://dx.doi.org/10.1016/j.phrs.2003.11.004
PMid:14998552 53. Rouse J, Cohen PS, Trigon S. A novel kinase cascade
triggered by stress and heat shock that stimulates MAPKAP
42. Chaparro-Huerta V, Flores-Soto ME, Gudi-o-Cabrera G, kinase-2 and phosphorylation of the small heat shock proteins.
Rivera-Cervantes MC, Bitzer-Quintero OK, Beas-Zrate C. Cell 1994; 78:1027-1037.
Role of p38 MAPK and pro-inflammatory cytokines http://dx.doi.org/10.1016/0092-8674(94)90277-1
expression in glutamate-induced neuronal death of neonatal
rats. Int J Dev Neurosci 2008; 26:487-495. 54. Freshney NW, Rawlinson L, Guesdon F. Interleukin-1
http://dx.doi.org/10.1016/j.ijdevneu.2008.02.008 activates a novel protein kinase cascade that results in the
PMid:18396383 phosphorylation of Hsp27. Cell 1994; 78:1039-1049.
http://dx.doi.org/10.1016/0092-8674(94)90278-X
43. Kim SW, Yu YM, Piao CS, Kim JB, Lee JK. Inhibition of
delayed induction of p38 mitogen-activated protein kinase 55. Shi Y, Gaestel M. In the cellular garden of forking paths: how
attenuates kainic acid-induced neuronal loss in the p38 MAPKs signal for downstream assistance. Biol Chem
hippocampus. Brain Res 2004; 1007:188-191. 2002; 383:1519-1536.
http://dx.doi.org/10.1016/j.brainres.2004.02.009 http://dx.doi.org/10.1515/BC.2002.173
PMid:15064151 PMid:12452429
44. Sweatt JD. The neuronal MAP kinase cascade: a biochemical 56. Gaestel M. MAPKAP kinasesMKstwo's company, three's
signal integration system subserving synaptic plasticity and a crowd. Nat Rev Mol Cell Biol 2006; 7:120-130.
memory. J Neurochem 2001; 76:1-10. http://dx.doi.org/10.1038/nrm1834
http://dx.doi.org/10.1046/j.1471-4159.2001.00054.x PMid:16421520
PMid:11145972 57. Kotlyarov A, Neininger A, Schubert C. MAPKAP kinase 2 is
45. Streit WJ, Mrak RE, Griffin WS. Microglia and essential for LPS-induced TNF- biosynthesis. Nat Cell Biol
neuroinflammation: a pathological perspective. J 1999; 1:94-97.
Neuroinflammation 2004; 1:14-17. http://dx.doi.org/10.1038/10061
http://dx.doi.org/10.1186/1742-2094-1-14 PMid:10559880
PMid:15285801 PMCid:PMC509427 58. Fyhrquist N, Matikainen S, Lauerma A. MK2 signaling:
46. Chao C, Hu S, Peterson P. Glia, cytokines, and neurotoxicity. lessons on tissue specificity in modulation of inflammation. J
Crit Rev Neurobiol 1995; 9:189-205. Invest Dermatol 2010; 130:342-344.
PMid:8581983 http://dx.doi.org/10.1038/jid.2009.372
PMid:20081887
47. Skaper SD. The brain as a target for inflammatory processes
and neuroprotective strategies. Ann N Y Acad Sciences 2007; 59. Neininger A, Kontoyiannis D, Kotlyarov A. MK2 targets
1122:23-34. AU-rich elements and regulates biosynthesis of tumor
necrosis factor and interleukin-6 independently at different
Geeta Ramesh
Neuroinflammation and Pain signaling

post-transcriptional levels. J Biol Chem 2002; 277:3065-3068. 204:355-65.


http://dx.doi.org/10.1074/jbc.C100685200 http://dx.doi.org/10.1016/j.expneurol.2006.11.016
PMid:11741878 PMid:17258708
60. Culbert AA, Skaper SD, Howlett DR. MAPKactivated protein 72. Jin SX, Zhuang ZY, Woolf CJ, Ji RR. p38 mitogen-activated
kinase 2 deficiency in microglia inhibits pro-inflammatory protein kinase is activated after a spinal nerve ligation in
mediator release and resultant neurotoxicity: relevance to spinal cord microglia and dorsal root ganglion neurons and
neuroinflammation in a transgenic mouse model of Alzheimer contributes to the generation of neuropathic pain. J Neurosci
disease. J Biol Chem 2006; 281:23658-23667. 2003; 23:4017-22.
http://dx.doi.org/10.1074/jbc.M513646200 PMid:12764087
PMid:16774924 73. Cao H, Gao YJ, Ren WH, Li TT, Duan KZ, Cui YH, et al.
61. Thomas T, Timmer M, Cesnulevicius-Hitti KE, Kotlyarov A, Activation of extracellular signal-regulated kinase in the
Gaestel M. MAPKAP kinase 2-deficiency prevents neurons anterior cingulate cortex contributes to the induction and
from cell death by reducing neuroinflammation- relevance in expression of affective pain. J Neurosci 2009; 29:3307-21.
a mouse model of Parkinson's disease. J Neurochem 2008; http://dx.doi.org/10.1523/JNEUROSCI.4300-08.2009
105:2039-2052. PMid:19279268 PMCid:PMC2682784
http://dx.doi.org/10.1111/j.1471-4159.2008.05310.x 74. Ji RR, Woolf CJ. Neuronal plasticity and signal transduction
PMid:18298661 in nociceptive neurons: implications for the initiation and
62. Correa SA, Eales KL. The Role of p38 MAPK and Its maintenance of pathological pain. Neurobiol Dis 2001;
Substrates in Neuronal Plasticity and Neurodegenerative 8:1-10.
Disease. J Signal Transduct 2012; 2012: 649079. http://dx.doi.org/10.1006/nbdi.2000.0360
63. Yasuda S, Sugiura H, Tanaka H, Takigama S, Yamagata K. PMid:11162235
p38 MAP kinase inhibitors as potential therapeutic drugs for 75. Sweitzer SM, Peters MC, Ma JY, Kerr I, Mangadu R,
neural diseases. Cent Nerv Syst Agents Med Chem 2011; Chakravarty S, et al. Peripheral and central p38 MAPK
1191:45-59. mediates capsaicin-induced hyperalgesia. Pain 2004; 111:
http://dx.doi.org/10.2174/187152411794961040 278-285.
64. Wolf CJ, Walters ET. Common patterns of plasticity http://dx.doi.org/10.1016/j.pain.2004.07.007
contributing to nociceptive sensitization in mammals and PMid:15363871
Aplysia. Trends Neurosci 1991; 14:4-8. 76. Dai Y, Iwata K, Fukuoka T, Kondo E, Tokunaga A,
65. Campbell JN, Meyer RA. Mechanisms of neuropathic pain. Yamanaka H, et al. Phosphorylation of extracellular
Neuron 2006; 52:77-92. signal-regulated kinase in primary afferent neurons by
http://dx.doi.org/10.1016/j.neuron.2006.09.021 noxious stimuli and its involvement in peripheral
PMid:17015228 PMCid:PMC1810425 sensitization. J Neurosci 2002; 22:7737-45.
PMid:12196597
66. Ji RR, Gereau RW, Malcangio M, Strichartz GR. MAP kinase
and pain. Brain Res Rev 2009; 60:135-148. 77. Kim SY, Bae JC, Kim JY, Lee HL, Lee KM, Kim DS, et al.
http://dx.doi.org/10.1016/j.brainresrev.2008.12.011 Activation of p38 MAP kinase in the rat dorsal root ganglia
PMid:19150373 PMCid:PMC2666786 and spinal cord following peripheral inflammation and nerve
injury. Neuroreport 2002; 13:2483-6.
67. Ji RR. Peripheral and central mechanisms of inflammatory http://dx.doi.org/10.1097/00001756-200212200-00021
pain, with emphasis on MAP kinases. Curr Drug Targets PMid:12499853
Inflamm Allergy 2004; 3:299-303.
http://dx.doi.org/10.2174/1568010043343804 78. Obata K, Yamanaka H, Kobayashi K, Dai Y, Mizushima T,
Katsura H, et al. Role of mitogen-activated protein kinase
68. Widmann C, Gibson S, Jarpe MB, Johnson GL. activation in injured and intact primary afferent neurons for
Mitogen-activated protein kinase: conservation of a mechanical and heat hypersensitivity after spinal nerve
three-kinase module from yeast to human. Physiol Rev 1999; ligation. J Neurosci 2004; 24:10211-22.
79:143-180. http://dx.doi.org/10.1523/JNEUROSCI.3388-04.2004
PMid:9922370 PMid:15537893
69. Ji RR, Kawasaki Y, Zhuang ZY, Wen YR, Zhang YQ. Protein 79. Schafers M, Svensson CI, Sommer C, Sorkin LS. Tumor
kinases as potential targets for the treatment of pathological necrosis factor-alpha induces mechanical allodynia after
pain. Handb Exp Pharmacol 2007; (177): 359-389. spinal nerve ligation by activation of p38 MAPK in primary
70. Svensson CI, Marsala M, Westerlund A, Calcutt NA, sensory neurons. J Neurosci 2003; 23:2517-21.
Campana WM, Freshwater JD, et al. Activation of p38 PMid:12684435
mitogen-activated protein kinase in spinal microglia is a 80. Svensson CI, Schafers M, Jones TL, Yaksh TL, Sorkin LS.
critical link in inflammation-induced spinal pain processing. J Covariance among age, spinal p38 MAP kinase activation and
Neurochem 2003; 86:1534-1544. allodynia. J Pain 2006; 7:337-45.
http://dx.doi.org/10.1046/j.1471-4159.2003.01969.x http://dx.doi.org/10.1016/j.jpain.2005.12.007
PMid:12950462 PMid:16632323
71. Xu JT, Xin WJ, Wei XH, Wu CY, Ge YX, Liu YL, et al. p38 81. Zhuang ZY, Gerner P, Woolf CJ, Ji RR. ERK is sequentially
activation in uninjured primary afferent neurons and in spinal activated in neurons, microglia, and astrocytes by spinal nerve
microglia contributes to the development of neuropathic pain ligation and contributes to mechanical allodynia in this
induced by selective motor fiber injury. Exp Neurol 2007;

8
Geeta Ramesh
Neuroinflammation and Pain signaling

neuropathic pain model. Pain 2005; 114:149-59. 84. Piao CS, Kim JB, Han PL, Lee JK. Administration of the p38
http://dx.doi.org/10.1016/j.pain.2004.12.022 MAPK inhibitor SB203580 affords brain protection with a
PMid:15733640 wide therapeutic window against focal ischemic insult. J
82. Edelmayer RM, Brederson JD, Jarvis MF, Bitner RS. Neurosci Res 2003; 73:537-544.
Biochemical and pharmacological assessment of MAP-kinase http://dx.doi.org/10.1002/jnr.10671
signaling along pain pathways in experimental rodent models: PMid:12898538
a potential tool for the discovery of novel antinociceptive
therapeutics. Biochem Pharmacol 2014; 87:390-8. To cite this article: Geeta Ramesh. Novel Therapeutic Targets in
http://dx.doi.org/10.1016/j.bcp.2013.11.019 Neuroinflammation and Neuropathic Pain. Inflamm Cell Signal
PMid:24300134 2014; 1: e111. doi: 10.14800/ics.111.
83. Saklatvala J. The p38 MAP kinase pathway as a therapeutic
target in inflammatory disease. Curr Opin Pharmacol. 2004,
4:372-377.
http://dx.doi.org/10.1016/j.coph.2004.03.009
PMid:15251131

Você também pode gostar