ArticlePDF AvailableLiterature Review

Mechanism of electroacupuncture on inflammatory pain: neural-immune-endocrine interactions

Authors:
  • Dana-Farber Cancer Institute/Harvard Medical School

Abstract and Figures

Nociceptive signals are transmitted by peripheral afferents to the central nervous system under pain condition, a process that involves various neurotransmitters and pathways. Electroacupuncture (EA) has been widely used as a pain management technique in clinical practice. Emerging studies have shown that EA can inhibit the induction and transmission of pain signals and, consequently, mediate anti-nociceptive and anti-inflammatory effects by rebalancing the neural-immune-endocrine interactions. This review summarizes the neural-immune-endocrine circuit including peripheral afferent and central efferent, contributing to EA-induced neuroimmune and neuroendocrine modulation in inflammatory pain models. The peripheral afferent circuit includes crosstalk among immune cells, inflammatory cytokines, peripheral nociceptors. In central efferent primarily involves the neuroinflammatory interactions between spinal nociceptive neurons and glial cells. Furthermore, the hypothalamic-pituitary-adrenal axis, sympathetic and vagal nervous may serve as an essential pathway involved in the mechanism of acupuncture-mediated analgesia within the interactions of the central, immune and endocrine systems. Overall, this review focuses on the interactions of neural-immune-endocrine in inflammatory pain, which may be underlying the mechanism of EA-induced anti-inflammatory and antinociceptive effect.
Content may be subject to copyright.
TOPIC
JTCM
|
www. journaltcm. com October 15, 2019
|
Volume 39
|
Issue 5
|
Online Submissions: http://ww w.journaltcm.com J Tradit Chin Med 2019 October 15; 39(5): 740-749
info@journaltcm.com ISSN 0255-2922
© 2019 JTCM. All rights reserved.
REVIEW
Mechanism of electroacupuncture on inflammatory pain: neural-im-
mune-endocrine interactions
Li Yuan, Yang Mingxiao, Wu Fan, Cheng Ke, Chen Haiyong, Shen Xueyong, Lao Lixing
aa
Li Yuan, Wu Fan, Cheng Ke, Shen Xueyong, Lao Lixing,
School of Acupuncture Moxibustion and Tuina, Shanghai
University of Traditional Chinese Medicine, Shanghai
201203, China
Yang Mingxiao, Chen Haiyong, Lao Lixing, School of Chi-
nese Medicine, The University of Hong Kong, Hong Kong,
China
Supported by the National Natural Science Foundation of
China (No. 81320108028, Efficacy and Neurobiological
Mechanisms of Traditional Moxibustion-based Laser Moxi-
bustion with Specific Wavelength on Inflammatory Pain),
the Three-Year Development Plan Project for Traditional Chi-
nese Medicine of Shanghai Municipal Health Commission
[ZY (2018-2020)-CCCX-2001-05], the National Basic Research
Program of China (973 Program, No. 2015CB554505), Shang-
hai Key Laboratory of acupuncture mechanism and acu-
point function (No. 14DZ2260500), Shanghai University of
Traditional Chinese Medicine budget research project
(No.18LK010), and Li Yuan's postgraduate innovation ability
project (No. A1-N192050102040105)
Correspondence to: Prof. Shen Xueyong, School of Acu-
puncture Moxibustion and Tuina, Shanghai University of Tra-
ditional Chinese Medicine, Shanghai 201203, China. snow-
ysh@hotmail.com; Prof. Lao Lixing, School of Chinese Medi-
cine, The University of Hong Kong, Hong Kong, China;
School of Acupuncture Moxibustion and Tuina, Shanghai
University of Traditional Chinese Medicine, Shanghai
201203, China. lxlao1@hku.hk
Telephone: +86-21-51322178;+852-39176476
Accepted: January 9, 2019
Abstract
Nociceptive signals are transmitted by peripheral
afferents to the central nervous system under pain
condition, a process that involves various neu-
rotransmitters and pathways. Electroacupuncture
(EA) has been widely used as a pain management
technique in clinical practice. Emerging studies
have shown that EA can inhibit the induction and
transmission of pain signals and, consequently, me-
diate anti-nociceptive and anti-inflammatory ef-
fects by rebalancing the neural-immune-endocrine
interactions. This review summarizes the neural-im-
mune-endocrine circuit including peripheral affer-
ent and central efferent, contributing to EA-in-
duced neuroimmune and neuroendocrine modula-
tion in inflammatory pain models. The peripheral
afferent circuit includes crosstalk among immune
cells, inflammatory cytokines, peripheral nocicep-
tors. In central efferent primarily involves the neuro-
inflammatory interactions between spinal nocicep-
tive neurons and glial cells. Furthermore, the hypo-
thalamic-pituitary-adrenal axis, sympathetic and va-
gal nervous may serve as an essential pathway in-
volved in the mechanism of acupuncture-mediated
analgesia within the interactions of the central, im-
mune and endocrine systems. Overall, this review
focuses on the interactions of neural-immune-en-
docrine in inflammatory pain, which may be under-
lying the mechanism of EA-induced anti-inflamma-
tory and antinociceptive effect.
© 2019 JTCM. All rights reserved.
Keywords: Electroacupuncture; Analgesia; Inflam-
matory pain; Neural-immune-endocrine interac-
tions; Review
INTRODUCTION
Inflammatory pain, which is caused by the activation
of peripheral nociceptors in the nervous system by nox-
ious chemicals and mechanical or thermal stimuli, is
not only a warning signal for local tissue damage and
nerve injury but can also be an indicator of systemic ill-
740
Li Y et al. / Review
JTCM
|
www. journaltcm. com October 15, 2019
|
Volume 39
|
Issue 5
|
ness.1Physiologically, nociceptive signals that are medi-
ated by central neural circuits lead to the perception of
pain. Pain perceived by the brain can have regulatory
effects that maintain the homeostasis of the immune,
autonomic and endocrine systems. The dysregulation
of these bidirectional signaling pathways results in
chronic inflammatory pain.2,3 During this process, pe-
ripheral tissue damage induces inflammatory responses
by triggering immune cells to release a series of inflam-
matory mediators. These mediators bind with recep-
tors expressed on nociceptive neurons, causing the neu-
rons to depolarize and generate action potentials,
which further transmit the nociceptive signals to the
spinal dorsal horn and the brain, contributing to the in-
duction and maintenance of perceived pain.4More-
over, peripheral inflammatory mediators, such as in-
flammatory cytokines, can be activated by vagal and
sympathetic nerves, which can aggravate pain and in-
flammation through cross-talk between the neuroim-
mune and neuroendocrine systems.3, 5
Electroacupuncture (EA), as a complementary and al-
ternative therapy, has been shown to have substantial
beneficial effects on pain relieving.6Though the mecha-
nisms of action underlying these effects remain to be
elucidated, EA may exert a comprehensive modulatory
effect on inflammatory pain, affecting the entire ner-
vous system [including the peripheral nervous system
(PNS) and the central nervous system (CNS)], the im-
mune system, and the endocrine system. Due to the
complexity of pain, a more comprehensive elucidate
the mechanisms underlying the EA-mediated regula-
tion of inflammatory pain within the interactions of
central, immune and endocrine systems is of great clini-
cal significance. This review summarizes the recent
progress towards understanding this process and pro-
poses a mechanism for the multisystem regulatory ef-
fects of EA during inflammatory pain.
Anatomical structure of acupuncture points
Acupoints can be sensed and transduced by a variety of
stimulation types, such as mechanical stimulation me-
diated by acupuncture needle, thermal stimulation by
moxibustion, electrical stimulation by EA, and radia-
tion by laser beams. The ability of acupoints to trans-
duce stimuli plays an important role in acupunc-
ture-mediated effects on pain.7,8 According to classical
acupuncture theories, disorders of the visceral organs
can be manifested at specific points, either on or under-
neath the skin surface, which is how acupoints have his-
torically been defined.9Despite considerable efforts to
better understand the anatomy and physiology of acu-
puncture points and meridians, the definitions and
characterizations of these structures remain inconclu-
sive. Early morphology studies analyzed the anatomical
structures surrounding acupoints, suggesting that ner-
vous system components, blood vessels, and musculosk-
eletal tissues represent important constituents of acu-
points.10-12 The anatomical structures surrounding acu-
puncture points transmit the neuroimmune and neuro-
inflammation signals induced by stimulation with EA
or manual acupuncture.13 Recent studies have shown
that mast cells (MCs), which are important compo-
nents of the immune system, could be potential effec-
tor cells at acupoints.14,15 EA stimulation at acupoints
has been shown to result in MC degranulation and af-
ferent nerve excitation,16 which is closely related to acu-
point sensitization17 and EA-mediated analgesia.15 Fol-
lowing acupuncture stimulation, MCs not only initiate
a neuroimmune response by releasing inflammatory
mediators but also play a crucial role in the cross-talk
among the circulatory, nervous and immune networks
at acupoints.16,18 A recent study showed that transcuta-
neous electrical acupoint stimulation at Binao (LI 14)
acupoint induced sensory nerve fibers to express calci-
tonin gene-related peptide and substance P, which bind
with the neurokinin 1 receptor on MCs. This interac-
tion resulted in the degranulation of MCs, releasing
5-hydroxytryptamine (5-HT), and thus producing an-
algesic effect similar to that of EA.19 Other studies have
also demonstrated that electrical stimulation at the Zu-
sanli (ST 36) acupoint activates the immune system by
regulating the production of immune cytokines, such
as interferon gamma (IFN-γ), interleukin-2 (IL-2) and
interleukin-17 (IL-17), and the differentiation and acti-
vation of splenic T cells.20
Peripheral afferent neuroimmune and
neuroendocrine modulation
Peripheral nociceptive afferent fibers include Aδ- and
C-fibers, which have peripheral axonal branches at no-
ciceptor terminals. These fibers are found in the dorsal
root ganglion (DRG), the trigeminal ganglion, the no-
dose ganglion of the vagal nerve, and the central axonal
branch, all of which form synaptic connections with
neurons in the spinal dorsal horn (SDH).4The nocicep-
tive signals induced by chemical, mechanical, or ther-
mal stimuli leading to changes in the responsiveness of
peripheral nociceptors and peripheral sensitization in-
cluding hyperalgesia and allodynic.
The neural mechanism underlying acupuncture/EA-
mediated analgesia has been addressed previously and
includes a wide spectrum of modulating effects mediat-
ed by peripheral afferent pathway, transmitters and
modulators.10,21 A large number of studies have ex-
plored the distinct roles played by the non-nociceptive
large afferent fiber (Aβ-fiber) and the nociceptive small
afferent fibers (Aδ- and C-fibers), which are found in
the peripheral neural pathways, during EA-induced an-
ti-nociceptive signaling.22 The transient receptor poten-
tial vanilloid receptors 1 (TRPV1) and 4 (TRPV4),
and the acid-sensing ion channel 3 (ASIC3) are mecha-
nosensitive channels associated with the release of ade-
nosine triphosphate (ATP) in various local tissues.23-25
Structurally, they are membrane channel proteins that
become permeable to cations, such as sodium and calci-
um, following mechanical stimulation. ASIC3, which
741
JTCM
|
www. journaltcm. com October 15, 2019
|
Volume 39
|
Issue 5
|
Li Y et al. / Review
mediates responses to acidic and mechanical stimuli, is
located primarily in the Aβ-fibers that innervate the
skin and muscles.26 TRPV1 belongs to the TRPV sub-
family and is highly expressed in nociceptive Aδ- and
C-fibers.27 Previous studies have shown that the ASIC3
and TRPV1 receptors are involved in the peripheral
sensations of EA stimulation, based on their distribu-
tions and functional properties.28 The segmental analge-
sic effects of low-intensity EA are mediated by afferent
Aβ-fibers and ASIC3, and the systemic analgesic ef-
fects of high-intensity EA are attributed to the activa-
tion of Aδ/C-fibers and TRPV1. Other studies have
found that TRPV1 is highly expressed at Zusanli (ST
36), which suggests that TRPV1 might act as an acu-
puncture-responding channel that senses the physical
stimulation of acupuncture, conducts the signal to
nerve terminals through ATP-induced calcium wave
propagation (CWP),29 and mediates the transduction
of EA signals to the CNS. TRPV1 receptors are also ex-
pressed in subepidermal connective tissue cells, where
they may play a role in the effects of EA on local tis-
sue.30 TRPV1, as a mechanical sensitive channel, plays
an important role in the transmission of the physical
stimulation caused by acupuncture or EA to neurologi-
cal signaling in nervous system. In addition, the activa-
tion of TRPV1 receptors upregulates the expression of
protein kinases and related downstream molecules,
such as protein kinase A (PKA), protein kinase C
(PKC), and Phosphoinositide 3-kinase (PI3K)-protein
kinase B (PKB/Akt) signaling pathway, during com-
plete Freund's adjuvant (CFA)-induced inflammatory
pain. Furthermore, TRPV1 can activate the cellular sig-
naling pathways including the mitogen-activated pro-
tein kinase (MAPK) family member p38, extracellu-
lar-regulated protein kinase (ERK), c-Jun N-terminal
kinase (JNK), and downstream nuclear factor-κB
(NF-κB) and cAMP response element binding protein
(CREB) during CFA-induced pain. Moreover, nocicep-
tive Nav1.7 and Nav1.8 channels have been shown to
be sensitized under pain conduction in both the DRG
and SDH. Inflammatory factors, such as glial fibrillary
acidic protein (GFAP), S100 calcium-binding protein
B (S100B), and receptor for advanced glycation end
products (RAGE) are also involved in this process.31
TRPV1 also play a crucial role during EA-mediated an-
algesia. EA applied to Zusanli (ST 36) can significantly
attenuate TRPV1 activation and TRPV1-mediated the
above cellular signaling pathways, thus inhibiting the
transmission of nociceptive signals.32,33
Furthermore, under the chronic pain condition, the
plasticity of nociceptive neurons in both the PNS and
the CNS are also altered, which can result in peripheral
and central sensitization.34 Crosstalk between peripher-
al nociceptors and immune cells mediates peripheral
sensitization. Moreover, the neural, immune, and endo-
crine systems communicate with each other via inflam-
matory cytokines (Figure 1).35 Peripheral inflammatory
cytokines are potential messengers that mediate the
Figure 1 EA-induced neuroimmune and neuroendocrine modulation circuit
This circuit includes peripheral afferent and central efferent pathways. The nociceptive stimulus induced by CFA activates the no-
ciceptor and the vagal and sympathetic terminals in the peripheral afferent pathway. Meanwhile, immune cells form a crosstalk
mechanism among the nociceptor, vagal afferent fibers and sympathetic nerve terminal through the release of inflammatory cy-
tokines, such as TNF-α, interleukin-1β, and IL-6. The nociceptive signal is then transmitted to the spinal cord and brain. The modu-
latory signals received from the neural, immune, endocrine systems are then integrated in the brain, which further conveys sec-
ondary neuroimmune and neuroendocrine modulatory signals through the ascending and descending pain control pathway, the
VN, SN, and HPA) axis to regulate inflammatory pain. In the central efferent pathway, vagal efferent fibers and sympathetic pre-
ganglionic neurons act on the adrenal gland to release glucocorticoids and catecholamines, which further inhibit the release of
inflammatory cytokines and eventually mitigate anti-inflammatory pain. EA: electroacupuncture; TNF-α: tumor necrosis factor α;
IL-1β: interleukin-1β; IL-6: interleukin-6;VN: vagal nerve; SN: sympathetic nerve; HPA: hypothalamic-pituitary-adrenal.
742
Li Y et al. / Review
JTCM
|
www. journaltcm. com October 15, 2019
|
Volume 39
|
Issue 5
|
communication between nociceptors and vagal and
sympathetic nerves. Inflammatory cytokines, such as
tumor necrosis factor α(TNF-α), interleukin-1β
(IL-1β), and interleukin-6 (IL-6), are released by pe-
ripheral immune cells to form an "inflammatory
soup", and these cytokines bind with ion channel re-
ceptors, G protein coupled receptors (GPCRs) and ty-
rosine kinase receptors (Trk) that are expressed on noci-
ceptive neurons to mediate peripheral sensitization at
the cellular and molecular levels.4,36 However, inflam-
matory cytokines can also activate the afferent neurons
of the vagal nerve and sympathetic nerve endings
through a central neural circuit; the efferents of the va-
gal and sympathetic pre-ganglionic nerves then act on
the adrenal gland, immune cells or nociceptive neu-
rons, causing the release of norepinephrine (NE) and
acetylcholine (ACh), which inhibit the release of pe-
ripheral inflammatory cytokines.37,38
EA inhibits peripheral sensitization by modulating the
neural, immune and endocrine systems (Figure 2). At
the peripheral level, EA can inhibit the production and
release of inflammatory cytokines through various path-
ways. During inflammation, EA promotes peripheral
immune cells to release opioids, cannabinoids (CB)
and adenosine (A), which further exert anti-inflamma-
tory effects via binding with μ-opioid receptors or
CB2, A1 and A2A receptors that are expressed on noci-
ceptive neurons or immune cells, respectively, to inhib-
it the release of TNF-α, IL-1β, and IL-6.39-42 In addi-
tion to inflammatory cytokines, chemokines also play a
role in inflammatory pain. During the early stages of
inflammation, the chemokine (C-X-C motif) ligand
(CXCL) CXCL2 stimulates opioid release by acting on
the chemokine (C-X-C motif) receptor (CXCR) CX-
CR2 in neutrophils.43 Studies performed in a rodent
model of CFA-induced inflammation have also found
that EA applied to acupoint Huantiao (GB 30) upregu-
lates the expression levels of CXCL10 and CXCR3,
and then stimulates the release of opioids from macro-
phages.44 Macrophages have bidirectional effects on in-
flammatory responses. The M1 macrophage releases
pro-inflammatory cytokines, while the M2 macro-
Figure 2 Mechanisms of EA induced-analgesia and anti-inflammatory effects on inflammatory pain, involving interactions among
the neural, immune, and endocrine systems, at both peripheral and central levels
The injection of CFA/Formalin/Carrageenan/Collagen generates peripheral nociceptive signals to further sensitize the peripheral
nociceptive afferent fibers. Following its initial integration in the DRG, the nociceptive information is transmitted to the SDH and
supraspinal cord through the ascending pathway. Meanwhile, the SDH receives signals that are projected from the descending
pain modulatory system in the brain stem (PAG-RVM/LC-SDH pathway). In addition, the VN, SN, and HPA axis are involved in pain
processing and induce anti-inflammatory responses. EA: electroacupuncture; CFA: complete Freund's adjuvant; DRG: dorsal root
ganglion; SDH: spinal dorsal horn; PAG: periaqueductal grey; RVM: rostral ventromedial medulla; LC: locus coeruleus; VN: vagal
nerve; SN: sympathetic nerve; HPA: hypothalamic-pituitary-adrenal.
743
JTCM
|
www. journaltcm. com October 15, 2019
|
Volume 39
|
Issue 5
|
Li Y et al. / Review
October 15, 2019
|
Volume 39
|
Issue 5
|
phage secretes anti-inflammatory cytokines to inhibit
inflammatory responses.45 EA inhibits M1 macro-
phages, promotes the transformation from M1 to M2
macrophages, and consequently enhances the M2-me-
diated release of interleukin-10 (IL-10), which allevi-
ates inflammatory pain.46 The inflammasome NACHT,
LRR and PYD domains-containing protein 3 (NL-
RP3), a member of the NOD-like receptor (NLR) fam-
ily, is located on macrophages and modulates inflam-
mation and pain by promoting IL-1βmaturation.47 EA
can inhibit NLRP3 release by promoting the expres-
sion of CB2 and β-endorphin, thereby impeding the
maturation of IL-1βand mitigating inflammatory
pain.48 Adenosine exerts anti-inflammatory effects pri-
marily through the activation of A2A receptors in the
periphery.49 EA applied to the acupoints Zusanli (ST
36) and Sanyinjiao (SP 6) increased the levels of A2A
receptors, decreased the release of TNF-α, and simulta-
neously reduced inflammation and hyperalgesia in col-
lagen-induced arthritis model rats.42,50 EA may also act
through MAPK signaling pathways, such as the phos-
phorylation of ERK and JNK, to regulate the expres-
sion levels of TNF-αand IL-1βat both the transcrip-
tional and post-translational levels.51,52 In addition, EA
activates vagal and sympathetic nerves and the HPA ax-
is to inhibit the levels of inflammatory cytokines,
which may affect peripheral sensitization.3,53
Central efferent neuroimmune and neuroendocrine
modulation
The CNS includes the spinal cord and supraspinal
structures. The SDH is one of the major tissues that
regulate the transfer and processing of nociceptive sig-
nals. The lamina ,and layers of the SDH,
which contain a large proportion of intrinsic neurons,
intermediate neurons and projection neurons, primari-
ly receive projections from peripheral nociceptive Aδ-
and C-fibers and establish multiple synaptic connec-
tions. In addition, under peripheral sensitization condi-
tions, spinal microglia and astrocytes become activated,
releasing inflammatory mediators and forming synap-
tic connections with dorsal horn neurons.54,55 Nocicep-
tive signals in the SDH are transported to the brain
stem, thalamus, and cortex through projection neurons
and the ascending pathway (Figure 3). The SDH also
receives signals from the descending pathway that can
inhibit or facilitate the transduction of nociceptive sig-
nals.56 The change of synaptic plasticity in the SDH,
through neuroimmune interactions among spinal noci-
ceptive neurons, glial cells, and T lymphocytes, eventu-
ally results in central sensitization.57,58
The periaqueductal gray (PAG) and the rostral ventro-
medial medulla (RVM) are the major brain regions in-
volved in descending pain control. The analgesic ef-
fects of EA at the spinal cord level are also mediated by
descending inhibitory pathways, which regulate the re-
lease of classical neurotransmitters, such as opioids,
5-HT, norepinephrine (NE), and γ-aminobutyric acid
Figure 3 Anti-inflammatory and anti-nociceptive signaling
pathway induced by EA through peripheral, spinal, and su-
praspinal mechanisms during inflammatory pain
EA inhibited a variety of inflammatory mediators (TNF-α,
IL-1β, IL-6, NGF, and ATP) and promoted the peripheral re-
lease of opioid peptides, cannabinoids and adenosine to
suppress peripheral sensitization. In the spinal dorsal horn,
EA inhibited the postsynaptic excitation of NMDARs and
AMPARs, the MAPK phosphorylation-induced cellular signal-
ing, and the release of glial activation-dependent inflamma-
tory mediators to further attenuate central sensitization.
The EA-induced modulation of pain and inflammation at
the brain level also involves interactions among the central,
immune and endocrine systems. EA modulates the descend-
ing pain inhibitory pathway (PAG-RVM/LC-SDH) to induce
5-HT and NE expression in the SDH. In addition, the expres-
sion of peripheral inflammatory cytokines was regulated by
the HPA axis and the sympathetic and vagal nervous sys-
tems, which were associated with EA-induced neuroim-
mune and neuroendocrine modulations. EA: electroacu-
puncture; CFA: complete Freund's adjuvant; Tc: T lympho-
cyte cell; TNF-α: tumor necrosis factor α; IL-1β: interleu-
kin-1β; IL-6: interleukin-6; IL-17: interleukin-17; IL-10: inter-
leukin-10; IL-33: interleukin-33; P2XR: purinergic P2X recep-
tor; TRPV1: transient receptor potential vanilloid 1; CB: can-
nabinoids; CB2R: cannabinoids 2 receptor; CB1R: cannabi-
noids1 receptor; u-OR: u-opioid receptor; A1R: adrenergic 1
receptors; A2R: adrenergic 2 receptors; DRG: dorsal root gan-
glion; SCDH: spinal cord dorsal horn; SP: substance P; CGRP:
calcitonin-gene related peptide; 5-HT: 5-hydroxytryptamine;
5-HT1AR: 5-hydroxytryptamine 1A receptors; 5-HT2R: 5-hy-
droxytryptamine 2 receptors; NE: norepinephrine; μ-/δ-R: μ-/
δ-opioid receptors; GABA: γ-aminobutyric acid; GABAA/BR:
GABA A/B receptors; Glu: glutamate; α2AR: α2 adrenergic re-
ceptor; NMDAR: N-methyl-d-aspartate receptor; AMPAR: α
amino- 3-hydroxy -5- methyl-4-isoxazole-propionic acid; DA:
Dopamine; FNK: fractalkine; GPCRs: G protein-coupled re-
ceptors; PKC: protein kinase C; MAPK: mitogen-activated
protein kinase; NF-kB: nuclear factor-kB; CREB: cAMP re-
sponse element binding protein; RVM: rostral ventromedial
medulla; LC: locus coeruleus; PAG: periaqueductal grey.
744
Li Y et al. / Review
JTCM
|
www. journaltcm. com October 15, 2019
|
Volume 39
|
Issue 5
|
(GABA), and their corresponding receptors (Figure
2).10,21 Studies have demonstrated that CB1 receptors
are expressed on both GABAergic and glutamatergic
neurons in the PAG, which has different effects on as-
cending and descending pain modulation.59 EA can up-
regulate CB1 expression in GABAergic neurons, inhib-
iting GABA release from PAG neurons, and subse-
quently disinhibiting the release of 5-HT in the RVM,
which promotes the inhibition of chronic pain by de-
scending inhibitory pathways.60 Glial activation is re-
cruited during EA-mediated neuroimmune modula-
tion in the CNS. EA inhibits glial activation and reduc-
es the levels of inflammatory cytokines and other in-
flammatory mediators released by microglia and astro-
cytes, which suppresses central sensitization (Figure
2).61 Inflammation promotes ATP release and binding
with the P2X7 receptors on glial cells, which further ac-
tivates the chemokine fractalkine (FTK). FTK binds
with the receptor CX3CR to induce p38 phosphoryla-
tion, causing glial activation and the release of TNF-α,
brain-derived neurotrophic factor and other inflamma-
tory mediators, which modulate central sensitization.62
EA can inhibit the ATP-FTK-p38 signaling pathway
and suppress glial cell activation, thereby reducing the
expression levels of TNF-αand IL-1β.63 Excitatory glu-
tamatergic neurons, which primarily express α-amino-
3- hydroxy- 5- methyl- 4- isoxazole-propionic acid
(AMPA) and N-methyl-D-aspartate (NMDA) recep-
tors (NMDARs), promote the transmission of nocicep-
tive signals to the spinal cord and mediate central sensi-
tization. The NMDAR subtype NR1 plays a major
role during the regulation of NMDAR excitability,64,65
and the phosphorylation of NR1 promotes the central
sensitization. Inflammatory cytokines released by glial
cells, such as IL-1βand interleukin-17 (IL-17), also
promote the NR1 phosphorylation.66,67 EA inhibits the
IL-1β- and IL-17-induced NR1 phosphorylation in gli-
al cells, alleviating CFA-induced inflammatory pain.68
Moreover, EA also regulates the production and release
of TNF-α, IL-1β, neurokinin-1, and cyclooxygenase-2
at the transcriptional and post-translation levels
through spinal cell signaling pathways, such as the
PKA, PKC, and downstream NF-κB, and CREB path-
ways, which eventually inhibit central sensitization.69-73
At the supraspinal cord level, the brain stem, hypothal-
amus, thalamus, limbic system, cortex, and other im-
portant brain regions are integrated to manage pain in-
formation, including pain related sensory components,
anxiety, depression and other affective components,
which involves the comprehensive regulation of the
nervous, immune and endocrine systems in the brain
(Figure 2). On the one hand, EA regulates pain sensa-
tions and pain aversion through pain-related brain re-
gions in the CNS.74-77 On the other hand, the central ef-
ferents of the hypothalamic-pituitary-adrenal (HPA)
axis, and the sympathetic and the vagal nervous sys-
tems regulate the expression of peripheral inflammato-
ry cytokines (Figure 1).78-80 The adrenal gland is closely
associated with the process of EA-mediated neuroim-
mune and neuroendocrine modulation. The primary
glucocorticoid released from the adrenal cortex is corti-
sol in humans and corticosterone in rodents. The adre-
nal medulla releases catecholamines, including epineph-
rine, NE and dopamine (DA). Peripheral nociceptive
stimuli are transmitted through the spinal cord to the
paraventricular nucleus in the hypothalamus, which
further activates the HPA axis to release glucocorti-
coids and inhibit inflammatory responses.3,81 Previous
studies have demonstrated that EA stimulated the acti-
vation of the HPA axis to release glucocorticoids and
increased the levels of peripheral serum corticosterone,
while inhibiting the expression of inflammatory cyto-
kines, thus alleviating inflammatory pain induced by
CFA.82,83 The neuroimmune and neuroendocrine mod-
ulation induced by vagal and sympathetic nerves is pri-
marily mediated by the release of ACh and NE. The va-
gal nerve acts on the adrenal gland to release ACh
through the afferent nucleus tractus solitarius and the
efferent dorsal motor nucleus. ACh then binds to α7
nicotinic cholinergic receptors expressed on macro-
phages to inhibit the release of TNF-α.84,85 Sympathetic
preganglionic neurons may stimulate the adrenal gland
to release ACh, while postganglionic neurons directly
release NE, independent of the adrenal gland, via α2-/
β-adrenergic receptors in T lymphocytes, which fur-
ther inhibits inflammation. In addition, the postgangli-
onic neurons of the vagal nerve also inhibit the produc-
tion of inflammatory cytokines in the spleen by activat-
ing the sympathetic adrenergic splenic nerve via
a7nAChRs at the celiac ganglion. NE from the splenic
nerve activates T lymphocytes to inhibit cytokine pro-
duction in splenic macrophages.3,5,86 The application of
different EA frequencies results in the selective activa-
tion of sympathetic preganglionic and postganglionic
neurons, which results in different neuroimmune and
neuroendocrine modulatory effects.80 The vagal nerve
plays a role in EA-induced neuroimmune and neuroen-
docrine modulation.78,79 A previous study also suggest-
ed that EA application stimulated the vagal nerve
caused the adrenal gland to release DA, and decreased
TNF-αlevels in the serum and spleen.79 Furthermore,
vagotomy, adrenalectomy and splenectomy can affect
the neuroimmune and neuroendocrine modulation in-
duced by EA.78,79,83 Although the vagal nerve mediates
the anti-inflammatory effects of EA, the role of the va-
gal nerve during pain perception remains poorly under-
stood.
DISCUSSION
The mechanism underlying EA-mediated pain reduc-
tion is multifaceted, ranging from the activation of sin-
gle systems to multisystem connections. The nervous,
immune, and endocrine systems are fundamental to
the autoregulation of the body, and these systems are
all involved in the EA-mediated analgesia.3This article
745
JTCM
|
www. journaltcm. com October 15, 2019
|
Volume 39
|
Issue 5
|
Li Y et al. / Review
describes the anti-inflammatory and anti-nociceptive
response induced by EA application, with a particular
focus on the role played by neural-immune-endocrine
interactions during this process. During inflammation,
peripheral inflammatory cytokines sensitize nociceptive
neurons to transmit nociceptive signals to the spinal
cord and supraspinal cord. Meanwhile, through the
HPA axis and the sympathetic and vagal nervous sys-
tems, nociceptive signals can stimulate the release of
NE and ACh from the adrenal gland, inhibiting the ex-
pression of inflammatory cytokines and forming a neu-
roimmune and neuroendocrine modulatory circuit
(Figure 1). This circuit includes interactions among pe-
ripheral afferents and central efferents that are induced
by the application of EA and mediates the modulation
of the neuroimmune and neuroendocrine systems dur-
ing inflammatory pain. The immune cells and cyto-
kines sensitize the nociceptive neurons, which in turn
activate the immune response. Furthermore, the HPA
axis and sympathetic and vagal nerves inhibit inflam-
mation through interactions with immune cells and no-
ciceptive neurons, forming a feedback loop.3,5,36,87 The
peripheral afferent circuit includes crosstalk among im-
mune cells, inflammatory cytokines, peripheral noci-
ceptive neurons, the HPA axis, and sympathetic and va-
gal nerves. In central efferent pathways, changes in the
synaptic plasticity of spinal nociceptive neurons and gli-
al cells can lead to neuroimmune interactions. Further-
more, nociceptive signals in the neural, immune and
endocrine systems are integrated in corresponding func-
tional brain areas, further modulating the interactions
of neuroimmune and neuroendocrine under pain con-
dition. The anti-inflammatory and anti-nociceptive sig-
naling pathways are regulated by EA at the peripheral,
spinal, and supraspinal levels in inflammatory pain
models, and the mechanisms of EA-induced analgesia
are associated with the inhibition of peripheral and cen-
tral sensitization (Figure 2).21 Peripherally, EA inhibits
the release of pain-related inflammatory mediators dur-
ing inflammatory responses caused by tissue injury, the
expression of pro-nociceptive receptors in peripheral
neurons, and the phosphorylation of cellular signaling
pathways, all of which desensitize peripheral pain per-
ception. Furthermore, EA also promotes the release of
peripheral opioid peptides, cannabinoids and adenos-
ine to inhibit peripheral inflammatory pain. Centrally,
EA inhibits the activation of spinal cord excitatory neu-
rons, including NMDAR and AMPAR-mediated post-
synaptic excitation, the activation of MAPK phosphor-
ylation-induced cellular signaling pathways, the activa-
tion of glial cells, and the release of inflammatory medi-
ators, such as inflammatory cytokines, chemokines,
and ATP, thereby inhibiting central sensitization.10, 21 In
addition, EA enhances descending inhibitory pathways
to prohibit the transmission of pain. At the brain level,
EA modulates pain and inflammation through interac-
tions with the central nervous, immune and endocrine
systems. Furthermore, the peripheral inflammatory cy-
tokine-dependent anti-inflammatory response, mediat-
ed by the HPA axis and the sympathetic and vagal ner-
vous systems, is also involved during EA-mediated anal-
gesia (Figure 3).3Consequently, the multiple modulato-
ry effects of EA on inflammatory pain may be associat-
ed with neural-immune-endocrine interactions.
However, further studies on the EA-induced neuroim-
mune and neuroendocrine modulation of inflammato-
ry pain are required because the neuroimmune and
neuroendocrine modulation mediated by EA is not the
simple addition of EA-induced analgesia to anti-inflam-
matory effects; instead, this modulation represents an
integrated effect that requires interactions among the
nervous, immune and endocrine systems. This article
focused on the inflammatory cytokines that are in-
volved in the interactions among nociceptive neurons,
vagal afferent nerves and sympathetic nerve terminals.
Previous studies showed that crosstalk between sympa-
thetic nerve terminals and peripheral nociceptors in-
volves bradykinin, prostaglandin E2, nerve growth fac-
tor, and adrenaline, in addition to adenosine receptors,
which causes the sensitization of nociceptive neu-
rons.88-90 The vagal afferent nerve may have similar
properties to nociceptive neurons, including discrimi-
native responsiveness to potentially noxious physical
and chemical stimuli, peripheral sensitization and the
capacity to induce central sensitization.91,92 Future stud-
ies are necessary to explore other inflammatory media-
tors involved in nociceptor- and sympathetic/vagal
nerve-mediated neuroimmune and neuroendocrine
modulation to further clarify their roles during the po-
tentiation of inflammatory pain. In addition, although
there are several studies investigating the mechanism
through which EA acts on inflammatory pain in the
nervous system, mechanistic perspectives on the EA-in-
duced neuroimmune and neuroendocrine modulation
of inflammatory pain involving the HPA axis and the
sympathetic/vagal nervous systems are still lacking.
The elucidation of the mechanisms that underly the ef-
fects of EA on inflammatory pain from multiple per-
spectives may further contribute to the clinical practice
of pain management, and studying EA-mediated neu-
roimmune and neuroendocrine modulation is impor-
tant for inflammatory pain control.
REFERENCES
1 Classification of chronic pain. Descriptions of chronic
pain syndromes and definitions of pain terms. Prepared by
the International Association for the Study of Pain, Sub-
committee on Taxonomy. Pain Suppl 1986; 3: S1-S226.
2Chovatiya R, Medzhitov R. Stress, inflammation, and de-
fense of homeostasis. Mol Cell 2014; 54(2): 281-288.
3Ulloa L, Quiroz Gonzalez S, Torres Rosas R. Nerve stimu-
lation: immunomodulation and control of inflammation.
Trends Mol Med 2017; 23(12): 1103-1120.
4Basbaum AI, Bautista DM, Scherrer G, Julius D. Cellu-
lar and molecular mechanisms of pain. Cell 2009; 139(2):
746
Li Y et al. / Review
JTCM
|
www. journaltcm. com October 15, 2019
|
Volume 39
|
Issue 5
|
267-284.
5Talbot S, Foster SL, Woolf CJ. Neuroimmunity: physiolo-
gy and pathology. Annu Rev Immunol 2016; 34: 421-447.
6Butts R, Dunning J. Peripheral and spinal mechanisms of
pain and dry needling mediated analgesia: a clinical re-
source guide for health care professionals. Int J Phys Med
Rehabil 2016; 4(2): 1000327.
7Furlan AD, van Tulder MW, Cherkin DC, et al. Acupunc-
ture and dry-needling for low back pain. Cochrane Data-
base Syst Rev 2005; (1): Cd001351.
8Melzack R, Stillwell DM, Fox EJ. Trigger points and acu-
puncture points for pain: correlations and implications.
Pain 1977; 3(1): 3-23.
9Li F, He T, Xu Q, et al. What is the acupoint? a prelimi-
nary review of acupoints. Pain Med 2015; 16(10):
1905-1915.
10 Zhao ZQ. Neural mechanism underlying acupuncture an-
algesia. Prog Neurobiol 2008; 85(4): 355-375.
11 Kuo TC, Lin CW, Ho FM. The soreness and numbness ef-
fect of acupuncture on skin blood flow. Am J Chin Med
2004; 32(1): 117-129.
12 Lee BC, Ogay V, Kim KW, et al. Acupuncture muscle
channel in the subcutaneous layer of rat skin. J Acupunct
Meridian Stud 2008; 1(1): 13-19.
13 Wu ML, Xu DS, Bai WZ, et al. Local cutaneous nerve ter-
minal and mast cell responses to manual acupuncture in
acupoint LI4 area of the rats. J Chem Neuroanat 2015;
68: 14-21.
14 Kalesnikoff J, Galli SJ. Antiinflammatory and immuno-
suppressive functions of mast cells. Methods mol bio
2011; 677: 207-220.
15 Zhang D, Ding G, Shen X, et al. Role of mast cells in acu-
puncture effect: a pilot study. Explore (NY) 2008; 4(3):
170-177.
16 Chen B, Li MY, Guo Y, Zhao X, Lim HC. Mast cell-de-
rived exosomes at the stimulated acupoints activating the
neuro-immune regulation. Chin J Integr Med 2017; 23
(11): 878-880.
17 Ding N, Jiang J, Qin P, Wang Q, Hu J, Li Z. Mast cells
are important regulator of acupoint sensitization via the se-
cretion of tryptase, 5-hydroxytryptamine, and histamine.
PLoS One 2018; 13(3): e0194022.
18 Yu LL, Li L, Rong PJ, et al. Changes in responses of neu-
rons in spinal and medullary subnucleus reticularis dorsalis
to acupoint stimulation in rats with visceral hyperalgesia.
Evid Based Complement Alternat Med 2014; 2014:
768634.
19 Chen LZ, Kan Y, Zhang ZY, et al. Neuropeptide initiated
mast cell activation by transcutaneous electrical acupoint
stimulation of acupoint LI4 in rats. Sci Rep 2018; 8(1):
13921.
20 Chen L, Xu A, Yin N, et al. Enhancement of immune cy-
tokines and splenic CD4+ T cells by electroacupuncture at
ST 36 acupoint of SD rats. PLoS One 2017; 12(4):
e0175568.
21 Zhang R, Lao L, Ren K, Berman BM. Mechanisms of acu-
puncture-electroacupuncture on persistent pain. Anesthesi-
ology 2014; 120(2): 482-503.
22 Kagitani F, Uchida S, Hotta H. Afferent nerve fibers and
acupuncture. Auton Neurosci 2010; 157(1-2): 2-8.
23 Delmas P, Hao J, Rodat Despoix L. Molecular mecha-
nisms of mechanotransduction in mammalian sensory neu-
rons. Nat Rev Neurosci 2011; 12(3): 139-153.
24 Mihara H, Boudaka A, Sugiyama T, Moriyama Y, Tomina-
ga M. Transient receptor potential vanilloid 4
(TRPV4)-dependent calcium influx and ATP release in
mouse oesophageal keratinocytes. J Physiol 2011; 589(Pt
14): 3471-3482.
25 Page AJ, Brierley SM, Martin CM, et al. Different contri-
butions of ASIC channels 1a, 2, and 3 in gastrointestinal
mechanosensory function. Gut 2005; 54(10): 1408-1415.
26 Li WG, Xu TL. ASIC3 channels in multimodal sensory
perception. ACS Chem Neurosci 2011; 2(1): 26-37.
27 Jara Oseguera A, Simon SA, Rosenbaum T. TRPV1: on
the road to pain relief. Curr Mol Pharmacol 2008; 1(3):
255-269.
28 Xin J, Su Y, Yang Z, et al. Distinct roles of ASIC3 and
TRPV1 receptors in electroacupuncture-induced segmen-
tal and systemic analgesia. Front Med 2016; 10(4):
465-472.
29 Wu SY, Chen WH, Hsieh CL, Lin YW. Abundant expres-
sion and functional participation of TRPV1 at Zusanli
acupoint (ST 36) in mice: mechanosensitive TRPV1 as an
"acupuncture-responding channel". BMC Complement
Altern Med 2014; 14: 96.
30 Abraham TS, Chen ML, Ma SX. TRPV1 expression in
acupuncture points: response to electroacupuncture stimu-
lation. J Chem Neuroanat 2011; 41(3): 129-136.
31 Yang J, Hsieh CL, Lin YW. Role of transient receptor po-
tential vanilloid 1 in electroacupuncture analgesia on
chronic inflammatory pain in mice. Biomed Res Int 2017:
5068347.
32 Lu KW, Hsu CK, Hsieh CL, Yang J, Lin YW. Probing the
effects and mechanisms of electroacupuncture at ipsilateral
or contralateral ST 36-ST37 acupoints on CFA-induced
inflammatory pain. Sci Rep 2016; 6: 22123.
33 Liao HY, Hsieh CL, Huang CP, Lin YW. Electroacupunc-
ture attenuates CFA-induced inflammatory pain by sup-
pressing Nav1.8 through S100B, TRPV1, opioid, and ade-
nosine pathways in mice. Sci Rep 2017; 7: 42531.
34 Ji RR, Nackley A, Huh Y, Terrando N Maixner W. Neuro-
inflammation and central sensitization in chronic and
widespread pain. Anesthesiology 2018;129(2): 343-366.
35 Maier SF, Watkins LR. Cytokines for psychologists: impli-
cations of bidirectional immune-to-brain communication
for understanding behavior, mood, and cognition. Psychol
Rev 1998; 105(1): 83-107.
36 Pinho-Ribeiro FA, Verri WA Jr, Chiu IM. Nociceptor
sensory neuron-immune interactions in pain and inflam-
mation. Trends Immunol 2017; 38(1): 5-19.
37 Johnston GR, Webster NR. Cytokines and the immuno-
modulatory function of the vagus nerve. Br J Anaesth
2009; 102(4): 453-462.
38 Szelényi J, Kiss JP, Puskás E, Szelényi M, Vizi ES. Contri-
bution of differently localized α2- and β-adrenoceptors in
the modulation of TNF-αand IL-10 production in endo-
toxemic mice. Ann N Y Acad Sci 2000; 917(1): 145-153.
39 Jiang YL, He XF, Shen YF, et al. Analgesic roles of periph-
eral intrinsic met-enkephalin and dynorphin A in
long-lasting inflammatory pain induced by complete
747
JTCM
|
www. journaltcm. com October 15, 2019
|
Volume 39
|
Issue 5
|
Li Y et al. / Review
Freund's adjuvant in rats. Exp Ther Med 2015; 9(6):
2344-2348.
40 Su TF, Zhao YQ, Zhang LH, et al. Electroacupuncture re-
duces the expression of proinflammatory cytokines in in-
flamed skin tissues through activation of cannabinoid CB2
receptors. Eur J Pain 2012; 16(5): 624-635.
41 Goldman N, Chen M, Fujita T, et al. Adenosine A1 recep-
tors mediate local anti-nociceptive effects of acupuncture.
Nat Neurosci 2010; 13(7): 883-888.
42 Li QH, Xie WX, Li XP, et al. Adenosine A2A receptors
mediate anti-inflammatory effects of electroacupuncture
on synovitis in mice with collagen-induced arthritis. Evid
Based Complement Alternat Med 2015; 2015: 809560.
43 Rittner HL, Labuz D, Schaefer M, et al. Pain control by
CXCR2 ligands through Ca2 +-regulated release of opioid
peptides from polymorphonuclear cells. FASEB J 2006; 20
(14): 2627-2629.
44 Wang Y, Gehringer R, Mousa SA, Hackel D, Brack A,
Rittner HL. CXCL10 controls inflammatory pain via opi-
oid peptide-containing macrophages in electroacupunc-
ture. PLoS One 2014; 9(4): e94696.
45 Sutterwala FS, Noel GJ, Salgame P, Mosser DM. Reversal
of proinflammatory responses by ligating the macrophage
Fcgamma receptor type . J Exp Med 1998; 188(1):
217-222.
46 da Silva MD, Bobinski F, Sato KL, Kolker SJ, Sluka KA,
Santos AR. IL-10 cytokine released from M2 macrophages
is crucial for analgesic and anti-inflammatory effects of
acupuncture in a model of inflammatory muscle pain.
Mol Neurobiol 2015; 51(1): 19-31.
47 Davis BK, Wen H, Ting JP. The inflammasome NLRs in
immunity, inflammation, and associated diseases. Annu
Rev Immunol 2011; 29: 707-735.
48 Gao F, Xiang HC, Li HP, et al. Electroacupuncture inhib-
its NLRP3 inflammasome activation through CB2 recep-
tors in inflammatory pain. Brain Behav Immun 2018; 67:
91-100.
49 Sawynok J. Adenosine receptor targets for pain. Neurosci-
ence 2016; 338: 1-18.
50 Ye TS, Du ZH, Li ZH, et al. Repeated electroacupuncture
persistently elevates adenosine and ameliorates collagen-in-
duced arthritis in rats. Evid Based Complement Alternat
Med 2016; 2016: 3632168.
51 Chen H, Shao X, Li L, et al. Electroacupuncture serum in-
hibits TNF-α-mediated chondrocyte inflammation via the
Ras-Raf-MEK1/2-ERK1/2 signaling pathway. Mol Med
Rep 2017; 16(5): 5807-5814.
52 Park JY, Park JJ, Jeon S, et al. From peripheral to central:
the role of ERK signaling pathway in acupuncture analge-
sia. J Pain 2014; 15(5): 535-549.
53 Kavoussi B, Ross BE. The neuroimmune basis of anti-in-
flammatory acupuncture. Integr Cancer Ther 2007; 6(3):
251-257.
54 Ji RR, Berta T, Nedergaard M. Glia and pain: is chronic
pain a gliopathy? Pain 2013; 154 Suppl 1: S10-S28.
55 Austin PJ, Moalem-Taylor G. The neuro-immune bal-
ance in neuropathic pain: involvement of inflammatory
immune cells, immune-like glial cells and cytokines. J
Neuroimmunol 2010; 229(1-2): 26-50.
56 Ossipov MH, Morimura K, Porreca F. Descending pain
modulation and chronification of pain. Curr Opin Sup-
port Palliat Care 2014; 8(2): 143-151.
57 Latremoliere A, Woolf CJ. Central sensitization: a genera-
tor of pain hypersensitivity by central neural plasticity. J
Pain 2009; 10(9): 895-926.
58 Grace PM, Hutchinson MR, Maier SF, Watkins LR. Path-
ological pain and the neuroimmune interface. Nat Rev Im-
munol 2014; 14(4): 217-231.
59 Samineni VK, Grajales-Reyes JG, Copits BA, et al. Diver-
gent modulation of nociception by glutamatergic and GA-
BAergic neuronal subpopulations in the periaqueductal
gray. eNeuro 2017; 4(2): e0129.
60 Yuan XC, Zhu B, Jing XH, et al. Electroacupuncture po-
tentiates cannabinoid receptor-mediated descending inhib-
itory control in a mouse model of knee osteoarthritis.
Front Mol Neurosci 2018; 11: 112.
61 Lin L, Skakavac N, Lin X, et al. Acupuncture-induced an-
algesia: the role of microglial inhibition. Cell Transplant
2016; 25(4): 621-628.
62 Clark AK, Malcangio M. Fractalkine/CX3CR1 signaling
during neuropathic pain. Front Cell Neurosci 2014; 8:
121.
63 Gao YH, Li CW, Wang JY, et al. Effect of electroacupunc-
ture on the cervicospinal P2X7 receptor/fractalkine/
CX3CR1 signaling pathway in a rat neck-incision pain
model. Purinergic Signal 2017; 13(2): 215-225.
64 Gao X, Kim HK, Chung JM, Chung K. Enhancement of
NMDA receptor phosphorylation of the spinal dorsal
horn and nucleus gracilis neurons in neuropathic rats.
Pain 2005; 116(1-2): 62-72.
65 Zou X, Lin Q, Willis WD. Enhanced phosphorylation of
NMDA receptor 1 subunits in spinal cord dorsal horn and
spinothalamic tract neurons after intradermal injection of
capsaicin in rats. J Neurosci 2000; 20(18): 6989-6997.
66 Zhang RX, Li A, Liu B, et al. IL-1ra alleviates inflammato-
ry hyperalgesia through preventing phosphorylation of
NMDA receptor NR-1 subunit in rats. Pain 2008; 135(3):
232-239.
67 Meng X, Zhang Y, Lao L, et al. Spinal interleukin-17 pro-
motes thermal hyperalgesia and NMDA NR1 phosphory-
lation in an inflammatory pain rat model. Pain 2013; 154
(2): 294-305.
68 Meng X, Lao L, Shen X, at al. Electroacupuncture inhib-
its spinal interleukin-17a to alleviate inflammatory pain in
a rat model. Open Pain J 2013; 6: 183-189.
69 Fang JQ, Fang JF, Liang Y, Du JY. Electroacupuncture me-
diates extracellular signal-regulated kinase 1/2 pathways in
the spinal cord of rats with inflammatory pain. BMC
Complement Altern Med 2014; 14: 285.
70 Du JY, Fang JQ, Liang Y, Fang JF. Electroacupuncture at-
tenuates mechanical allodynia by suppressing the spinal
JNK1/2 pathway in a rat model of inflammatory pain.
Brain Res Bull 2014; 108: 27-36.
71 Liang Y, Fang JQ, Du JY, Fang JF. Effect of electroacu-
puncture on activation of p38MAPK in spinal dorsal horn
in rats with complete Freund's adjuvant-induced inflam-
matory pain. Evid Based Complement Alternat Med
2012; 2012: 568273.
72 Fang JQ, Du JY, Liang Y, Fang JF. Intervention of elec-
troacupuncture on spinal p38 MAPK/ATF-2/VR-1 path-
748
Li Y et al. / Review
JTCM
|
www. journaltcm. com October 15, 2019
|
Volume 39
|
Issue 5
|
way in treating inflammatory pain induced by CFA in
rats. Mol Pain 2013; 9: 13.
73 Hu ML, Zhou FY, Liu JJ, Ding Y, Zhong JM, Ding MX.
Electroacupuncture inhibits the activation of p38MAPK
in the central descending facilitatory pathway in rats with
inflammatory pain. Evid Based Complement Alternat
Med 2017; 2017: 7531060.
74 Yi M, Zhang H, Lao L, Xing GG, Wang Y. Anterior cin-
gulate cortex is crucial for contra- but not ipsi-lateral elec-
tro-acupuncture in the formalin-induced inflammatory
pain model of rats. Mol Pain 2011; 7: 61.
75 Du J, Fang J, Wen C, Shao X, Liang Y, Fang J. The effect
of electroacupuncture on PKMzeta in the ACC in regulat-
ing anxiety-like behaviors in rats experiencing chronic in-
flammatory pain. Neural Plast 2017; 2017: 3728752.
76 Sun J, Shao X-m, Fang F, Shen Z, Wu YY, Fang JQ. Elec-
troacupuncture alleviates retrieval of pain memory and its
effect on phosphorylation of cAMP response ele-
ment-binding protein in anterior cingulate cortex in rats.
Behav Brain Funct 2015; 11(1): 9.
77 Wu YY, Jiang YL, He XF, et al. 5-HT in the dorsal raphe
nucleus is involved in the effects of 100-Hz electro-acu-
puncture on the pain-depression dyad in rats. Exp Ther
Med 2017; 14(1): 107-114.
78 Lim HD, Kim MH, Lee CY, Namgung U. Anti-inflam-
matory effects of acupuncture stimulation via the vagus
nerve. PLoS One 2016; 11(3): e0151882.
79 Torres-Rosas R, Yehia G, Peña G, et al. Dopamine medi-
ates vagal modulation of the immune system by electroacu-
puncture. Nat Med 2014; 20(3): 291-295.
80 Kim HW, Uh DK, Yoon SY, et al. Low-frequency elec-
troacupuncture suppresses carrageenan-induced paw in-
flammation in mice via sympathetic post-ganglionic neu-
rons, while high-frequency EA suppression is mediated by
the sympathoadrenal medullary axis. Brain Res Bull 2008;
75(5): 698-705.
81 Silverman MN, Sternberg EM. Glucocorticoid regulation
of inflammation and its functional correlates: from HPA
axis to glucocorticoid receptor dysfunction. Ann NY Acad
Sci 2012; 1261: 55-63.
82 Li A, Lao L, Wang Y, et al. Electroacupuncture activates
corticotrophin-releasing hormone-containing neurons in
the paraventricular nucleus of the hypothalammus to alle-
viate edema in a rat model of inflammation. BMC Com-
plement Altern Med 2008; 8: 20.
83 Li A, Zhang RX, Wang Y, et al. Corticosterone mediates
electroacupuncture-produced anti-edema in a rat model of
inflammation. BMC Complement Altern Med 2007; 7:
27.
84 Wang H, Yu M, Ochani M, et al. Nicotinic acetylcholine
receptor α7 subunit is an essential regulator of inflamma-
tion. Nature 2002; 421(6921): 384-388.
85 Corsi-Zuelli FMDG, Brognara F, Quirino GFDS, et al.
Neuroimmune interactions in schizophrenia: focus on va-
gus nerve stimulation and activation of the alpha-7 nico-
tinic acetylcholine receptor. Front Immunol 2017; 8: 618.
86 Szelényi J, Kiss JP, Vizi ES. Differential involvement of
sympathetic nervous system and immune system in the
modulation of TNF-alpha production by alpha2- and be-
ta-adrenoceptors in mice. J Neuroimmunol 2000; 103(1):
34-40.
87 Ashley NT, Demas GE. Neuroendocrine-immune cir-
cuits, phenotypes, and interactions. Horm Behav 2017;
87: 25-34.
88 Schlereth T, Birklein F. Sympathetic nervous system and
pain. Neuromolecular Med 2008; 10(3): 141-147.
89 Khasar SG, Miao FJP, Jänig W, Levine JD. Modulation of
bradykinin-induced mechanical hyperalgesia in the rat by
activity in abdominal vagal afferents. Eur J Neurosci 1998;
10(2): 435-444.
90 Andreev NY, Dimitrieva N, Koltzenburg M, McMahon
SB. Peripheral administration of nerve growth factor in
the adult rat produces a thermal hyperalgesia that requires
the presence of sympathetic post-ganglionic neurones.
Pain 1995; 63(1): 109-115.
91 Kollarik M, Ru F, Brozmanova M. Vagal afferent nerves
with the properties of nociceptors. Auton Neurosci 2010;
153(1-2): 12.
92 Randich A, Gebhart GF. Vagal afferent modulation of no-
ciception. Brain Res Rev 1992; 17(2): 77-99.
749
... The rats were tested for allodynia on Days 0, 8,15,22,30,32,34,36, and 38 as previously described by Zeng et al. 26 First, the rats were placed in Plexiglas cubicles with a stainless steel wire grid floor for 30 min to acclimatize. Then, a series of von Frey filaments from 0.4 to 26 g were applied one by one to the rat scrotal skins for 1 to 2 s each time. ...
... Chronic pain is often persistent and poorly treated, which is a crucial point of focus in acupuncture analgesia. 36 This study indicated the effect of EA on alleviating chronic pelvic pain in CNP rats. It is known that EA could play an analgesic role in various ways. ...
Article
Objective: To investigate the anti-inflammatory effect of electroacupuncture (EA) on chronic prostatitis/chronic pelvic pain syndrome (CP/CPPS), also known as chronic nonbacterial prostatitis (CNP), and explore its underlying mechanism. Methods: A CNP rat was established by surgical castration combined with 17-β estradiol injection in male Sprague-Dawley rats for thirty consecutive days. The CNP rats received EA treatment once a day for eight days. Chronic pelvic pain was evaluated by mechanical withdrawal threshold measurement. The histological change was assessed by hematoxylin-eosin staining. The inflammatory cytokines in prostates were determined by enzyme-linked immunosorbent assays. The expressions of toll-like receptor 4 (TLR4), myeloid differentiation factor 88 (MyD88), inhibitors of kappa-B alpha (IκBα), and nuclear factor-kappa B (NF-κB) were detected by Western blotting. The nuclear translocation of NF-κB and the location of TLR4 were observed with immunofluorescence staining. Results: The results showed that EA decreased the prostate index, upregulated the mechanical withdrawal threshold, restored the histomorphology of the prostate, reduced the inflammatory factor levels, inhibited NF-κB p65 nuclear translocation, and downregulated the expression levels of critical proteins involved in the TLR4/NF-κB signaling pathway in prostates. Conclusions: Our findings suggested that EA could relieve pelvic pain and attenuate prostatic inflammation in estradiol-induced CNP rats. The underlying mechanism may be related to the inhibition of the TLR4/NF-κB signaling pathway.
... Electroacupuncture (EA), which combines acupuncture and electric stimulation, is widely used as a clinical pain management technique [16]. Previous studies have shown that EA relieves pain and improves physical and psychological health of patients with LDH [17,18]. ...
... 9 Acupuncture stimulates energy pathways through specific acupoints to rebalance "Qi" within body and organ systems, to regulate blood circulation and to affect physiological system function. 10,11 Biological mechanisms such as central sensitization, 12 neurotransmitters, 13 immune regulation, 14 oxidative stress, 15 and inflammatory action 16 may be involved. It has been proven to have certain effects in many aspects, such as postoperative or poststroke cognitive impairment, 17,18 angina pectoris, 19 emesis, 20 etc. ...
... Based on analysis of these results, it can be seen that Lao Lixing's team is a very influential team, devoting themselves to mechanistic research from a neuro-immuno-endocrine perspective and improving the quality of randomized controlled trials in acupuncture. [24][25][26] Analysis of Journals ...
Article
Full-text available
Purpose Electroacupuncture is widely used to pain management. A bibliometric analysis was conducted to identify the hotspots and trends in research on electroacupuncture for pain. Methods We retrieved studies published from 1994–2022 on the topic of pain relief by electroacupuncture from the Web of Science Core Collection database. We comprehensively analysed the data with VOSviewer, CiteSpace, and bibliometrix. Seven aspects of the data were analysed separately: annual publication outputs, countries, institutions, authors, journals, keywords and references. Results A total of 2030 papers were analysed, and the number of worldwide publications continuously increased over the period of interest. The most productive country and institution in this field were China and KyungHee University. Evidence-Based Complementary and Alternative Medicine was the most productive journal, and Pain was the most co-cited journal. Han Jisheng, Fang Jianqiao, and Lao Lixing were the most representative authors. Based on keywords and references, three active areas of research on EA for pain were mechanisms, randomized controlled trials, and perioperative applications. Three emerging trends were functional magnetic resonance imaging (fMRI), systematic reviews, and knee osteoarthritis. Conclusion This study comprehensively analysed the research published over the past 28 years on electroacupuncture for pain treatment, using bibliometrics and science mapping analysis. This work presents the current status and landscape of the field and may serve as a valuable resource for researchers. Chronic pain, fMRI-based mechanistic research, and the perioperative application of electroacupuncture are among the likely foci of future research in this area.
... Previous works implicated that EA showed protective effects on cartilage and pain relieving. 31,32 Our present study revealed a novel mechanism about the effect of EA treatment on OA from an epigenetic role-DNA methylation and microRNA interaction concept. EA has been shown to affect DNA methylation in many diseases. ...
Article
Objective: To explore whether electroacupuncture (EA) could alleviate osteoarthritis (OA) through affecting the DNA methylation regulated transcription of miR-146a and miR-140-5p. Methods: Sixty male eight-week-old Sprague-Dawley rats were divided into three groups: normal group (normal healthy rats; no treatment), model group (OA rats; no treatment) and EA group (OA rats treated with EA). Safranin O staining and modified Mankin's score were performed to evaluate the histopathological alterations and degeneration of cartilage 8 weeks after 8 consecutive weeks of treatment. Quantitative real time polymerase chain reaction (qRT-PCR) assay was employed to evaluate the expression of miR-146a in the cartilage tissue and miR-140-5p in the synovium tissue, respectively. The bisulfite sequencing analysis and quantitative methylation specific PCR (qMSP) were used to analyze the status of methylation in the regulatory regions of miR-146a and miR-140-5p. Chromatin immunoprecipitation (ChIP) assay were performed to assess the binding of nuclear factor-kappa B (NF-κB) and signal transducer and activator of transcription 3 (SMAD-3) in the regulatory regions of miR-146a and miR-140-5p. Western blot analysis was performed to detect the expressions of DNA Methyltransferase 1 (DMNT1), DNA Methyltransferase 3A (DMNT3A), and DNA Methyltransferase 3A (DMNT3b), NF-κB, SMAD3 levels. Results: Our results showed that EA treatment significantly upregulated miR-146a and miR-140-5p expressions. qMSP analysis showed that EA significantly decreased methylation levels of miR-140-5p regulated region and miR-146a promoter in OA cartilage and synovium. Bisulfite DNA sequencing (BDS) and ChIP analysis showed that EA significantly increased binding affinity of SMAD3 and NF-kB on the hypermethylated miR-140 regulatory region and miR-146a promoter, respectively. Western Blot analysis demonstrated that EA also significantly decreased expressions of methylation related proteins- DMNT1, DMNT3a, and DMNT3b as well as NF-κB and SMAD3. Conclusions: Electroacupuncture stimulating Neixiyan (EX-LE5) and Dubi (ST35) may alleviate OA affecting the DNA methylation regulated transcription of miR-146a and miR-140-5p.
... Inflammatory pain is a complex condition characterised by multiple mechanisms, such as hyperexcitability and/or sensitisation of primary nociceptive neurons or nociceptors and neural-immune-endocrine interactions [1][2][3][4]. Inflammatory pain is one of the major contributors to the health care burden and is associated with many chronic diseases [5,6]. Despite the high prevalence and severity of inflammatory pain, the medications currently available to treat inflammatory pain are unsatisfactory, such as nonsteroidal anti-inflammatory drugs (NSAIDs), opioids and some adjuvant medications [7]. ...
Article
Full-text available
Elevated excitability of glutamatergic neurons in the lateral parabrachial nucleus (PBL) is associated with the pathogenesis of inflammatory pain, but the underlying molecular mechanisms are not fully understood. Sodium leak channel (NALCN) is widely expressed in the central nervous system and regulates neuronal excitability. In this study, chemogenetic manipulation was used to explore the association between the activity of PBL glutamatergic neurons and pain thresholds. Complete Freund's adjuvant (CFA) was used to construct an inflammatory pain model in mice. Pain behaviour was tested using von Frey filaments and Hargreaves tests. Local field potential (LFP) was used to record the activity of PBL glutamatergic neurons. Gene knockdown techniques were used to investigate the role of NALCN in inflammatory pain. We further explored the downstream projections of PBL using cis-trans-synaptic tracer virus. The results showed that chemogenetic inhibition of PBL glutamatergic neurons increased pain thresholds in mice, whereas chemogenetic activation produced the opposite results. CFA plantar modelling increased the number of C-Fos protein and NALCN expression in PBL glutamatergic neurons. Knockdown of NALCN in PBL glutamatergic neurons alleviated CFA-induced pain. CFA injection induced C-Fos protein expression in central nucleus amygdala (CeA) neurons, which was suppressed by NALCN knockdown in PBL glutamatergic neurons. Therefore, elevated expression of NALCN in PBL glutamatergic neurons contributes to the development of inflammatory pain via PBL-CeA projections.
... Furthermore, multiple studies have shown that acupuncture can act on multiple aspects, including the neuroendocrine, immune, metabolic, and hemodynamic systems, to produce a regulatory effect, thereby improving fatigue symptoms in cancer patients [53]. In addition, acupuncture has been proven to significantly improve the immune function of patients with advanced cancer, thereby improving fatigue, depression, and sleep disorders [54]. ...
Article
Full-text available
Objective Acupuncture has become a popular complementary treatment in oncology. This study is based on RNA-Seq transcriptome sequencing technology to investigate the molecular mechanisms underlying the effect of acupuncture-mediated regulation of the Leptin/AMPK signaling pathway on mitochondrial dysfunction-induced fatigue in breast cancer patients after chemotherapy. Methods Peripheral blood samples from 10 patients with post-operative chemotherapy for breast cancer were selected for transcriptome sequencing to screen the key molecular pathways involved in fatigue after chemotherapy in breast cancer patients. Besides, peripheral blood samples were collected from 138 post-operative chemotherapy patients with breast cancer to study the composite fatigue and quality of life scores. Flow cytometry was used to detect T lymphocyte subsets in peripheral blood-specific immune cells. In addition, a blood cell analyzer was used to measure peripheral blood leukocyte counts, and MSP-PCR was used to detect mitochondrial DNA mutations in peripheral blood leukocytes. Results Transcriptome bioinformatics analysis screened 147 up-regulated mRNAs and 160 down-regulated mRNAs. Leptin protein was confirmed as the key factor. Leptin was significantly higher in the peripheral blood of breast cancer patients who developed fatigue after chemotherapy. Acupuncture treatment effectively improved post-chemotherapy fatigue and immune status in breast cancer patients, suppressed the expression of Leptin/AMPK signaling pathway-related factor and leukocyte counts, and significantly reduced the rate of mitochondrial DNA mutations in peripheral blood leukocytes. Conclusion The Leptin/AMPK signaling pathway may be the key molecular pathway affecting the occurrence of fatigue after chemotherapy in breast cancer patients. Leptin may improve post-chemotherapy fatigue in breast cancer patients by activating AMPK phosphorylation and alleviating mitochondrial functional impairment.
... This stimulation can also effectively overcome the phenomenon of electroacupuncture tolerance. Furthermore, electroacupuncture has been found to promote nerve fiber regeneration, which is often associated with dysplasia of nerve fiber in the colon wall, a common factor in secondary constipation [40][41][42]. Studies have also shown that electroacupuncture can regulate the content of brain and intestinal peptides in the body, thereby improving the brain-intestinal axis pathway and regulating intestinal function and movement. ...
Article
Full-text available
Objective Secondary constipation refers to constipation that occurs after certain diseases or medications, such as acute stroke or opioids, and the efficacy of electroacupuncture for secondary constipation is controversial. So, this study aimed to explore the efficacy and safety of electroacupuncture for secondary constipation through a meta-analysis and systematic review. Method We retrieved articles from PubMed, Embase, Cochrane Library, Web of Science, CNKI, Wanfang, and VIP databases up to 28 February 2023. The study was screened strictly according to inclusion and exclusion criteria. Revman5.4 was used for quality evaluation; grade rating was used for index evaluation, and stata15.0 was used for data consolidation analysis. Result Thirteen randomized controlled studies, involving a total of 1437 people (722 electroacupuncture and 715 control groups), were included in this review. Meta-analysis results indicated that electroacupuncture significantly improved constipation overall response (RR = 1.31, 95%CI: 1.11, 1.55, P < 0.001), reduced defecation straining score (MD = − 0.46, 95%CI: − 0.67, − 0.251, P < 0.001), increased weekly complete spontaneous bowel movements (MD = 0.41, 95%CI: 0.20, 0.63, P = 0.002), and increased in the weekly spontaneous bowel movements (MD = 0.80, 95%CI (0.49, 01.11), P < 0.001), and electroacupuncture had no effect on change stool consistency score compared (MD = − 0.03, 95%CI (− 0.38, 0.33), P = 0.88) and did not increase adverse events (RR = 0.50, 95%CI: 0.18, 1.44, P = 0.20). Conclusion According to the current studies, the overall relief rate of patients with secondary constipation after electroacupuncture treatment was improved, the defecation pressure score was reduced, the weekly natural defecation was more complete, and adverse reactions were not increased. Electroacupuncture therefore shows potential for treating constipation, but more high-quality studies are needed to confirm these findings.
Article
Full-text available
Acupuncture is a traditional medicinal practice in China that has been increasingly recognized in other countries in recent decades. Notably, several reports have demonstrated that acupuncture can effectively aid in pain management. However, the analgesic mechanisms through which acupuncture provides such benefits remain poorly understood. Purinergic signaling, which is mediated by purine nucleotides and purinergic receptors, has been proposed to play a central role in acupuncture analgesia. On the one hand, acupuncture affects the transmission of nociception by increasing adenosine triphosphate dephosphorylation and thereby decreasing downstream P2X3, P2X4, and P2X7 receptors signaling activity, regulating the levels of inflammatory factors, neurotrophic factors, and synapsin I. On the other hand, acupuncture exerts analgesic effects by promoting the production of adenosine, enhancing the expression of downstream adenosine A1 and A2A receptors, and regulating downstream inflammatory factors or synaptic plasticity. Together, this systematic overview of the field provides a sound, evidence-based foundation for future research focused on the application of acupuncture as a means of relieving pain.
Article
Full-text available
Objective This paper aims to review the current evidence on electroacupuncture as an effective and safe therapy for cancer pain management. Methods Five databases were searched from their inception through November 11, 2022. Only the randomized controlled trials that meet the eligibility criteria were finally included in the study. Literature screening and data extraction were performed independently by two reviewers, and RevMan 5.3 used for meta-analysis. Results A total of 17 RCTs met our inclusion criteria. We used 8 indicators to estimate the meta-analysis results, most of which proved statistically significant, including VAS scores, NRS scores, and KPS scores. To be specific, VAS scores (MD = −1.41, 95% CI: −2.42 to −0.41, P = 0.006) and NRS scores (MD = −1.19, 95% CI: −1.72 to −0.66, P < 0.0001) were significantly lower in the treatment group compared to the control group. The treatment group's KPS scores (MD = 5.48, 95% CI: 3.27 to 7.69, P < 0.00001) were higher than those of the control group. Also, in the treatment group, the number of burst pain (MD = −2.66, 95% CI: −3.32 to −1.99, P < 0.00001) and side effect rates (RR = 0.51, 95% CI: 0.39 to 0.67, P < 0.00001) greatly reduced, while the response rate (RR = 1.17, 95% CI: 1.09 to 1.26, P < 0.0001) significantly increased compared to the control group. Conclusion This study demonstrates the advantages of electroacupuncture in the treatment of cancer pain. Meanwhile, rigorous RCTs should be designed and conducted in the future to further demonstrate the exact efficacy of electroacupuncture. Systematic Review Registration https://www.crd.york.ac.uk/PROSPERO/ , identifier CRD42022376148.
Article
Full-text available
Transcutaneous electrical acupoint stimulation (TEAS) has been consistently used clinically for its ease of operation, non-invasiveness and painlessness, in contrast to the characteristics of inserted needles. However, the mechanism remains unknown. The aim of this study was to investigate the local response of TEAS at Hegu acupoint (LI4). Immunohistochemistry was used to measure the expression of tryptase-positive mast cells, neuropeptides of the calcitonin gene-related peptide (CGRP) and substance P (SP) in LI4. Mast cells were also labelled with serotonin (5-HT), neurokinin-1 receptor (NK-1R) and toluidine blue. The results showed that cutaneous CGRP and SP immune-positive (CGRP-IP or SP-IP) nerve fibres in LI4 were more highly expressed. There were high degrees of mast cell aggregation and degranulation with release of 5-HT near the CGRP-IP or SP-IP nerve fibres and blood vessels after TEAS. The degranulation of mast cells (MCs) was accompanied by expression of NK-1R after TEAS. Either mast cell membrane stabilizer (Disodium cromoglycate) or NK-1R antagonist (RP 67580) diminished the accumulation and degranulation of MCs induced by TEAS. Taken together, the findings demonstrated that TEAS induced sensory nerve fibres to express CGRP and SP, which then bound to the NK-1R on MCs, after which MCs degranulated and released 5-HT, resulting in TEAS-initiated acupuncture-like signals.
Article
Full-text available
Knee osteoarthritis (KOA) is a highly prevalent, chronic joint disorder, which can lead to chronic pain. Although electroacupuncture (EA) is effective in relieving chronic pain in the clinic, the involved mechanisms remain unclear. Reduced diffuse noxius inhibitory controls (DNIC) function is associated with chronic pain and may be related to the action of endocannabinoids. In the present study, we determined whether EA may potentiate cannabinoid receptor-mediated descending inhibitory control and inhibit chronic pain in a mouse model of KOA. We found that the optimized parameters of EA inhibiting chronic pain were the low frequency and high intensity (2 Hz + 1 mA). EA reversed the reduced expression of CB1 receptors and the 2-arachidonoylglycerol (2-AG) level in the midbrain in chronic pain. Microinjection of the CB1 receptor antagonist AM251 into the ventrolateral periaqueductal gray (vlPAG) can reversed the EA effect on pain hypersensitivity and DNIC function. In addition, CB1 receptors on GABAergic but not glutamatergic neurons are involved in the EA effect on DNIC function and descending inhibitory control of 5-HT in the medulla, thus inhibiting chronic pain. Our data suggest that endocannabinoid (2-AG)-CB1R-GABA-5-HT may be a novel signaling pathway involved in the effect of EA improving DNIC function and inhibiting chronic pain.
Article
Full-text available
Mast cells (MCs) play a crucial role in mediating the establishment of networks among the circulatory, nervous and immune system at acupoints. However, the changes which occur in MCs during acupoint sensitization, i.e. the dynamic transformation of an acupoint from a "silenced" to an "activated" status, remain uncharacterized. To investigate the morphological and functional changes of MCs as an aid to understanding the cellular mechanism underlying acupoint sensitization, a rat model of knee osteoarthritis (OA) was induced by an injection of mono-iodoacetate (MIA) on day 0. On day 14, toluidine blue and immunofluorescence staining were used to observe the recruitment and degranulation of MCs and the release of mast cell co-expressed mediators: tryptase, 5-hydroxytryptamine (5-HT) and histamine (HA) at the acupoints Yanglingquan (GB34), Heding (EX-LE2) and Weizhong (BL40). Results showed that the number of MCs as well as the percentages of degranulated and extensively degranulated MCs at the acupoints GB34 and EX-LE2 in the light (A), mild (B), heavy (C) osteoarthritis groups were larger than those in the normal control (N) and normal saline (NS) groups (p < 0.01). Comparisons among the A, B and C groups suggested that the number and the degranulation extent of the MCs at the acupoints GB34 and EX-LE2 were positively correlated with the severity of the disease. Some MCs in the A, B and C group showed the release of 5-HT, HA, and tryptase in degranulation at the acupoints GB34 and EX-LE2. Such changes in MCs were not observed at the acupoint BL40. In conclusion, this study confirmed that acupoint sensitization is associated with the increase in recruitment and degranulation levels of MCs on a acupoint-specific and disease severity-dependent manner. The release of tryptase, 5-HT, and HA during MC degranulation is likely to be one of the cellular mechanisms occurring during acupoint sensitization.
Article
Full-text available
Chronic inflammatory pain may result from peripheral tissue injury or inflammation, increasing the release of protons, histamines, adenosine triphosphate, and several proinflammatory cytokines and chemokines. Transient receptor potential vanilloid 1 (TRPV1) is known to be involved in acute to subacute neuropathic and inflammatory pain; however, its exact mechanisms in chronic inflammatory pain are not elucidated. Our results showed that EA significantly reduced chronic mechanical and thermal hyperalgesia in the chronic inflammatory pain model. Chronic mechanical and thermal hyperalgesia were also abolished in TRPV1 −/− mice. TRPV1 increased in the dorsal root ganglion (DRG) and spinal cord (SC) at 3 weeks after CFA injection. The expression levels of downstream molecules such as pPKA, pPI3K, and pPKC increased, as did those of pERK, pp38, and pJNK. Transcription factors (pCREB and pNF κ B) and nociceptive ion channels (Nav1.7 and Nav1.8) were involved in this process. Inflammatory mediators such as GFAP, S100B, and RAGE were also involved. The expression levels of these molecules were reduced in EA and TRPV1 −/− mice but not in the sham EA group. Our data provided evidence to support the clinical use of EA for treating chronic inflammatory pain.
Article
Full-text available
The mitogen-activated protein kinases (MAPKs), especially p38MAPK, play a pivotal role in chronic pain. Electroacupuncture (EA) relieves inflammatory pain underlying the descending pathway, that is, the periaqueductal gray (PAG), the rostral ventromedial medulla (RVM), and the spinal cord dorsal horn (SCDH). However, whether EA antagonizes inflammatory pain through regulation of p38MAPK in this descending facilitatory pathway is unclear. Complete Freund’s adjuvant (CFA) was injected into the hind paw of rats to establish inflammatory pain model. EA was administrated for 30 min at Zusanli and Kunlun acupoints at 0.5, 24.5, 48.5, and 72.5 h, respectively. The paw withdrawal threshold (PWT), paw edema, and Phosphor-p38MAPK-Immunoreactivity (p-p38MAPK-IR) cells were measured before (0 h) and at 1, 3, 5, 7, 25, and 73 h after CFA or saline injection. EA increased PWT at 1, 3, 25, and 73 h and inhibited paw edema at 25 and 73 h after CFA injection. Moreover, the increasing number of p-p38MAPK-IR cells which was induced by CFA was suppressed by EA stimulation in PAG and RVM at 3 and 5 h and in SCDH at 5, 7, 25, and 73 h. These results suggest that EA suppresses inflammation-induced hyperalgesia probably through inhibiting p38MAPK activation in the descending facilitatory pathway.
Article
Full-text available
The Ras‑Raf‑mitogen‑activated protein kinase kinase (MEK)1/2‑extracellular signal‑regulated kinase (ERK)1/2 signaling pathway contributes to the release of chondral matrix‑degrading enzymes and accelerates the degradation of articular cartilage. Electroacupuncture (EA) treatment has been widely used for the treatment of osteoarthritis (OA); however, the mechanism underlying the effects of EA on OA remains unclear. Therefore, the present study evaluated the anti‑inflammatory effects and potential underlying mechanisms of EA serum (EAS) on tumor necrosis factor (TNF)‑α‑mediated chondrocyte inflammation. A total of 30 Sprague Dawley rats were randomly divided into three groups: The blank group; experimental group I, which received 15 min of EA treatment; and experimental group II, which received 30 min of EA treatment. Subsequently, serum samples were obtained. Chondrocytes were isolated from the knee cartilage of Sprague Dawley rats, and were identified using collagen type II immunohistochemistry. TNF‑α‑treated chondrocytes were used as a cell model, and subsequently the cells were treated with EAS from each group for various durations. The results demonstrated that EAS treatment significantly promoted the viability and inhibited the apoptosis of TNF‑α‑treated chondrocytes. In addition, interleukin (IL)‑1β concentration was significantly increased in the model group compared with in the control group, whereas EAS significantly reduced IL‑1β concentration in TNF‑α‑treated chondrocytes. Furthermore, the protein expression levels of Ras, Raf and MEK1/2 were reduced in the EAS groups compared with in the model group. EAS also significantly inhibited the phosphorylation of ERK1/2, and the expression of downstream regulators matrix metalloproteinase (MMP)‑3 and MMP‑13. In conclusion, these results indicated that EAS may inhibit TNF‑α‑mediated chondrocyte inflammation via the Ras‑Raf‑MEK1/2‑ERK1/2 signaling pathway in vitro, thus suggesting that EAS may be considered a potential therapeutic strategy for the treatment of OA.
Article
Full-text available
The pain‑depression dyad is becoming widespread in the clinic and is attracting increasing attention. A previous study by our group found that 100‑Hz electro‑acupuncture (EA), but not 2‑, 50‑and 2/100‑Hz EA, was effective against the reserpine‑induced pain‑depression dyad. This finding is in contrast to the fact that low‑frequency EA is commonly used to treat supraspinal‑originating diseases. The present study aimed to investigate the mechanism underlying the effects of 100‑Hz EA on the pain‑depression dyad. Repeated reserpine injection was found to induce allodynia and depressive behav­iors in rats. It decreased 5‑hydroxytryptamine (5‑HT) levels and immunoreactive expressions in the dorsal raphe nucleus (DRN). 100‑Hz EA alleviated the pain‑depression dyad and upregulated 5‑HT in the DRN of reserpine‑injected rats. Intracerebroventricular injection of para‑chlorophenylalanine, an inhibitor of 5‑HT resynthesis, suppressed the upregulation of 5‑HT in the DRN by 100‑Hz EA and partially counteracted the analgesic and anti‑depressive effects of 100‑Hz EA. The present study was the first to demonstrate that 5‑HT in the DRN is involved in mediating the analgesic and anti‑depres­sive effects of 100‑Hz EA on the pain‑depression dyad. This finding provided a scientific basis for high‑frequency EA as a potential treatment for the pain‑depression dyad.
Article
Chronic pain is maintained in part by central sensitization, a phenomenon of synaptic plasticity, and increased neuronal responsiveness in central pain pathways after painful insults. Accumulating evidence suggests that central sensitization is also driven by neuroinflammation in the peripheral and central nervous system. A characteristic feature of neuroinflammation is the activation of glial cells, such as microglia and astrocytes, in the spinal cord and brain, leading to the release of proinflammatory cytokines and chemokines. Recent studies suggest that central cytokines and chemokines are powerful neuromodulators and play a sufficient role in inducing hyperalgesia and allodynia after central nervous system administration. Sustained increase of cytokines and chemokines in the central nervous system also promotes chronic widespread pain that affects multiple body sites. Thus, neuroinflammation drives widespread chronic pain via central sensitization. We also discuss sex-dependent glial/immune signaling in chronic pain and new therapeutic approaches that control neuroinflammation for the resolution of chronic pain.
Article
Neuronal stimulation is an emerging field in modern medicine to control organ function and re-establish physiological homeostasis during illness. Transdermal nerve stimulation with electroacupuncture is currently endorsed by the World Health Organization (WHO) and the National Institutes of Health (NIH), and is used by millions of people to control pain and inflammation. Recent advances in electroacupuncture may permit activation of specific neuronal networks to prevent organ damage in inflammatory and infectious disorders. Experimental studies of nerve stimulation are also providing new information on the functional organization of the nervous system to control inflammation and its clinical implications in infectious and inflammatory disorders. These studies may allow the design of novel non-invasive techniques for nerve stimulation to help to control immune and organ functions.
Article
The therapeutic effect of electroacupuncture (EA) on inflammatory pain has been well recognized clinically. The inflammasome promotes the maturation of the inflammatory cytokines, and EA can stimulate cannabinoid CB2 receptors in inflamed tissues. In this study we investigated whether EA inhibits NLRP3 inflammasome activation through CB2 receptors and thus relieving inflammatory pain. Assay of Caspase-1 activity and western blotting revealed that complete Freund's adjuvant (CFA) injection activated the NLRP3 inflammasome in the skin tissue in rats, which was attenuated by EA treatment. Immunofluorescence labeling showed that NLRP3 inflammasome elicited by CFA in the skin macrophages were decreased by EA. Nociceptive behavioral tests demonstrated that in CB2 receptor knockout mice, the EA effects on NLRP3 inflammasomes were largely attenuated. In addition, in vitro studies in a macrophage cell line showed that CB2 receptor stimulation inhibited the NLRP3 inflammasome activation. Thus, our results suggest a novel signaling pathway through which CB2 receptors are involved in the analgesic effect of EA on inflammatory pain. Stimulation of CB2 receptors inhibits NLRP3 inflammasome activation in inflamed skin tissues. These results suggest that EA reduces the inflammatory pain by inhibiting the activation of NLRP3 inflammasome through CB2 receptors. Our findings provide novel information about the mechanisms through which EA and CB2 receptor activation reduce inflammatory pain.