ArticlePDF AvailableLiterature Review

NGF/TrkA Signaling as a Therapeutic Target for Pain

Authors:
  • Hyogo Medical University Faculty of Medicine

Abstract and Figures

Nerve growth factor (NGF) was first discovered approximately 60 years ago by Rita Levi-Montalcini as a protein that induces the growth of nerves. It is now known that NGF is also associated with Alzheimer's disease and intractable pain, and hence, it, along with its high-affinity receptor, tropomyosin receptor kinase (Trk) A, is considered to be 1 of the new targets for therapies being developed to treat these diseases. Anti-NGF antibody and TrkA inhibitors are known drugs that suppress NGF/TrkA signaling, and many drugs of these classes have been developed thus far. Interestingly, local anesthetics also possess TrkA inhibitory effects. This manuscript describes the development of an analgesic that suppresses NGF/TrkA signaling, which is anticipated to be 1 of the new methods to treat intractable pain.
Content may be subject to copyright.
REVIEW ARTICLE
NGF/TrkA Signaling as a Therapeutic Target
for Pain
Munetaka Hirose, MD, PhD*; Yoshihiro Kuroda, PhD
; Eri Murata, VMD, MS
*Department of Anesthesiology and Pain Medicine, Hyogo College of Medicine, Hyogo;
Department of Pharmaceutical Health Care, Faculty of Pharmaceutical Sciences, Himeji
Dokkyo University, Hyogo;
Department of Anesthesiology and Reanimatology, Faculty of
Medical Sciences, University of Fukui, Fukui, Japan
&Abstract: Nerve growth factor (NGF) was first discov-
ered approximately 60 years ago by Rita Levi-Montalcini as
a protein that induces the growth of nerves. It is now
known that NGF is also associated with Alzheimer’s disease
and intractable pain, and hence, it, along with its high-
affinity receptor, tropomyosin receptor kinase (Trk) A, is
considered to be 1 of the new targets for therapies being
developed to treat these diseases. Anti-NGF antibody and
TrkA inhibitors are known drugs that suppress NGF/TrkA
signaling, and many drugs of these classes have been
developed thus far. Interestingly, local anesthetics also
possess TrkA inhibitory effects. This manuscript describes
the development of an analgesic that suppresses NGF/TrkA
signaling, which is anticipated to be 1 of the new methods
to treat intractable pain. &
Key Words: local anesthetic, nerve growth factor, NGF,
pain, tropomyosin receptor kinase, protein kinase, TrkA, anti-
NGF antibody, tanezumab, review
INTRODUCTION
Levi-Montalcini, who continually conducted research on
the growth of nerve fibers, discovered that mouse
sarcomas transplanted into chicken embryos secrete a
factor into the blood which induces sensory and sympa-
thetic nerve growth.
1,2
Furthermore, it was demonstrated
that sympathetic neurons become denatured when an
antiserum against this factor is injected into newborn
mammals.
3
This factor, indispensable for the prenatal
growth of sensory and sympathetic nerves, was named
“nerve growth factor (NGF).” A tissue sample isolated
from a mouse submandibular gland revealed that NGF is
composed of 118 amino acid residue sequences.
4
More-
over, it was discovered that there are 2 NGF receptors, 1
with high affinity and the other with low affinity for
NGF,
5
and these were later named tropomyosin receptor
kinase (Trk) A and p75 neurotrophin receptor
(p75NTR), respectively. Subsequently, aside from
NGF, brain-derived neurotrophic factor (BDNF), neu-
rotrophin-3 (NT-3), and neurotrophin-4/5 (NT-4/5)
were discovered as other factors involved in nerve
growth. These were collectively termed “neurotrophic
factors.” Thereafter, the receptors for each of these
factors, TrkB and TrkC, which have very similar amino
acid sequences as TrkA, were discovered. TrkB was
shown to be the receptor for BDNF and NT-4/5, while
TrkC was indicated as the receptor for NT-3.
6
Currently, NGF is known not only for its function in
prenatal nerve growth, but also for its significant role in
pain and immune function in adults. This manuscript
Address correspondence and reprint requests to: Munetaka Hirose,
MD, PhD, Department of Anesthesiology and Pain Medicine, Hyogo
College of Medicine, 1-1 Mukogawa-cho, Nishinomiya, Hyogo 663-8501,
Japan. E-mail: mhirose@hyo-med.ac.jp.
Conflict of Interests: The authors have no conflict of interests or
financial ties to disclose.
Submitted: February 15, 2015; Revision accepted: June 15, 2015
DOI. 10.1111/papr.12342
©2015 World Institute of Pain, 1530-7085/15/$15.00
Pain Practice, Volume , Issue , 2015 
describes the development of analgesics that target
NGF/TrkA signaling.
TRKA ACTIVATION BY NGF
TrkA is expressed in various organs and tissues, such as
the peripheral nervous system, central nervous system,
immune tissue, digestive tract, adrenal cortex, prostate,
uterus, kidney, and skin.
6,7
NGF binds to TrkA on the
cell membrane, which possesses tyrosine kinase activity
that phosphorylates tyrosine in amino acid residues. As
shown in Figure 1, the activation loop of TrkA is
wedged in the center of the enzymatic activity site in an
inactivate state, and this prevents the adenosine triphos-
phate (ATP) from entering the site, consequently
suppressing the tyrosine kinase activity.
8
When the
NGF dimer binds to the TrkA dimer,
9
the activation
loop is released from the center of the enzymatic activity
site, following which TrkA autophosphorylates (pY)
tyrosine residues (Y676, Y680, Y681) on the contralat-
eral activation loop with ATP
8
(Figures 1 and 2). This
activated form of TrkA phosphorylates other intracel-
lular matrix proteins, which then trigger the intracellu-
lar signal transduction system of NGF/TrkA signaling,
transmitting signals into the nucleus.
In particular, NGFs that act on the peripheral
nociceptive neuron terminals bind to TrkA on the cell
membrane, which are then taken up by endosomes and
are subsequently transported in a retrograde manner
through the axon to the dorsal root ganglia cell bodies.
There, the downstream intracellular signal transduction
system is activated, producing various types of
proteins.
10
NGF/TRKA SIGNALING AND PAIN
NGFs are involved in pain in 2 distinct ways. The first is
during the fetal period via the growth of nerve fibers that
transmit pain sensations, and the other is via the role
played during adulthood in inducing pain.
Congenital Insensitivity to Pain
When NGF-mediated nerve growth is absent during the
fetal period, insensitivity to pain develops. In 1976,
when it was already known that NGF is essential in the
formation of sensory and sympathetic nerves, the blood
levels of NGF were measured in patients with congenital
insensitivity to pain, who inherently do not sense pain.
However, this study did not investigate the cause of this
disorder.
11
With advances in molecular biology, it was subse-
quently discovered that a genetic mutation in TrkA was
the cause of congenital insensitivity to pain together
with anhidrosis. This is a disorder in which nociceptive
and sympathetic nerves are missing. It is an extremely
rare disorder where patients suffer repetitive injuries
because they do not feel pain, and develop high
temperatures upon exercising due to their inability to
sweat, both caused by defective tyrosine kinase activity
of TrkA.
12,13
In addition, reportedly, in a variant
congenital insensitivity to pain (hereditary sensory and
autonomic neuropathy type 5), where there is insensi-
tivity to pain but normal ability to sweat, a mutation in
the NGF gene
14
leads to a defect in the physiological
actions of prenatal NGF.
NGF-induced Pain in Adulthood
When NGF actions are continuously suppressed by
anti-NGF antibodies from the prenatal through the
neonatal period, the growth of sensory and sympa-
thetic nerves is completely inhibited. However, if NGF
actions are suppressed postnatally, only a portion of
nerve growth is inhibited.
15
This suggests a difference
in physiological actions of NGF between prenatal and
postnatal stages.
Due to its nerve-growing properties, NGF gained
interest in the late 1980s as a candidate target for a
therapeutic agent for central and peripheral nervous
system disorders, and a study that administered NGF
into animals was conducted.
16
This study revealed that
hyperalgesia develops when NGF is administered to
adult rats, following which NGF became known as 1 of
Figure 1. Activation of high-affinity receptor TrkA by NGF.
2HIROSE ET AL.
the chemical substances that induce pain during adult-
hood.
17
In a study that investigated the effects of NGF in
humans, its intravenous administration induced whole-
body muscle pain, and subcutaneous administration at
the same dose induced hyperalgesia of the skin at the
injection site in addition to whole-body muscle pain.
Muscle pain continued for nearly a week, and hyperal-
gesia of the skin persisted for several weeks.
18
Subsequently, the mechanism of action of NGF was
investigated. Physical pain can be divided into 2 types:
nociceptive and neuropathic pain. Nociceptive pain is
defined as “pain that arises from actual or threatened
damage to non-neural tissue and is due to the activation
of nociceptors,” and is generally interpreted as physio-
logical pain.
19
In contrast, neuropathic pain is defined as
“pain caused by a lesion or disease of the somatosensory
nervous system”
19
and is considered to be a pathological
pain that does not arise normally. Both types of pain can
develop into chronic pain that persists for a long period
of time, with treatments for such conditions being
difficult to determine. NGF, interleukins, and tumor
necrosis factor (TNF)-aare secreted by inflammatory
cells in injured tissue and by Schwann cells in damaged
nerves and are involved in both types of pain.
20,21
When
tissue injury occurs, NGF expression at the site of injury
increases.
22,23
NGFs secreted by inflammatory cells act
on TrkA located on the cell membrane of the sensory
nerve endings, phosphorylating other proteins associ-
ated with pain, which induces a conformational change
and increases the expression of these proteins. It is
believed that these in turn elicit peripheral sensitization
in the peripheral nerve and central sensitization in the
spinal cord, inducing the onset of a hypersensitive
reaction (hyperalgesia) in response to pain, together
with allodynia, a painful response to a stimulus that is
not normally painful.
2426
In addition to neuropathic pain in the arm or leg
ipsilateral to peripheral nerve lesions, mirror image pain
also occurs in the contralateral sites.
27
NGF may be
involved in the mechanisms of mirror image pain
pathogenesis.
28
NGF as a Cause of Pain
It is curious why NGF, an essential protein for the
prenatal growth of sensory and sympathetic nerves,
plays a role in inducing pain postnatally. Although the
exact reason is unknown, it is possible that when an
inflammatory response occurs due to tissue damage,
NGF required for the repair of the injured peripheral
nerve, which occurs simultaneously with this inflamma-
tory response, acts on the surrounding sensory nerves
that are not injured, thereby inducing pain and protect-
ing the injured site. This may, consequently, promote the
repair of injured peripheral nerves.
In conditions in which the inflammatory response
continues and nociceptive pain is constant, such as with
autoimmune diseases, or in a disease state that induces
neuropathic pain, NGF triggers peripheral and central
sensitization, thereby causing hyperalgesia and allody-
nia. In such pathological conditions, an analgesic that
blocks NGF/TrkA signaling would be extremely useful.
NGF/TRKA SIGNALING AND THE DEVELOPMENT
OF ANALGESICS
In modern society, approximately 20 to 30% of adults
suffer from refractory pain such as chronic low back
Figure 2. Amino acid sequences of the
TrkA activation loop and TrkA activity
inhibitor IPTRK3.
TrkA and Pain 3
pain that cannot be suitably alleviated even with the use
of various analgesics. This is a large societal issue as it
leads to a decreased productivity.
29
The development of
novel analgesics is, therefore, desirable. The NGF/TrkA
signaling that is involved in both nociceptive and
neuropathic pain is considered to be 1 of the important
targets for such therapeutic development.
3032
In
particular, clinical studies on anti-NGF antibody have
already been conducted, and its clinical use as a new
analgesic is anticipated in the future. Additionally, the
development of drugs that block TrkA activity is also
ongoing.
Classification of NGF/TrkA Inhibitors
Drugs that suppress NGF actions are classified into the
following 3 types
30,31
: (1) drugs that capture NGF; (2)
drugs that inhibit the binding of NGF and TrkA; and (3)
drugs that directly inhibit the enzymatic activity of TrkA
(Figure 3). Many NGF/TrkA inhibitors have been
developed in the past, as described below (Table 1).
3348
Anti-NGF Antibodies. Anti-NGF antibody is the oldest
known NGF/TrkA inhibitor; it captures NGF. Its
efficacy in suppressing nociceptive pain was demon-
strated in the early 1990s,
33
and later, its analgesic
effects were shown in various experimental animal
models of nociceptive and neuropathic pain. Trunk burn
injury in rats is known to induce hyperalgesia at the
bottom of the hind paw in an area outside the region of
burn injury. It was later shown that NGF produced at
the burn site induces hyperalgesia at a distant location
and that this phenomenon can be suppressed by an anti-
NGF antibody.
49
Subsequently, anti-NGF antibody was clinically used
in humans, and a clinical study of tanezumab was
conducted in the USA to investigate its efficacy in
osteoarthritis of the knee and hip.
34
Although a long-
term analgesic effect was achieved with intravenous
administration of the drug every 2 months, the possible
development of side effects, including hyperesthesia,
hypalgesia, and exacerbation of osteoarthritis and
osteonecrosis, was noted and further study was discon-
tinued in 2010. Later, it was revealed that the exacer-
bation of osteoarthritis and osteonecrosis was
associated with the long-term usage combined with
nonsteroidal anti-inflammatory drugs (NSAIDs) and
with high doses of tanezumab.
50
Studies have resumed,
and anti-NGF antibody may eventually gain use as an
analgesic, not only for osteoarthritis, but also for
cancer
51,52
and postoperative pain.
53
TrkA Activity Inhibitors. As the enzymatic activity site
of TrkA is located intracellularly (Figure 1), it is
necessary for TrkA activity inhibitors to have the ability
to enter the cell (Figure 3). For this reason, these drugs
are either small molecules or require the addition of a
peptide that promotes cell membrane permeability (cell-
penetrating peptide).
The first reported cell-penetrating peptide was an
amino acid sequence (Tat: YGRKKRRQRRR) within a
segment of the acquired immune deficiency syndrome
(AIDS) virus protein
54
. One of the enigmatic properties
of this cell-penetrating peptide is that its addition to
large molecules, such as deoxyribonucleic acid (DNA),
oligonucleotides, peptides, and proteins, which cannot
normally cross the cell membrane, enables them to pass
through the cell membrane.
Previously, we created a peptide in which this Tat
peptide is bound to a TrkA activity-suppressing peptide
with a linker (e-aminocaproic acid: acp), to develop a
TrkA activity inhibitor (IPTRK3) (Figure 2).
42
As TrkA
activity-suppressing peptide has an amino acid sequence
that is partially equivalent to that of the TrkA activation
loop, it is thought to suppress TrkA activity by acting as
Figure 3. Site of action of NGF/TrkA signaling inhibitor.
Table 1. NGF/TrkA Signaling Inhibitors
Drugs capturing NGF Anti-NGF antibody (ABT-110, alpha-D11,
AMG403, fulranumab, Medi-578,
muMab911, REGN475, tanezumab)
3335
TrkA-IgG
36
TrkAd5
37
Inhibitors of
NGF binding to TrkA
Anti-TrkA antibody (MNAC13)
38
ALE0540
39
PD90780
40
TrkA inhibitors ARRY-470
41
, ARRY-872
CT-327, CT-335, CT-340
IPTRK3
4245
K252a
46,47
NMS-P626
TrkA antisense oligodeoxynucleotide
48
4HIROSE ET AL.
a decoy for the activation loop and embedding itself into
the center of the enzymatic activity site. IPTRK3 has
been reported to inhibit nociceptive pain induced by
Freund’s complete adjuvant in rats,
43
neuropathic pain
generated by partial sciatic nerve ligation in mice,
44
and
cancer pain caused by malignant melanoma inoculation
into mouse hind paws.
45
Subsequently, it was demon-
strated that IPTRK3 also suppresses the activity of
numerous protein kinases (Erk, Janus kinase (JAK), p38,
protein kinase C (PKC)) that are involved in pain
(unpublished data) (Figure 4), and the development of
small molecule TrkA activity inhibitors that more
selectively suppress TrkA activity is presently under-
way.
55
The ability of TrkA activity inhibitors to penetrate
the cell membrane makes it possible for them to be
taken up by the central nervous system. However, the
fact that NGF depletion in the central nervous system
can induce Alzheimer’s disease
56
and that activation of
NGF/TrkA signaling may suppress the onset of
Alzheimer’s disease
57
may be a significant deterrent
to the use of TrkA activity inhibitors. Surprisingly,
TrkA inhibitors have conversely been indicated to be a
potential therapeutic agent for Alzheimer’s disease.
58
Studies investigating the effects of long-term adminis-
tration of TrkA activity inhibitors on the central
nervous system are needed.
LOCAL ANESTHETICS AND NGF/TRKA SIGNALING
Although it is not well known, local anesthetics suppress
NGF/TrkA signaling. In cell culture experiments, where
neurite outgrowth occurred with the addition of NGF
into the petri dish, it was reported that 40 to 50 lMof
lidocaine suppresses this NGF-mediated neurite out-
growth.
59,60
Lidocaine is known to bind to the cytoplasmic linker
between domains III and IV of the sodium channel,
61
and the amino acid sequence of this linker is extremely
similar to the amino acid sequences of the activation
loops of the insulin receptor and of TrkA (Figure 5).
59,62
For this reason, lidocaine at a dose of 40 lMinhibits
Figure 4. Inhibitory action of IPTRK3 against the activity of
various protein kinases.
1163
1488
1489
1490
1158
1162
672 682
676 680681
linker of
sodium channel
Activation loop of
TrkA
A
ctivation loop of
insulin receptor
kinase
= autophosphorylation sites = basic amino acid
= acidic amino acid = neutral amino acid
Figure 5. Amino acid sequence
similarities in the sodium channel,
insulin receptor, and TrkA.
TrkA and Pain 5
the tyrosine kinase activity of both insulin receptors and
TrkA.
59,62,63
As lidocaine toxicity occurs at a blood concentration
of 5 lg/mL (20 lM), intravenously administered lido-
caine does not reach a concentration that suppresses
TrkA activity. However, with local injection of lido-
caine (1% lidocaine is equivalent to approximately
40 mM) in nerve blocks, even if the injected dose
becomes less concentrated after diffusion into the nerve
fibers (1% lidocaine after diffusion in the vicinity of the
nerves achieves a concentration of approximately 100 to
400 lM), it is still considered to adequately suppress
TrkA activity. This suggests that local anesthetics used
in nerve blocks not only inhibit sodium channels, but
also potentially suppress TrkA activity. Figure 6 shows
molecular interactions between TrkA and ATP and also
between TrkA and lidocaine using UCSF Chimera
version 1.10.1.
64
Lidocaine probably inhibits tyrosine
kinase activity of TrkA by blocking ATP-binding site of
TrkA and may suppress neuropathic pain or mirror
image pain.
22,23,28
NGF/TRKA SIGNALING AND CANCER PAIN
Activation of NGF/TrkA signaling induces tumor pro-
gression, and either NGF or TrkA is a therapeutic target
against cancer.
65,66
Therefore, NGF/TrkA inhibitors are
expected to be useful for both cancer pain and tumori-
genesis.
45,66
TrkA inhibitor, which decreases proliferation of
melanoma cells, suppresses melanoma-induced cancer
pain in mice.
45
In bone metastasis model of prostate
carcinoma or sarcoma, however, neither anti-NGF
antibodies nor TrkA inhibitors, which suppress cancer
pain in rodents, showed any inhibitory effects on tumor
growth.
41,51,67
Further investigations are needed for
potential therapeutic strategies using NGF/TrkA inhibi-
tors to suppress tumorigenesis in addition to cancer
pain.
CONCLUSION
NGF, a factor that possesses physiological features
indispensable to the growth of sensory and sympathetic
nerves prenatally, becomes a chemical substance that
produces pain postnatally. If tissue injury is associated
with a prolonged inflammatory response or if the
damaged nerve does not regenerate into its original
state, pathological pain ensues. In such situations,
analgesics that suppress NGF/TrKA signaling might be
considered to be effective therapy. For instance, NGF/
TrkA inhibitors could be administered in the perioper-
ative period to prevent refractory chronic postoperative
pain following surgeries that are prone to cause periph-
eral nerve damage, such as thoracotomy (associated
with chronic post-thoracotomy pain syndrome) and
mastectomy (associated with chronic postmastectomy
pain syndrome). Moreover, NGF/TrkA inhibitors are
also candidate therapeutic agents for clinical use in the
treatment of chronic pain caused by osteoarthritis and
cancer pain.
REFERENCES
1. Abbott A. One hundred years of Rita. Nature.
2009;458:564567.
2. Levi-Montalcini R. Effects of mouse tumor transplan-
tation on the nervous system. Ann NY Acad Sci. 1952;55:330
343.
3. Levi-Montalcini R, Booker B. Destruction of the
sympathetic ganglia in mammals by an antiserum to a nerve
growth protein. Proc Natl Acad Sci USA. 1960;46:384391.
4. Angeletti RH, Bradshaw RA. Nerve growth factor
from mouse submaxillary gland: amino acid sequence. Proc
Natl Acad Sci USA. 1971;68:24172420.
5. Landreth GE, Shooter EM. Nerve growth factor
receptors on PC12 cells: ligand-induced conversion from
low- to high-affinity states. Proc Natl Acad Sci USA.
1980;77:47514755.
6. Shelton DL, Sutherland J, Gripp J, et al. Human trks:
molecular cloning, tissue distribution, and expression of
AB
Figure 6. Potential binding site of
lidocaine in TrkA. (A) Interaction
between TrkA and ATP. (B)
Interaction between TrkA and
lidocaine.
6HIROSE ET AL.
extracellular domain immunoadhesins. J Neurosci.
1995;15:477491.
7. Shibayama E, Koizumi H. Cellular localization of the
Trk neurotrophin receptor family in human non-neuronal
tissues. Am J Pathol. 1996;148:18071818.
8. Cunningham ME, Greene LA. A function-structure
model for NGF-activated TRK. EMBO J. 1998;17:7282
7293.
9. Wiesmann C, de Vos AM. Nerve growth factor:
structure and function. Cell Mol Life Sci. 2001;58:748759.
10. Delcroix J-D, Valletta JS, Wu C, Hunt SJ, Kowal AS,
Mobley WC. NGF signaling in sensory neurons: evidence that
early endosomes carry NGF retrograde signals. Neuron.
2003;39:6984.
11. Siggers DC. Nerve growth factor and some inherited
neurological conditions. Proc R Soc Med. 1976;69:183184.
12. Indo Y, Tsuruta M, Hayashida Y, et al. Mutations in
the TRKA/NGF receptor gene in patients with congenital
insensitivity to pain with anhidrosis. Nat Genet. 1996;13:485
488.
13. Miranda C, Di Virgilio M, Selleri S, et al. Novel
pathogenic mechanisms of congenital insensitivity to pain with
anhidrosis genetic disorder unveiled by functional analysis of
neurtrophic tyrosine receptor kinase type 1/nerve growth
factor receptor mutations. J Biol Chem. 2002;277:64556462.
14. Einarsdottir E, Carlsson A, Minde J, et al. A mutation
in the nerve growth factor beta gene (NGFB) causes loss of
pain perception. Human Mol Genet. 2004;8:799805.
15. Koltzenburg M. The changing sensitivity in the life of
the nociceptor. Pain. 1999;(Suppl. 6):S93S102.
16. Kromer LF. Nerve growth factor treatment after brain
injury prevents neuronal death. Science. 1987;235:214216.
17. Lewin GR, Ritter AM, Mendell LM. Nerve growth
factor induced hyperalgesia in the neonatal and adult rat. J
Neurosci. 1993;13:21362148.
18. Petty BG, Cornblath DR, Adornato BT, et al. The
effect of systemically administered recombinant human nerve
growth factor in healthy human subjects. Ann Neurol.
1994;36:244246.
19. Loeser JD, Treede RD. The Kyoto protocol of IASP
basic pain terminology. Pain. 2008;137:473477.
20. Thacker MA, Clark AK, Marchand F, McMahon SB.
Pathophysiology of peripheral neuropathic pain: immune cells
and molecules. Anesth Analg. 2007;105:838847.
21. Basbaum AI, Bautista DM, Scherrer G, Julius D.
Cellular and molecular mechanisms of pain. Cell.
2009;139:267284.
22. Wu C, Boustany L, Liang H, Brennan TJ. Nerve
growth factor expression after plantar incision in the rat.
Anesthesiology. 2007;107:128135.
23. Wu C, Erickson MA, Xu J, Wild KD, Brennan TJ.
Expression profile of nerve growth factor after muscle incision
in the rat. Anesthesiology. 2009;110:140149.
24. Woolf CJ, Costigan M. Transcriptional and posttrans-
lational plasticity and the generation of inflammatory pain.
Pro Natl Acad Sci USA. 1999;96:77237730.
25. Zhuang ZY, Xu H, Clapham DE, Ji RR. Phosphatidyli-
nositol 3-Kinase activates ERK in primary sensory neurons and
mediates inflammatory heat hyperalgesia through TRPV1
sensitization. J Neurosci. 2004;24:83008309.
26. Ueda H. Peripheral mechanisms of neuropathic pain
involvement of lysophosphatidic acid receptor-mediated
demyelination. Mol Pain. 2008;4:11.
27. Jancalek R. Signaling mechanisms in mirror image pain
pathogenesis. Ann Neurosci. 2011;18:123127.
28. Cheng CF, Cheng JK, Chen CY, et al. Mirror-image
pain is mediated by nerve growth factor produced from tumor
necrosis factor alpha-activated satellite glia after peripheral
nerve injury. Pain. 2014;155:906920.
29. Elzahaf RA, Tashani OA, Unsworth BA, Johnson MI.
The prevalence of chronic pain with an analysis of countries
with a human development index less than 0.9: a systematic
review without meta-analysis. Curr Med Res Opin.
2012;30:12211229.
30. Hefti FF, Rosenthal A, Walicke PA, et al. Novel class
of pain drugs based on antagonism of NGF. Trends Pharmacol
Sci. 2006;27:8591.
31. Watson JJ, Allen SJ, Dawbarn D. Targeting nerve
growth factor in pain. BioDrugs. 2008;22:349359.
32. Xian C, Zhou XF. Treating skeletal pain: limitations of
conventional anti-inflammatory drugs, and anti-neurotrophic
factor as a possible alternative. Nat Clin Pract Rheumatol.
2009;5:9298.
33. Urschel BA, Brown PN, Hulsebosch CE. Differential
effects on sensory nerve processes and behavioral alterations in
the rat after treatment with antibodies to nerve growth factor.
Exp Neurol. 1991;114:4452.
34. Lane NE, Schnitzer TJ, Birbara CA, et al. Tanezumab
for the treatment of pain from osteoarthritis of the knee. N
Engl J Med. 2010;363:15211531.
35. Garber K. Fate of novel painkiller mAbs hangs in
balance. Nat Biotechnol. 2011;29:173174.
36. McMahon SB, Bennett DL, Priestley JV, Shelton DL.
The biological effects of endogenous nerve growth factor on
adult sensory neurons revealed by a trkA-IgG fusion molecule.
Nat Med. 1995;1:774780.
37. Watson JJ, Fahey MS, van der Worm E, et al.
TrkAd5; a novel therapeutic agent for treatment of inflam-
matory pain and asthma. J Pharmacol Exp Ther.
2006;316:11221129.
38. Ugolini G, Marinelli S, Covaceuszach S, Cattaneo A,
Pavone F. The function neutralizing anti-TrkA antibody
MNAC13 reduces inflammatory and neuropathic pain. Pro
Natl Acad Sci USA. 2007;104:29852990.
39. Owolabi JB, Rizkalla G, Tehim A, et al. Characteriza-
tion of antiallodynic actions of ALE-0540, a novel nerve
growth factor receptor antagonist, in the rat. J Pharmacol Exp
Ther. 1999;289:12711276.
40. Colquhoun A, Lawrance GM, Shamovsky IL, Riopelle
RJ, Ross GM. Differential activity of the nerve growth factor
(NGF) antagonist PD90780 [7-(benzolylamino)-4,9-dihydro-
4-methyl-9-oxo-pyrazolo[5,1-b]quinazoline-2-carboxylic acid]
TrkA and Pain 7
suggests altered NGF-p75NTR interactions in the presence of
TrkA. J Pharmacol Exp Ther. 2004;310:505511.
41. Ghilardi JR, Freeman KT, Jimenez-Andrade JM, et al.
Administration of a tropomyosin receptor kinase inhibitor
attenuates sarcoma-induced nerve sprouting, neuroma forma-
tion and bone cancer pain. Mol Pain. 2010;6:87.
42. Hirose M, Takatori M, Kuroda Y, et al. Effect of
synthetic cell-penetrating peptide on TrkA activity in PC12
cells. J Pharmacol Sci. 2008;106:107113.
43. Ueda K, Hirose M, Murata E, et al. Local administra-
tion of a synthetic cell-penetrating peptide antagonizing TrkA
function suppresses inflammatory pain in rats. J Pharmacol
Sci. 2010;112:438443.
44. Ma W-Y, Murata E, Ueda K, et al. A synthetic
cell-penetrating peptide antagonizing TrkA function sup-
presses neuropathic pain in mice. J Pharmacol Sci.
2010;114:7984.
45. Tabata M, Murata E, Ueda K, Kuroda Y, Kato-Kogoe
N, Hirose M. Effects of TrkA inhibitory peptide, IPTRK3, on
cancer pain in a mouse melanoma model. J Anesth.
2012;26:545551.
46. Winston JH, Toma H, Shenoy M, et al. Acute pancre-
atitis results in referred mechanical hypersensitivity and
neuropeptide up-regulation that can be suppressed by the
protein kinase inhibitor K252a. J Pain. 2003;4:329337.
47. Sung B, Lim G, Mao J. Altered expression and uptake
activity of spinal glutamate transporters after nerve injury
contribute to the pathogenesis of neuropathic pain in rats. J
Neurosi. 2003;23:28992910.
48. Summer GJ, Puntillo KA, Miaskowski C, Dina OA,
Green PG, Levine JD. TrkA and PKC-epsilon in thermal burn-
induced mechanical hyperalgesia in the rat. J Pain.
2006;7:884891.
49. Ueda M, Hirose M, Takei N, et al. Nerve growth
factor induces systemic hyperalgesia after thoracic burn injury
in the rat. Neurosci Lett. 2002;328:97100.
50. http://www.fda.gov/downloads/AdvisoryCommittees/
CommitteesMeetingMaterials/Drugs/ArthritisAdvisoryCom-
mittee/UCM295205.pdf#search=‘Tanezumabarthritisadvisory
committeebriefingdocument’
51. Halvorson KG, Kubota K, Sevcik MA, et al. A block-
ing antibody to nerve growth factor attenuates skeletal pain
induced by prostate tumor cells growing in bone. Cancer Res.
2005;65:94269435.
52. Selvaraj D, Kuner R. Molecular players of tumor-nerve
interactions. Pain. 2015;156:67.
53. Majuta LA, Longo G, Fealk MN, McCaffrey G,
Mantyh PW. Orthopedic surgery and bone fracture pain are
both significantly attenuated by sustained blockade of nerve
growth factor. Pain. 2015;156:157165.
54. Nagahara H, Vocero-Akbani AM, Snyder EL, et al.
Transduction of full-length TAT fusion proteins into mam-
malian cells: TAT-p27
Kip1
induces cell migration. Nat Med.
1998;4:14491452.
55. Stachel SJ, Sanders JM, Henze DA, et al. Maximizing
diversity from a kinase screen: identification of novel and
selective pan-Trk inhibitors for chronic pain. J Med Chem.
2014;57:58005816.
56. Calissano P, Matrone C, Amadoro G. Nerve growth
factor as a paradigm of neurotrophins related to Alzheimer’s
disease. Dev Neurobiol. 2010;70:372383.
57. Matrone C, Barbagallo APM, La Rossa LR, et al. APP
is phosphorylated by TrkA and regulates NGF/TrkA signaling.
J Neurosci. 2011;31:1175611761.
58. Zhang Q, Descamps O, Hart MJ, et al. Paradoxical
effect of TrkA inhibition in Alzheimer’s disease Models. J
Alzheimers Dis. 2014;40:605617.
59. Takatori M, Kuroda Y, Hirose M. Local anesthetics
suppress nerve growth factor-mediated neurite outgrowth by
inhibition of tyrosine kinase activity of TrkA. Anesth Analg.
2006;102:462467.
60. Onizuka S, Shiraishi S, Tamura R, et al. Lidocaine
treatment during synapse reformation periods permanently
inhibits NGF-induced excitation in an identified reconstructed
synapse of Lymnaea stagnalis. J Anesth. 2012;26:4553.
61. Kuroda Y, Miyamoto K, Tanaka K, et al. Interactions
between local anesthetics and Na+channel inactivation gate
peptides in phosphatidylserine suspensions as studied by 1H-
NMR spectroscopy. Chem Pharm Bull. 2000;48:12931298.
62. Hirose M, Kuroda Y, Sawa S, et al. Suppression of
insulin signaling by a synthetic peptide KIFMK suggests the
cytoplasmic linker between DIII-S6 and DIV-S1 as a local
anaesthetic binding site on the sodium channel. Br J Pharma-
col. 2004;142:222228.
63. Hirose M, Martyn JAJ, Kuroda Y, Marunaka Y,
Tanaka Y. Mechanism of suppression of insulin signaling with
lignocaine. Br J Pharmacol. 2002;136:7680.
64. Pettersen EF, Goddard TD, Huang CC, et al. UCSF
Chimera-a visualization system for exploratory research and
analysis. J Comput Chem. 2004;25:16051612.
65. Nakagawa A. Trk receptor tyrosine kinases: a bridge
between cancer and neural development. Cancer Lett.
2001;169:107114.
66. Wang W, Chen J, Guo X. The role of nerve growth
factor and its receptors in tumorigenesis and cancer pain.
Biosci Trends. 2014;8:6874.
67. Mantyh WG, Jimenez-Andrade JM, Stake JI, et al.
Blockade of nerve sprouting and neuroma formation markedly
attenuates the development of late stage cancer pain. Neuro-
science. 2010;171:588598.
8HIROSE ET AL.
... The NGF secreted by resident and inflammatory cells and keratinocytes in injured tissue acts via TrkA receptors present on primary sensory nerve endings and participates in peripheral sensitization [4]. Hence, NGF/TrkA signaling is considered as a primary target for the development of pain therapeutics [5] leading to the development of anti-NGF antibodies and TrkA antagonists [6,7]. However, for these analgesics to be effective with the least possible side effects, the detailed mechanisms involved in peripheral sensitization need to be evaluated further. ...
Preprint
Full-text available
Glutamate functions as the major excitatory neurotransmitter for primary sensory neurons and has a crucial role in sensitizing peripheral nociceptor terminals producing sensitization. Glutaminase (GLS) is the synthetic enzyme that converts glutamine to glutamate. GLS-immunoreactivity (-ir) and enzyme activity are elevated in dorsal root ganglion (DRG) neuronal cell bodies during chronic peripheral inflammation, but the mechanism for this GLS elevation is yet to be fully characterized. It has been well established that, after nerve growth factor (NGF) binds to its high-affinity receptor tropomyosin receptor kinase A (TrkA), a retrograde signaling endosome is formed. This endosome contains the late endosomal marker Rab7GTPase and is retrogradely transported via axons to the cell soma located in DRG. This complex is responsible for regulating the transcription of several critical nociceptive genes. Here, we show that this retrograde NGF signaling mediates the expression of GLS in DRG neurons during the process of peripheral inflammation. We disrupted the normal NGF/TrkA signaling in adjuvant-induced arthritic (AIA) Sprague Dawley rats by pharmacological inhibition of TrkA or blockade of Rab7GTPase, which significantly attenuated the expression of GLS in DRG cell bodies. These results indicate that NGF/TrkA signaling is crucial for the production of glutamate and has a vital role in the development of neurogenic inflammation. In addition, our pain behavioural data suggest that Rab7GTPase can be a potential target for attenuating peripheral inflammatory pain.
... 54 TrkA, which are high-affinity-NGF receptors, are found on nociceptors and sympathetic efferent fibers, 83 and signaling through the NGF/TrkA pathway has been a target for treatment of pain. 84 Transdermal ES after NGF administration to the hindlimbs of pigs resulted in a hyperalgesia, which was increased with higher-frequency stimulation Most post-ganglionic axons exit the sympathetic ganglia (SG) through the gray rami and into the ventral and dorsal rami of the spinal nerves to innervate distal targets. Some will send projections to their adjacent DRG (sensory neurons in blue) to reach blood vessels and the surface of the DRG. ...
Article
Full-text available
Peripheral nerve injuries (PNIs) are common and devastating. The current standard of care relies on the slow and inefficient process of nerve regeneration after surgical intervention. Electrical stimulation (ES) has been shown to both experimentally and clinically result in improved regeneration and functional recovery after PNI for motor and sensory neurons; however, its effects on sympathetic regeneration have never been studied. Sympathetic neurons are responsible for a myriad of homeostatic processes that include, but are not limited to, blood pressure, immune response, sweating, and the structural integrity of the neuromuscular junction. Almost one quarter of the axons in the sciatic nerve are from sympathetic neurons, and their importance in bodily homeostasis and the pathogenesis of neuropathic pain should not be underestimated. Therefore, as ES continues to make its way into patient care, it is not only important to understand its impact on all neuron subtypes, but also to ensure that potential adverse effects are minimized. This piece gives an overview of the effects of ES in animals models and in humans while offering a perspective on the potential effects of ES on sympathetic axon regeneration.
... On the other hand, NGF receptors can activate the immune response and promote the release of inflammatory cytokines, such as interleukin and TNF, which then act on nerve fiber endings. 34,35 In addition, in recent years, studies have directly pointed out that intestinal hypersensitivity induced by NGF is related to synaptic plasticity. A previous study found that colonic synaptic density, SYN, and PSD-95 expression were increased in rats with intestinal hypersensitivity induced by water avoidance stress. ...
Article
Full-text available
Purpose Diarrhea-predominant irritable bowel syndrome (D-IBS) is a frequent functional gastrointestinal disease that affects health and quality of life owing to its high incidence and recurrence rate. Tongxie-Yaofang (TXYF) is a traditional Chinese medicine prescribed for D-IBS. However, the therapeutic mechanism of TXYF has not been fully elucidated. This study aimed to investigate the effects of TXYF on visceral hypersensitivity in stress-induced D-IBS rats and the underlying mechanisms. Methods Electromyographic (EMG) activity of the external oblique muscles and the abdominal withdrawal reflex (AWR) score captured by Barostat were used to quantify the effect of TXYF on visceral sensitivity. Transmission electron microscopy (TEM) was used to observe the ultrastructure of the enteric nervous system (ENS). For molecular detection, the colonic expression of enteric glial cell’s (EGC’s) activation markers, glial fibrillary acidic protein (GFAP) and calcium-binding protein S100β, NGF, TrkA, synaptic plasticity-related factors, synaptophysin (SYN) and postsynaptic density-95 (PSD-95), glutamate, glutamate receptors α-amino-3-hydroxy-5-methyl-4-isoxazole-propionic acid receptor (AMPAR), and N-methyl-D-aspartate receptor (NMDAR) were detected by immunohistochemistry, enzyme-linked immunosorbent assay, and real-time PCR. An ex vivo experiment was conducted to measure the EGC-induced NGF release. Results TXYF decreased the EMG activity and AWR scores in rats with D-IBS. Under TEM, TXYF improved the dense and irregular nerve arrangement, narrowed the synaptic cleft, and decreased the number of synaptic vesicles in D-IBS rats. In addition, TXYF decreased the expression of GFAP, S100β, SYN, and PSD-95; down-regulated the levels of NGF, TrkA, and glutamate; and reduced the mRNA expression of AMPAR1, NMDAR1, and NMDAR2B. In an ex vivo experiment, TXYF decreased NGF release in D-IBS rats, and this trend disappeared under EGC inhibition. Conclusion TXYF alleviated visceral hypersensitivity in D-IBS rats possibly by improving synaptic plasticity through inhibiting the activity of EGCs and the NGF/TrkA signaling pathway in the colon.
... The PAR2, Tyrosine kinase-type receptor (Trk), and multiple signal transduction pathways are involved in the process (Ji et al., 2002;Kim et al., 2014). One emerging target for treating intractable pain is the NGF and its receptor Tyrosine kinase-type receptor A (TrkA), which has a high affinity for NGF (Hirose et al., 2016). Additionally, the combination of NGF and TrkA might sensitize and improve TRPV1 expression (Donnerer et al., 2005). ...
Article
Full-text available
Irritable Bowel Syndrome (IBS) is a complex functional gastrointestinal disorder primarily characterized by chronic abdominal pain, bloating, and altered bowel habits. Chronic abdominal pain caused by visceral Hypersensitivity (VH) is the main reason why patients with IBS seek medication. Significant research effort has been devoted to the efficacy of acupuncture as a non-drug alternative therapy for visceral-hyperalgesia-induced IBS. Herein, we examined the central and peripheral analgesic mechanisms of acupuncture in IBS treatment. Acupuncture can improve inflammation and relieve pain by reducing 5-hydroxytryptamine and 5-HT3A receptor expression and increasing 5-HT4 receptor expression in peripheral intestinal sensory endings. Moreover, acupuncture can also activate the transient receptor potential vanillin 1 channel, block the activity of intestinal glial cells, and reduce the secretion of local pain-related neurotransmitters, thereby weakening peripheral sensitization. Moreover, by inhibiting the activation of N-methyl-D-aspartate receptor ion channels in the dorsal horn of the spinal cord and anterior cingulate cortex or releasing opioids, acupuncture can block excessive stimulation of abnormal pain signals in the brain and spinal cord. It can also stimulate glial cells (through the P2X7 and prokinetic protein pathways) to block VH pain perception and cognition. Furthermore, acupuncture can regulate the emotional components of IBS by targeting hypothalamic-pituitary-adrenal axis-related hormones and neurotransmitters via relevant brain nuclei, hence improving the IBS-induced VH response. These findings provide a scientific basis for acupuncture as an effective clinical adjuvant therapy for IBS pain.
Article
Full-text available
Glutamate functions as the major excitatory neurotransmitter for primary sensory neurons and has a crucial role in sensitizing peripheral nociceptor terminals producing sensitization. Glutaminase (GLS) is the synthetic enzyme that converts glutamine to glutamate. GLS-immunoreactivity (-ir) and enzyme activity are elevated in dorsal root ganglion (DRG) neuronal cell bodies during chronic peripheral inflammation, but the mechanism for this GLS elevation is yet to be fully characterized. It has been well established that, after nerve growth factor (NGF) binds to its high-affinity receptor tropomyosin receptor kinase A (TrkA), a retrograde signaling endosome is formed. This endosome contains the late endosomal marker Rab7GTPase and is retrogradely transported via axons to the cell soma located in the DRG. This complex is responsible for regulating the transcription of several critical nociceptive genes. Here, we show that this retrograde NGF signaling mediates the expression of GLS in DRG neurons during the process of peripheral inflammation. We disrupted the normal NGF/TrkA signaling in adjuvant-induced arthritic (AIA) Sprague Dawley rats by the pharmacological inhibition of TrkA or blockade of Rab7GTPase, which significantly attenuated the expression of GLS in DRG cell bodies. The results indicate that NGF/TrkA signaling is crucial for the production of glutamate and has a vital role in the development of neurogenic inflammation. In addition, our pain behavioral data suggest that Rab7GTPase can be a potential target for attenuating peripheral inflammatory pain.
Article
Full-text available
Pancreatic ductal adenocarcinoma is one of the most lethal solid malignancies, characterized by its aggressiveness and metastatic potential, with a 5-year survival rate of only 13%. Progress in the management of metastatic disease has been modest. A robust connection between nervous system and tumor progression exists, with prominent neural alterations having been observed during pancreatic cancer’s progression, including neural hypertrophy, neural density, and neural remodeling. The pancreatic tumor microenvironment includes s set of cells and structures that constantly dialogue with cancer cells, influencing its growth and behavior. The microglia is key cellular components of the tumor microenvironment, and Schwann cells are the principal glial cells in the peripheral neural system. Schwann cells can regulate changes in the tumor microenvironment and immune responses by secreting a variety of factors and can support a tumor’s invasion of nerves and distant metastasis, with further pain exacerbation. Schwann cells secrete various pain-related molecules, such as the neural growth factor, to mediate the activation of primary sensory neurons, leading to pain induction. The binding of the neural growth factor to tropomyosin receptor kinase A is an important signaling mechanism for pain perception in humans. Consequently, directing efforts towards targeting neural invasion may provide an alternative strategy to improve the prognosis of and alleviate pain in patients with pancreatic cancer.
Article
Nerve growth factor ( NGF ) and its receptor, tropomyosin receptor kinase A ( TrkA ), are known to play important roles in the immune and nervous system. However, the effects of NGF on the osteogenic differentiation of dental pulp stem cells ( DPSCs ) remain unclear. This study aimed to investigate the role of NGF on the osteogenic differentiation of DPSCs in vitro and the underlying mechanisms. DPSCs were cultured in osteogenic differentiation medium containing NGF (50 ng/ mL ) for 7 days. Then osteogenic‐related genes and protein markers were analysed using qRT‐PCR and Western blot, respectively. Furthermore, addition of NGF inhibitor and small interfering RNA ( siRNA ) transfection experiments were used to elucidate the molecular signalling pathway responsible for the process. NGF increased osteogenic differentiation of DPSCs significantly compared with DPSCs cultured in an osteogenic‐inducing medium. The NGF inhibitor Ro 08‐2750 ( 10 μM ) and siRNA ‐mediated gene silencing of NGF receptor, TrkA and ERK signalling pathways inhibitor U0126 ( 10 μM ) suppressed osteogenic‐related genes and protein markers on DPSCs . Furthermore, our data revealed that NGF ‐upregulated osteogenic differentiation of DPSCs may be associated with the activation of MEK / ERK signalling pathways via TrkA . Collectively, NGF was capable of promoting osteogenic differentiation of DPSCs through MEK / ERK signalling pathways, which may enhance the DPSCs ‐mediated bone tissue regeneration.
Article
Full-text available
The progressive worsening of disc degeneration and related nonspecific back pain are prominent clinical issues that cause a tremendous economic burden. Activation of reactive oxygen species (ROS) related inflammation is a primary pathophysiologic change in degenerative disc lesions. This pathological state is associated with M1 macrophages, apoptosis of nucleus pulposus cells (NPC), and the ingrowth of pain-related sensory nerves. To address the pathological issues of disc degeneration and discogenic pain, we developed MnO2@TMNP, a nanomaterial that encapsulated MnO2 nanoparticles with a TrkA-overexpressed macrophage cell membrane (TMNP). Consequently, this engineered nanomaterial showed high efficiency in binding various inflammatory factors and nerve growth factors, which inhibited inflammation-induced NPC apoptosis, matrix degradation, and nerve ingrowth. Furthermore, the macrophage cell membrane provided specific targeting to macrophages for the delivery of MnO2 nanoparticles. MnO2 nanoparticles in macrophages effectively scavenged intracellular ROS and prevented M1 polarization. Supportively, we found that MnO2@TMNP prevented disc inflammation and promoted matrix regeneration, leading to downregulated disc degenerative grades in the rat injured disc model. Both mechanical and thermal hyperalgesia were alleviated by MnO2@TMNP, which was attributed to the reduced calcitonin gene-related peptide (CGRP) and substance P expression in the dorsal root ganglion and the downregulated Glial Fibrillary Acidic Protein (GFAP) and Fos Proto-Oncogene (c-FOS) signaling in the spinal cord. We confirmed that the MnO2@TMNP nanomaterial alleviated the inflammatory immune microenvironment of intervertebral discs and the progression of disc degeneration, resulting in relieved discogenic pain.
Article
Full-text available
Citation: Terracina, S.; Ferraguti, G.; Tarani, L.; Fanfarillo, F.; Tirassa, P.; Ralli, M.; Iannella, G.; Polimeni, A.; Lucarelli, M.; Greco, A.; et al. Nerve Growth Factor and Autoimmune Diseases. Curr. Issues Mol. Biol. 2023, 45, 8950-8973. https://doi. Abstract: NGF plays a crucial immunomodulatory role and increased levels are found in numerous tissues during autoimmune states. NGF directly modulates innate and adaptive immune responses of B and T cells and causes the release of neuropeptides and neurotransmitters controlling the immune system activation in inflamed tissues. Evidence suggests that NGF is involved in the pathogenesis of numerous immune diseases including autoimmune thyroiditis, chronic arthritis, multiple sclerosis, systemic lupus erythematosus, mastocytosis, and chronic granulomatous disease. Furthermore, as NGF levels have been linked to disease severity, it could be considered an optimal early biomarker to identify therapeutic approach efficacy. In conclusion, by gaining insights into how these molecules function and which cells they interact with, future studies can devise targeted therapies to address various neurological, immunological, and other disorders more effectively. This knowledge may pave the way for innovative treatments based on NGF manipulation aimed at improving the quality of life for individuals affected by diseases involving neurotrophins.
Article
Chronic low back pain mainly attributed to intervertebral disc (IVD) degeneration. Endogenous damage-associated molecular patterns (DAMPs) in the injured IVD, particularly mitochondria-derived nucleic acid molecules (CpG DNA), play a primary role in the inflammatory responses in macrophages. M1-type macrophages form a chronic inflammatory microenvironment by releasing pro-inflammatory factors and nerve growth factor (NGF) that induce nerve growth into the inner annulus fibrosus, resulting in persistent hyperalgesia. We fabricated an amphiphilic polycarbonate that naturally forms cationic nanoparticles (cNP) in aqueous solutions, with the hydrophobic core loaded with TrkA-IN-1, an antagonist against the NGF receptor (TrkA). The drug delivery nanoparticles were denoted as TI-cNP. TrkA-IN-1 and TI-cNP were added to the decellularized annulus fibrosus matrix (DAF) hydrogel to form hybrid hydrogels, denoted as TI-DAF and TI-cNP-DAF, respectively. As a result, TrkA-IN-1 showed a delayed release profile both in TI-DAF and TI-cNP-DAF. Each mole of cNP could bind approximately 3 mol of CpG DNA to inhibit inflammation. cNP-DAF and TI-cNP-DAF significantly inhibited the M1 phenotype induced by CpG DNA. TI-DAF and TI-cNP-DAF reduced neurite branching and axon length, and inhibited the expression of neurogenic mediators (CGRP and substance P) in the presence of NGF. Besides, TI-cNP-DAF relieved mechanical hyperalgesia, reduced CGRP and substance P expression in the dorsal root ganglion, and downregulated GFAP and c-FOS signaling in the spinal cord in the rat disc herniation model. Summarily, TI-cNP-DAF, a novel composite IVD hydrogel, efficiently mediated the inflammatory environment, inhibited nerve ingrowth and sensitization, and could be clinically applied for treating discogenic pain. STATEMENT OF SIGNIFICANCE: : Discogenic lower back pain, related to intervertebral disc degeneration (IDD), imposes a tremendous health and economic burden globally. M1-type macrophages release pro-inflammatory factors and nerve growth factor (NGF) that induce nerve growth into the inner annulus fibrosus, resulting in persistent hyperalgesia and discogenic pain. Reconstructing matrix integrity and modulating the inflammatory microenvironment are promising strategies for preventing the ingrowth and activation of neurites. The TI-cNP-DAF hydrogel recovers tissue integrity, alleviates inflammation, and delivers the TrkA antagonist to inhibit the activity of NGF, thus restraining hyperinnervation and nociceptive input. Due to its simple production process, injectability, and acellular strategy, the hydrogel is operable and holds great potential for treating discogenic lower back pain.
Article
Full-text available
It is now clear that a peripheral nerve lesion affects contralateral non-lesioned structures, and thus such a lesion can result in mirror image pain. The pathogenesis is still not exactly known, but there are some possible signaling pathways in the contralateral reaction of the nerve tissue after unilateral nerve injury. Potential signaling pathways of contralateral changes can be generally divided into humoral and neuronal mechanisms. Damage to peripheral nerves or spinal roots produces a number of breakdown products with development of an aseptic inflammatory reaction. Released immunomodulatory cytokines are believed to be transported via blood or cerebrospinal fluid into the contralateral part of the body affecting spinal roots, dorsal root ganglia or peripheral nerves. Because neurons are elements of a highly organized network, injury to the peripheral neuron results in signals that travel transneuronally into the central nervous system and affects the contralateral homonymous neurons. There is also evidence that spinal glia creates and maintain pathological pain. Additionally, there may be compensatory changes in behavior of animals with an impact on contralateral neurons, such as altered stance and motor performance or autonomic reflex changes. Although the transneuronal signaling pathway appears to be plausible, the humoral signaling pathway or other communication systems cannot be excluded at this time. Knowledge about these processes has clinical implications for the understanding of chronic neuropathic pain states, and, therefore, further studies will be necessary. Understanding signaling mechanisms in mirror image pain pathogenesis may provide novel therapeutic targets for the management of neuropathic pain.
Article
Full-text available
An unbiased screen for compounds that block amyloid-β protein precursor (AβPP) caspase cleavage identified ADDN-1351, which reduced AβPP-C31 by 90%. Target identification studies showed that ADDN-1351 is a TrkA inhibitor, and, in complementary studies, TrkA overexpression increased AβPP-C31 and cell death. TrkA was shown to interact with AβPP and suppress AβPP-mediated transcriptional activation. Moreover, treatment of PDAPP transgenic mice with the known TrkA inhibitor GW441756 increased sAβPPα and the sAβPPα to Aβ ratio. These results suggest TrkA inhibition-rather than NGF activation-as a novel therapeutic approach, and raise the possibility that such an approach may counteract the hyperactive signaling resulting from the accumulation of active NGF-TrkA complexes due to reduced retrograde transport. The results also suggest that one component of an optimal therapy for Alzheimer's disease may be a TrkA inhibitor.
Article
The number of patients suffering from postoperative pain due to orthopedic surgery and bone fracture is projected to dramatically increase because the human life span, weight, and involvement in high-activity sports continue to rise worldwide. Joint replacement or bone fracture frequently results in skeletal pain that needs to be adequately controlled for the patient to fully participate in needed physical rehabilitation. Currently, the 2 major therapies used to control skeletal pain are nonsteroidal anti-inflammatory drugs and opiates, both of which have significant unwanted side effects. To assess the efficacy of novel therapies, mouse models of orthopedic and fracture pain were developed and evaluated here. These models, orthopedic surgery pain and bone fracture pain, resulted in skeletal pain-related behaviors that lasted 3 weeks and 8 to 10 weeks, respectively. These skeletal pain behaviors included spontaneous and palpation-induced nocifensive behaviors, dynamic weight bearing, limb use, and voluntary mechanical loading of the injured hind limb. Administration of anti-nerve growth factor before orthopedic surgery or after bone fracture attenuated skeletal pain behaviors by 40% to 70% depending on the end point being assessed. These data suggest that nerve growth factor is involved in driving pain due to orthopedic surgery or bone fracture. These animal models may be useful in developing an understanding of the mechanisms that drive postoperative orthopedic and bone fracture pain and the development of novel therapies to treat these skeletal pains.
Article
We have identified several series of small molecule inhibitors of TrkA with unique binding modes. The starting leads were chosen to maximize the structural and binding mode diversity derived from a high throughput screen of our internal compound collection. These leads were optimized for potency and selectivity employing a structure based drug design approach adhering to the principles of ligand efficiency to maximize binding affinity without overly relying on lipophilic interactions. This endeavor resulted in the identification of several small molecule pan-Trk inhibitors series that exhibit high selectivity for TrkA/B/C versus a diverse panel of kinases. We have also demonstrated efficacy in both inflammatory and neuropathic pain models upon oral dosing. Herein we describe the identification process, hit-to-lead progression, and binding profiles of these selective pan-Trk kinase inhibitors.
Article
The nerve growth factor (NGF) is a growth factor that belongs to the neurotrophin family. NGF has two structurally different receptors, the p75 neurotrophin receptor (p75NTR) and the tropomyosin-related kinase A (TrkA). Interaction of NGF with its receptors regulates a variety of physiological processes of neuronal system. Recent studies have shown that NGF and its receptors were involved in the regulation of tumourigenesis by either supporting or suppressing tumor growth depending on the tumor types. This review summarizes the current views of NGF and its receptors in tumorigenesis and cancer pain.