You are on page 1of 18

Brain, Behavior, and Immunity 95 (2021) 344–361

Contents lists available at ScienceDirect

Brain Behavior and Immunity


journal homepage: www.elsevier.com/locate/ybrbi

Microglial IL-10 and β-endorphin expression mediates gabapentinoids


antineuropathic pain
Khalil Ali Ahmad, Rana Muhammad Shoaib, Muhammad Zaeem Ahsan, Meng-Yan Deng, Le Ma,
Evhy Apryani, Xin-Yan Li, Yong-Xiang Wang *
King’s Lab, Shanghai Jiao Tong University School of Pharmacy, 800 Dongchuan Road, Shanghai 200240, China

A R T I C L E I N F O A B S T R A C T

Keywords: Gabapentinoids are recommended first-line treatments for neuropathic pain. They are neuronal voltage-
Gabapentinoids dependent calcium channel α2δ-1 subunit ligands and have been suggested to attenuate neuropathic pain via
Spinal cord interaction with neuronal α2δ-1 subunit. However, the current study revealed their microglial mechanisms
Neuropathic pain
underlying antineuropathic pain. Intrathecal injection of gabapentin, pregabalin and mirogabalin rapidly
Microglia
IL-10/β-endorphin
inhibited mechanical allodynia and thermal hyperalgesia, with projected ED50 values of 30.3, 6.2 and 1.5 µg (or
α2δ-1 176.9, 38.9 and 7.2 nmol) and Emax values of 66%, 61% and 65% MPE respectively for mechanical allodynia.
Intrathecal gabapentinoids stimulated spinal mRNA and protein expression of IL-10 and β-endorphin (but not
dynorphin A) in neuropathic rats with the time point parallel to their inhibition of allodynia, which was observed
in microglia but not astrocytes or neurons in spinal dorsal horns by using double immunofluorescence staining.
Intrathecal gabapentin alleviated pain hypersensitivity in male/female neuropathic but not male sham rats,
whereas it increased expression of spinal IL-10 and β-endorphin in male/female neuropathic and male sham rats.
Treatment with gabapentin, pregabalin and mirogabalin specifically upregulated IL-10 and β-endorphin mRNA
and protein expression in primary spinal microglial but not astrocytic or neuronal cells, with EC50 values of 41.3,
11.5 and 2.5 µM and 34.7, 13.3 and 2.8 µM respectively. Pretreatment with intrathecal microglial metabolic
inhibitor minocycline, IL-10 antibody, β-endorphin antiserum or μ-opioid receptor antagonist CTAP (but not κ- or
δ-opioid receptor antagonists) suppressed spinal gabapentinoids-inhibited mechanical allodynia. Immunofluo­
rescence staining exhibited specific α2δ-1 expression in neurons but not microglia or astrocytes in the spinal
dorsal horns or cultured primary spinal cells. Thus the results illustrate that gabapentinoids alleviate neuropathic
pain through stimulating expression of spinal microglial IL-10 and consequent β-endorphin.

1. Introduction mirogabalin, a more potent analog, has also been approved in Japan to
treat peripheral neuropathic pain (Deeks 2019). Including gabapentin,
Gabapentin and pregabalin are anticonvulsant drugs that have been pregabalin and mirogabalin, gabapentinoids provide a major new class
prescribed as first-line treatments for neuropathic pain, including post­ of analgesics for neuropathic pain and pain treatment, in addition to
herpetic neuralgia, peripheral diabetic neuropathy and fibromyalgia. opioids and NSAIDs.
Their pain indications have been extended to postoperative pain, With the widespread use of gabapentinoids, it has been an active area
headache, chronic low back pain and visceral pain (Stahl et al. 2013). to figure out their molecular mechanisms underlying analgesia in
Their labeled and off-labeled pain uses, together with other indication neuropathic pain. Gabapentin and pregabalin were first discovered to
uses, have trebled in the United States over the past two decades bind to a high-affinity binding site in rat brain homogenates (Suman-
(González-Bueno et al. 2015), probably driven by raising awareness of Chauhan et al. 1993), which was later found to be the α2δ-1 subunit of
the importance of non-opioid pharmacologic therapies. Recently, voltage-gated Ca2+ channels (Gee et al. 1996). The voltage-gated Ca2+

Abbreviations: IL, interleukin; MOR, µ-opioid receptor; KOR, κ-opioid receptor; DOR, δ-opioid receptor; LPS, lipopolysaccharide; NASAIDs, non-ster­
oidal antiinflammatory drugs; CFA, complete Freund’s adjuvant; CRF, corticotropin-releasing factor; GABA, γ-aminobutyric acid; NMDA, N-methyl-D-aspartate; GLP-
1R, glucagon-like peptide-1 receptors; α7nAChRs, α7-nicotinic acetylcholine receptors; GPR40, G-protein receptor 40; PBS, phosphate-buffered saline.
* Corresponding author.
E-mail address: yxwang@sjtu.edu.cn (Y.-X. Wang).

https://doi.org/10.1016/j.bbi.2021.04.007
Received 27 December 2020; Received in revised form 11 April 2021; Accepted 12 April 2021
Available online 20 April 2021
0889-1591/© 2021 Elsevier Inc. All rights reserved.
K.A. Ahmad et al. Brain Behavior and Immunity 95 (2021) 344–361

channels (particularly N-type calcium channels) are associated with the activation (Rosa et al., 2017; Wodarski et al., 2009). On the other hand,
two auxiliary α2δ-1 and β subunits, and their coexpression with the main gabapentin was reported to exert antinociception in neuropathic rats via
pore-forming α1 subunit leads to a significant rise in the cell Ca2+ cur­ upregulation of spinal antiinflammatory cytokine IL-10 and subsequent
rent amplitude and a role in Ca2+ channels membrane incorporation. inhibition of proinflammatory cytokines containing TNF-α, IL-6 and IL-
Neuronal α2δ-1 has been accepted to be the target site for gabapenti­ 1β (Lee et al. 2013). By the same mode, the intrathecal administration of
noids (Cheng and Chiou, 2006; Stahl et al., 2013), with binding affinities gabapentin also enhanced morphine antinociception (Bao et al. 2014b).
(Kd) in nanomolar (Domon et al., 2018a, 2018b; Li et al., 2011; Suman- Thus the present study intended to investigate the microglial mecha­
Chauhan et al., 1993). α2δ-1 is expressed predominantly in spinal dorsal nisms underlying gabapentinoids analgesia in neuropathic pain, mainly
horn neurons, localized in the superficial laminae and moderately the relationship with the spinal IL-10/β-endorphin microglial signaling
expressed postsynaptically on deeper neurons (Li et al., 2006; Bauer pathway. Firstly, we evaluated the inhibitory effects of intrathecal
et al., 2009). It was discovered that peripheral nerve injury upregulated gabapentin, pregabalin and mirogabalin on mechanical allodynia in a
presynaptic α2δ-1 expression and trafficking, which accounted for neuropathic pain model. Then we determined whether gabapentinoids
neuropathic pain development (Bauer et al., 2009; Hoppa et al., 2012; specifically expressed IL-10 and β-endorphin in spinal microglia. Addi­
Zhou and David, 2013). It has been suggested that gabapentin and tionally, we directly measured gabapentinoids on IL-10 and β-endorphin
pregabalin interfere with anterograde trafficking of α2δ-1 from dorsal expression in microglial cells. Further, we explored the causal rela­
root ganglion neurons to the presynaptic terminals of the dorsal horns, tionship between their analgesia and microglial IL-10 and β-endorphin
resulting in reduced release of pain neurotransmitters such as glutamate expression. Lastly, we made efforts to identify molecule targets for
and spinal sensitization, and subsequently attenuate neuropathic pain gabapentinoids to express IL-10 and β-endorphin in microglia. Our re­
(Stahl et al., 2013; Hendrich et al., 2008). However, the gabapentinoids sults demonstrated that spinal gabapentinoids produced fast anti­
results are inconsistent with inhibition of Ca2+ currents, pain transmitter neuropathic effects through microglial IL-10 and β-endorphin
release and α2δ-1 upregulation and trafficking (Bauer et al., 2010; Chen expression.
et al., 2018; Bannister et al., 2011; Yang et al., 2014). There is also
discrepancy between gabapentinoids binding with high affinities to α2δ- 2. Materials and methods
1 subunit (in nanomolar range) and their low potencies in physiological
actions (in low micromolar range), particularly clinical analgesic dosage 2.1. Drugs and chemicals
up to 3,600 mg for gabapentin (Stahl et al. 2013). In addition, alteration
of α2δ-1 upregulation and trafficking is associated with chronic appli­ Gabapentin and pregabalin were gifts from Enhua Pharmaceuticals
cation (days) of gabapentinoids, while analgesia in pain hypersensitivity (Xuzhou, Jiangsu, China) and mirogabalin was purchased from Med­
effects relatively rapid (within 30 min of gabapentinoids administration) ChemExpress (NJ, USA). Minocycline hydrochloride and lidocaine hy­
(Yang et al., 2012; Bauer et al., 2009; Chen et al., 2018). Thus gaba­ drochloride were obtained from Yuanye Biotech (Shanghai, China) and
pentinoids may exert antineuropathic pain by other mechanisms. Chengdu Pharmaceuticals Group (Chengdu, China) respectively, while
Indeed, other receptor targets have also been suggested, including so­ DPCPX and GNTI were acquired from Sigma-Aldrich (USA). CTAP,
dium channels, hyperpolarization-activated cation channels and naltrindole and β-endorphin polyclonal antiserum were bought from
particularly α2δ-1-NMDA receptor complexes (Chen et al., 2018; Tae Abcam (Cambridge, UK), and recombinant rat-IL-10 antibody was from
et al., 2017; Yang et al., 2009). R&D Systems (USA). α-helical CRF(9–41) and Exendin9-39 were
Historically, only neurons were believed to be involved in the pa­ commercially synthesized by GL Biochem (Shanghai, China) and
thology of neuropathic pain. However, microglia and microglia-neuron Shanghai TASH Biotechnology Co. (China) respectively, with peptide
interactions have been associated with initial and maintenance of syn­ contents of 98%. GW1100 and α-helical CRF(9–41) were dissolved in
aptic plasticity and central sensitization after peripheral nerve injury in 70% polyethylene glycol (PEG400) and30% dimethyl sulfoxide (DMSO)
recent years (Tiwari et al., 2014; Scholz and Woolf, 2007; Ji et al., in 0.9% normal saline (Mao et al., 2019; Apryani et al., 2020), while
2013). In contrast to microglia activation (classic role) which releases DPCPX was suspended in 10% DMSO in 0.9% normal saline.
neuroinflammatory factors such as interleukin (IL)-6, IL-1β, tumor ne­
crosis factor (TNF)-α and brain-derived neurotrophic factor (BDNF), and 2.2. Experimental animals
subsequently provokes spinal dorsal horn neurons (Kawasaki et al.
2008), alternatively activated microglia release antiinflammatory fac­ One-day-old (sex unidentified) neonatal Wistar rats and male or fe­
tors such as IL-10, IL-4, nerve growth factor (NGF), glial cell line-derived male adult rats (180–200 g) were acquired from Institute of Shanghai
neurotrophic factor (GDNF), and endogenous opioid peptides have been Experimental Animals (China). Adult rats were kept in specific plastic
recently identified to provide tissue repairing, neuroprotective and boxes (4 per cage) at typical room temperature and humidity on 12 h
analgesic actions in neuropathic pain (Wu et al., 2018a; Li et al., 2016; dark / light cycles in the Central Animal Laboratory, under ad libitum
Huang et al., 2016; Franco and Fernandez-Suarez, 2015; Carniglia et al., feeding. The animals were adapted for 3 to 5 days for laboratory at­
2017). It has been reported that Aconitum-derived bulleyaconitine A, mosphere before operations or experiments. Experimental study groups
bullatine A, lappaconitine and isotalitizindine induced pain anti­ were allocated randomly, and the researchers were blinded to the
hypersensitivity through microglial dynorphin A expression (Shao et al., behavior tests. The Animal Care and Welfare Committee of Shanghai
2020; Huang et al., 2020, 2016; Li et al., 2016; Sun et al., 2018). Also, IL- Jiao Tong University (Shanghai, China) approved the animal research
10 administered intrathecally to neuropathic rats yielded remarkable protocols, which performed according to the international animal care
inhibition of mechanical allodynia and thermal hyperalgesia through guidelines (National Institutes of health, USA).
spinal microglial β-endorphin expression (Wu et al. 2018a). Interest­
ingly, this IL-10/β-endorphin pathway accounts for pain relieve effects 2.3. Primary neuronal and glial cells cultures
caused by agonism of the GLP-1Rs, α7nAChRs, and GPR40 in numerous
rodent pain models (Apryani et al., 2020; Mao et al., 2019; Wu et al., Neuronal and glial cells were obtained from the spinal cords of 1-
2017b). day-old neonatal rats. The extracted spinal cords were digested with
Gabapentinoids analgesia has been recently implicated in microglia, 0.05% trypsin. The cells were disassociated and then suspended in
although the findings were somewhat controversial. Gabapentin was DMEM supplemented with 10% (v/v) fetal bovine serum (FBS), strep­
reported to inhibit allodynia and hyperalgesia in rat models of CFA- tomycin (100 μg/mL), and penicillin (100 U/ml). For neuronal culture,
induced chronic myositis and monoarthritis and streptozotocin- the cells were then incubated for 1.5 h in 24-well cell culture plates in
induced diabetes, which was associated with inhibition of microglial the complete DMEM. The culture medium was then changed to the

345
K.A. Ahmad et al. Brain Behavior and Immunity 95 (2021) 344–361

Neurobasal (Invitrogen) containing B27 and 0.5 mM glutamine for medial surface of each hindpaw. The hindpaw withdrawal latency was
further culture. All experiments were started after 6 day of plating. considered as the duration (seconds) of time from the start of radiant
Harvested neurons showed a purity > 85% as determined by the NeuN heat application till a sudden withdrawal response. The cut-off of 30 s
immunoreactivity. was set to avoid tissue damage. Each hindpaw was tested independently
For glial cell cultures, cells suspension was incubated in pre-coated in triplicate with five minutes intervals. The final results were computed
(poly-L-lysine, 100 µg/ml) 75-cm2 cell culture flasks (1 × 107 cells/ from the mean of three independent measurements.
flask) and maintained at 37 ◦ C in a 5% carbon dioxide cell incubator. To
prepare microglial cells, after 8 days the aliquots were obtained by 2.6. RNA extraction and real-time qPCR
shaking the cultural flasks at 260 rpm, 37 ◦ C for 2 h. The cells suspension
was then plated in new plates and unattached cells were removed by Spinal lumbar enlargements from sham or neuropathic rats were
washing with serum-free DMEM. Harvested microglial cells showed a collected and homogenized mechanically by an electronic micro-
purity > 95%, as determined by the Iba-1 immunoreactivity. homogenizer (4,000 rpm for 15 s) in TRIzol (Invitrogen, Thermo­
To prepare astrocyte cells, after 11 days culture the flasks were also fisher). The primary cultured cell samples were also obtained and lysed
shaken for 2 h followed by the incubation with 10 mL of 0.05% trypsin- in TRIzol after drug treatment. The amount of 1 µg total RNAs were then
EDTA (Invitrogen) in a cell incubator for 15 min to separate the oligo­ transcribed reversely into cDNA using a ReverTra Ace qPCR RT kit
dendrocytes from astrocytes. After the trypsin neutralization by 10 mL following the manufacturer protocols (Toyobo Co., Osaka, Japan). Real-
of the complete DMEM medium, the floating cells suspension was dis­ time qPCR was afterwards performed by utilizing the RealmasterMix
carded. A nearly intact bed layer of astrocyte cells was then subcultured SYBR Green I (Yeasen Biotechnology) in a Mastercycler ep realplex
conventionally in new plates after trypsinization. Cultured astrocytes (Eppendorf, Hamburg) in mixture with either of the following specific
showed > 90% purity as verified by the GFAP immunoreactivity. primers: 5′ -CCAAGGTCATCCATGACAAC-3′ and 5′ -TCCA­
CAGTCTTCTGAGTGGC-3′ for GAPDH (Gong et al. 2014), 5′ -
2.4. Intrathecal catheterization and injection in rats GGCTCAGCACTGCTATGTTGCC-3′ and 5′ -AGCATGTGGGTCTGGCT­
GACTG-3′ for IL-10 (Wu et al. 2017b), 5′ -CCTATCGGGTGGAGCACTTC-
A polyethylene catheter (PE-10: 0.28 mm inner diameter and 0.61 3′ and 5′ -TGGCTCTTCTCGGAGGTCAT-3′ for the β-endorphin precursor
mm outer diameter; Clay Adams, Parsippany, NJ, USA) was implanted proopiomelanocortin (POMC) (Wu et al. 2018a), and 5′ -
into the rat’s spinal cord lumbar level under standard inhaled isoflurane CCTGTCCTTGTGTTCCCTGT-3′ and 5′ -AGAGGCAGTCAGGGTGAGAA-3′
anesthesia run by an anesthesiameter (Ugo Basile Gas Anesthesia Sys­ for the dynorphin precursor prodynorphin (PDYN) (Huang et al. 2016).
tem) as described previously (Huang et al., 2012; Lu et al., 2012). The Real-time qPCR was firstly used to verify the primers specificity through
precise catheter position was validated by injecting 4% lidocaine (10 μL) the melting curves and the relative mRNA expression of the targeted
2–3 days after recovery. Rats which showed no motor impairments, for genes was then determined by the (2 − ΔΔCt) method after normali­
example paralysis and claudication following implanting of the catheter zation to the housekeeping gene GAPDH (cycle threshold) values.
and showed instant bilateral paralysis of the hindlimbs following
intrathecal injection of lidocaine were included for additional experi­ 2.7. Immunofluorescence staining
mental research. For intrathecal delivery, 10 μL of drug solution (fol­
lowed by a 15-μL saline for flushing) was injected using a 50-μL The co-labeling immunostaining of IL-10, β-endorphin, α2δ-1, and/
microliter syringes (Shanghai Anting Micro-Injector Factory, Shanghai, or microglial, astrocytic and neuronal cellular biomarkers in frozen
China). sections of spinal cords and primary culture cells were performed and
pictured under a confocal microscope (TCS SP8, Leica Microsystems,
2.5. Neuropathic pain rat model and behavior assessment Wetzlar, Germany) as described previously (Huang et al. 2016). Sham or
neuropathic rats were undergone to anesthetize by injection of pento­
The rats were undergone to L5/L6 unilateral spinal nerve ligation barbital (50 mg/kg, intraperitoneal) and intracardially perfused with
(SNL) to produce peripheral neuropathy basically as described previ­ normal saline (100 mL) and then 4% paraformaldehyde (PFA, 60 mL).
ously (Kim and Chung, 1992). Briefly, rats L5 and L6 spinal nerves were The lumbar enlargements (L3–L6) were carefully dissected and fixed
carefully uncovered and ligated with 6–0 silk thread tightly under overnight at 4 ◦ C in 4% buffered PFA. Tissues were then dehydrated
inhaled isoflurane anesthesia. After 2 spinal nerves ligated, the lumbar gradually in 10%, 20% and 30% sucrose solutions at 4 ◦ C. Frozen sec­
fascia was sutured by absorbable suture in layers and the skin was tions were prepared in OCT embedding media (Leica Microsystems) and
stitched. Rats were allocated to retrieve afterwards, and only rats which sliced into 30-μm thickness. Spinal glial and neuronal cells were cultured
showed substantial ipsilateral paws mechanical allodynia to inoffensive in 24-well plates (5 × 104 cells/well) on coverslips and then fixed by 4%
mechanical stimulus (hindlimbs withdrawal threshold in the ipsilateral buffered PFA. Both cell coverslips and frozen sections were blocked by
surgery side < 8 g) and without motor- reflex shortages were selected for incubation for 1 h in goat serum (10%, v/v) and Triton X-100 (0.5%, v/
the additional research tests. Sham rats underwent the same surgical v) in PBS. Cell coverslips and frozen sections were kept with the specific
procedures described above except without spinal nerves ligation. primary antibodies: rat IL-10 antibody (1:200; rabbit polyclonal; Pro­
To assess the mechanical allodynia, rats were firstly adapted to teinTech Group, IL, USA), rat α2δ-1 antibody (1:200; rabbit polyclonal;
testing environment for 30 min, specifically on a metal grid in trans­ ProteinTech Group) and rat β-endorphin antibody (1:200; rabbit poly­
parent plexiglass boxes. The contralateral and ipsilateral hindlimb clonal; Phoenix Pharmaceuticals, USA), as well as antibodies against
withdrawal threshold was evaluated using an electronic Von Frey primary cellular biomarkers for overnight at 4 ◦ C. Spinal glial and
monofilament (2450 CE, USA). The hand held probe contacted to a neuronal cells were observed with the following biomarkers: NeuN
monofilament No.15 generated force (0.1 to 90 g) was used to stimulate (1:100 for frozen sections or 1:200 for cell coverslips, mouse polyclonal;
base of the hindpaws. The threshold was considered as the least force to Millipore, Darmstadt, Germany) for neurons, Iba-1 (1:100 for frozen
evoke the withdrawal response of the rat’s hindpaws. The mean of sections or 1:200 for cell coverslips, Millipore) for microglia and GFAP
triplicate readings at 5 min intervals was recorded for each hindlimb at (1:200 for frozen sections and cell coverslips, mouse polyclonal; Milli­
each time point (Li et al. 2016). pore) for astrocytes. IL-10, β-endorphin and α2δ-1 were observed by the
The Hargreaves test with heated glass (IITC Life Science Inc.) was following secondary antibody, Alexa-555-conjugated goat anti-rabbit
used to assess thermal hyperalgesia. Briefly, rats were acclimatized for (1:200, Abcam). Other primary antibodies were visualized by the sec­
the testing environment by placing them in a plexiglass box for at least ondary antibody, Alexa-488-conjugated goat anti-mouse (1:200,
thirty minutes. The radiant heat source was applied on the plantar Abcam). For cell coverslips, the 49,6-diamidino-2-phenylindole (DAPI,

346
K.A. Ahmad et al. Brain Behavior and Immunity 95 (2021) 344–361

Fig. 1. Inhibitory effects of intrathecal gabapentin (A), pregabalin (B) and mirogabalin (C) on mechanical allodynia of in neuropathic rats. Dose-response curves of
gabapentin (D), pregabalin (E) and mirogabalin (F) on mechanical allodynia 1 h after injection. Inhibitory effects of intrathecal gabapentinoids on thermal anti­
hyperalgesia in neuropathic rats (G). The data are presented as means ± SEM (n = 6 per group).

Beyotime Biotechnology, Shanghai, China) staining was applied to the 2.9. β-Endorphin , dynorphin a and IL-10 measurement
cell nuclei.
To quantify the IL-10-, β-endorphin-, α2δ-1-, NeuN-, GFAP-, and Iba- The concentrations of β-endorphin, dynorphin A and IL-10 were
1-immunopositive cell, photomicrographs of the medial three fourths of measured in the spinal lumbar enlargements from sham or neuropathic
the dorsal horns (laminas I-V) were captured by the confocal microscope rats 1 h after intrathecal drug treatments. The spinal lumber enlarge­
at 10 × or 40 × magnifications. A researcher who was known the ments supernatants collected after homogenizing (4,000 rpm for 15 s) in
experimental groups calculated the positive stained regions using 10 mM Tris-hydrochloric acid (pH 7.4) and then centrifuged (4000 rpm
ImageJ Software (National Institutes of Health, USA). Background and 4 ◦ C) for 15 min. Protein concentrations were also measured by the
fluorescence was excluded and only positive co-labeling from the cell standard method, bicinchoninic acid test following the commercial kit
surfaces was included by the high and low threshold set up. The same instructions (Beyotime Biotechnology). β-endorphin or IL-10 concen­
ImageJ software settings was applied for all control and treated groups. trations were detected in neuronal, astrocytic, and microglial culture
The mean percentage of the co-labeled areas was the positive fraction of cells. Cells were plated in 24-well culture plates (5 × 105 cells per well)
the total measured image region from 3 non-adjacent slices of every and gently washed before drug treatments with warm DMEM (1 mL)
spinal cord (Huang et al., 2016; Wu et al., 2017b). containing bovine serum albumin (2 mg/mL) and N-(2-hydroxyethyl)
piperazine-N0-2-ethanesulfonic acid (HEPES, 15 mmol/L). Cells were
2.8. Western blot then treated with the experimental drugs for 2 h and sample superna­
tants were preceded. According to the manufacturer instructions, spe­
Western blot was performed as described previously by (Fan et al. cific enzyme-linked fluorescent immunoassay kits were used to measure
2016). Briefly, the spinal lumbar enlargements were homogenized and the β-endorphin, dynorphin A (Phoenix Pharmaceuticals) and IL-10
proteins were extracted using the radio-immunoprecipitation assay (eBioscience) concentrations which were validated by running stan­
buffer containing 1% protease inhibitor phenylmethylsulfonyl fluoride. dard curves with the testing samples simultaneously to determine the
The lysates were centrifuged for 15 min at 12,000 rpm at 4 ◦ C. The unknown concentrations. The relative fluorescence units were detected
protein concentration of samples was measured using the bicinchoninic by a Microplate Reader (ThermoLab systems, Finland) and the IL-10,
acid assay (Beyotime Biotechnology). The proteins were separated by β-endorphin or dynorphin A concentrations were determined by com­
10% sodium dodecyl sulfate–polyacrylamide gel electrophores and then parisons with the calibration curves performed simultaneously.
transferred to polyvinylidene fluoride membranes by an electrophoresis
technique. The membranes were blocked in 5% skim milk powder in
2.10. Data calculation and statistical evaluation
Tris-based saline containing 0.1% Tween 20 at room temperature for 1
h, and then incubated with primary antibodies raised against the α2δ-1
To compute percentage of the maximal possible effect (% MPE) the
subunit (1:1000, #27452–1-AP, rabbit polyclonal, ProteinTech Group,
following formula was applied: (post-drug threshold in ipsilateral hin­
USA) and GAPDH (1:5000, #6004–1-Ig, mouse polyclonal, ProteinTech
dlimb – baseline threshold in ipsilateral hindlimb) / (baseline threshold
Group, USA) overnight at 4 ◦ C with gentle shanking. Protein bands were
in contralateral hindlimb –baseline threshold in ipsilateral hindlimb) ×
visualized using the Odyssey Infrared Imaging system (Li-Cor Bio­
100. The % MPE values near to 100 were regarded as normal mechanical
sciences, USA) after 1-hour incubation at room temperature with the
thresholds in the contralateral hindpaws, while values closed to 0 indi­
goat anti-rabbit IgG, Dylight 800-conjugate (1:10,000, #5151, Cell
cate mechanical allodynia in the ipsilateral hindpaws. For analysis of the
Signaling Technology Inc. USA) or goat anti-mouse IgG, Dylight 680-
dose–response curves, the parameters, i.e., the minimum effect,
conjugate (1:10,000, #5470, Cell Signaling Technology Inc. USA).
maximum effect (Emax), half-effective concentration or dose (EC50 or
Bands intensities were quantitated using the image analysis software
ED50), and Hill coefficient (n), were calculated by fitting nonlinear least-
(ImageJ Software, National Institutes of Health). The relative expression
squares curves to the relation Y = a + bx, where × = [C]n/(ECn50 + [C]n)
of the target protein was calculated after normalization to the GAPDH
or [D]n/(EDn50 + [D]n). The values of EC50 or ED50 and b (Emax) were
level. The experiments were repeated at least treble.
projected by yielding a minimum residual sum of squares of deviations
from the theoretical curve (Wang and Pang 1993).

347
K.A. Ahmad et al. Brain Behavior and Immunity 95 (2021) 344–361

Fig. 2. Effects of gabapentinoids intrathecally on IL-10, POMC, PDYN mRNA expression (A-C), and IL-10, β-endorphin and dynorphin A protein expression (D-F) in
neuropathic rats. Spinal lumbar enlargements were obtained 1 h after injection. Data are shown as means ± SEM (n = 6 per group). * P < 0.05 compared with the
control group, by one-way ANOVA followed by the post-hoc Bonferroni test.

Data are summarized as means ± SEM or 95% confidence limits. The 244.7 nmol) and 66% MPE, 6.2 µg or 38.9 nmol (95% confidence limits:
significance of the differences was calculated by unpaired and two-way 4.8 to 7.8 µg or 30.1 to 49.0 nmol) and 61% MPE, and 1.5 µg or 7.2 nmol
Student t-test or one-way or repeated-measures two-way ANOVA using (95% confidence limits: 1.1 to 2.0 µg or 5.2 to 9.6 nmol) and 65% MPE,
GraphPad Prism (version 8.0.1, GraphPad Software, Inc., USA). The respectively (Fig. 1D-1F). Gabapentinoids-inhibited thermal hyper­
post-hoc Bonferroni test was used when the effect of the drug (dose) (for algesia was also assessed in neuropathic rats. Four groups of SNL rats
the one-way ANOVA, the factor was drug [dose]; for the two-way received intrathecal injection of saline (10 µL), gabapentin (100 µg),
ANOVA, the factors were drug [dose], time, and their interaction) was pregabalin (30 µg) or mirogabalin (10 µg). Hindpaw withdrawal la­
statistically significant. Probability values were two-tailed and the sta­ tencies were measured with 5-min intervals before, and 0.5, 1, 2, and 4 h
tistical significance criterion p-value was 0.05. after intrathecal drug injection. As shown in Fig. 1G, spinal nerve injury
induced notable thermal hyperalgesia in the ipsilateral hindpaws
3. Results compared to the contralateral hindpaws. Intrathecal saline injection (10
µL) did not influence paw withdrawal latencies in both hindpaws.
3.1. Gabapentinoids reduced mechanical allodynia and thermal However, gabapentin, pregabalin and mirogabalin significantly inhibi­
hyperalgesia in neuropathic rats ted thermal hyperalgesia in the ipsilateral hindpaws (P < 0.05, by
repeated-measures two-way ANOVA followed by the post-hoc Bonfer­
A single injection of normal saline (10 µL) or gabapentin (3, 10, 30, roni test), without affecting withdrawal latencies in the contralateral
100 or 300 µg), pregabalin (1, 3, 10, 30 or 100 µg) or mirogabalin (0.3, hindpaws.
1, 3, 10 or 30 µg) administrated intrathecally to six groups of neuro­
pathic rats. The paw withdrawal responses were afterwards measured
with 5-min intervals before, and 0.5, 1, 2, and 4 h after the intrathecal 3.2. Gabapentinoids specifically stimulated spinal microglial IL-10 and
drug injection. As shown in Fig. 1A-1C, spinal nerve ligation remarkably β-endorphin expression
provoked mechanical allodynia in the ipsilateral hindpaws, compared
with the contralateral hindpaws. After intrathecal administration of To assess the effects of gabapentinoids in the expression of spinal IL-
normal saline, hindpaw withdrawal responses in neuropathic rats 10, β-endorphin and dynorphin A, four groups of neuropathic rats were
remained unchanged. Whereas intrathecal injection of gabapentinoids injected intrathecally with normal saline (10 μL), gabapentin (100 μg),
augmented withdrawal thresholds dose- and time-dependent in the pregabalin (30 μg) or mirogabalin (10 μg), and spinal lumber enlarge­
ipsilateral hindpaws, with peak effect at 1 h after injection (P < 0.05, by ments were isolated 1 h after intrathecal administration. Measured by
repeated-measures two-way ANOVA followed by the post-hoc Bonfer­ RT-qPCR, gabapentinoids considerably increased the mRNA IL-10 by
roni test), without affecting withdrawal thresholds in the contralateral 80% (Fig. 2A) and POMC expression by 77% (Fig. 2B), but failed to
hindpaws. The dose–response analyses were computed from the % MPE significantly up-regulate the gene expression of PDYN (Fig. 2C). Also,
values of each drug obtained 1 h after injection. The ED50 and Emax the effects of gabapentinoids in the IL-10, β-endorphin and dynorphin A
values projected for gabapentin, pregabalin and mirogabalin were 30.3 expression were further assessed by commercial fluorescent immuno­
µg or 176.9 nmol (95% confidence limits: 21.9 to 41.9 µg or 127.9 to assay kits. The baseline protein levels of IL-10, β-endorphin and
dynorphin were 6.4 ± 1.9, 22.5 ± 2.4 and 20.6 ± 3.8 pg/mg respectively

348
K.A. Ahmad et al. Brain Behavior and Immunity 95 (2021) 344–361

Fig. 3. Stimulatory effects of gabapentin administered intrathecally on spinal IL-10 microglial expression in neuropathic rats. Co-labeling immunostaining of IL-10
antibody with microglial (Iba-1) marker, astrocytic (GFAP) marker or neuronal (NeuN) marker in spinal dorsal horn (scale bar: 250 µm) and laminae I-V (scale bar:
50 µm, A-F). Yellow co-labeling of IL-10 and cellular markers shown by arrows. G-L. Stimulatory effect of gabapentin. The % immunolabeled surface areas of IL-10/
Iba-1 (M), IL-10/GFAP (N) and IL-10/NeuN (O) from the spinal dorsal horn laminae I-V were quantified using ImageJ software. Data are presented as means ± SEM
(n = 6 per group). * P < 0.05 compared with control group by unpaired and two-tailed Student t-test. (For interpretation of the references to colour in this figure
legend, the reader is referred to the web version of this article.)

in the spinal homogenates from the saline treated neuropathic rats. Quantitatively, gabapentin treatment augmented co-labeling of
Gabapentinoids intrathecally significantly increased expression of IL-10 β-endorphin/Iba-1 by 3.1-fold as measured by ImageJ Software
by 165% (Fig. 2D) and β-endorphin by 86% (Fig. 2E) but not dynorphin (Fig. 4M), but not β-endorphin/ GFAP (Fig. 4N) or β-endorphin/NeuN
A (Fig. 2F). (Fig. 4O).
To determine the spinal cell types that specifically elevated IL-10 To compare gabapentinoids-induced antihypersensitivity and stim­
expression after gabapentin intrathecal treatment, double label immu­ ulation of spinal IL-10 and β-endorphin expression, six groups of male
nostaining was applied by co-labeling of IL-10 with the cellular bio­ sham rats and male and female SNL rats were intrathecally injected with
markers of neurons (NeuN), astrocytes (GFAP), and microglia (Iba-1) in saline (10 µL) or gabapentin (100 µg). One hour later, the hindpaw
spinal frozen sections of neuropathic rats. Two groups received normal withdrawal thresholds or latencies were measured in the ipsilateral
saline (10 μL) or gabapentin (100 μg) intrathecally and the spinal lumber hindpaws. As shown in Fig. 5A and 5B, compared to saline treatment,
enlargements were obtained 1 h after injection. As shown, IL-10 staining intrathecal gabapentin failed to produce significant changes in with­
was co-localized widely with Iba-1 (Fig. 3A, 3B) of ipsilateral spinal drawal thresholds or latencies in male sham rats. In contrast, gabapentin
dorsal horns (I-V laminae), but less with the GFAP (Fig. 3C, 3D) or NeuN inhibited mechanical allodynia and thermal hyperalgesia in both male
(Fig. 3E, 3F). Gabapentin (100 µg) intrathecally stimulated IL-10 im­ and female SNL rats by nearly the same degrees. Rats were immediately
munostaining with Iba-1 (Fig. 3G, 3H), but did not stimulate the co- killed and spinal lumber enlargements were collocated after behavioral
labeling of IL-10/GFAP (Fig. 3I, 3 J) or IL-10/NeuN (Fig. 3K, 3L). The tests for IL-10 and β-endorphin gene and protein quantification.
double immunofluorescences staining of IL-10 and cellular biomarkers Compared to saline control, intrathecal gabapentin (100 µg) signifi­
were quantitatively measured to calculate immunolabeled area fractions cantly increased IL-10 mRNA expression by 60%, 76% and 78%, and
in spinal dorsal horns (laminae I-V) by ImageJ Software. Compared with POMC mRNA expression by 68%, 71% and 72% in male sham rats, male
saline groups, gabapentin significantly increased the co-labeling im­ and female SNL rats, respectively (Fig. 5C, 5D). In addition, treatment
munostaining of IL-10/Iba-1 by 2.9-fold, but not IL-10/GFAP (Fig. 3N) with gabapentin significantly increased IL-10 protein expression by
or IL-10/NeuN (Fig. 3O). 140%, 165% and 160%, and β-endorphin protein expression by 66%,
Additionally, we also determined the specific-cell types which 86% and 89% in these animals, respectively (Fig. 5E, 5F). Gabapentin-
elevated the β-endorphin expression after gabapentin treatment. Co- stimulated microglial IL-10 expression was further compared using the
labeling of β-endorphin with NeuN, GFAP and Iba-1 was performed in immunostaining technique. Frozen sections from the ipsilateral spinal
the spinal frozen sections from the aforementioned two groups of cords were co-labeled with the IL-10 antibody and microglial biomarker
neuropathic rats. β-Endorphin labeling was co-localized with Iba-1 Iba-1 antibody. As shown in Fig. 5G, intrathecal gabapentin remarkably
(Fig. 4A, 4B), GFAP (Fig. 4C, 4D) NeuN (Fig. 4E, 4F) in the ipsilateral increased co-immunostaining of IL-10/Iba-1 in male sham, and male and
dorsal horns (I–V laminae) of the neuropathic rats. Gabapentin mark­ female SNL rats. Quantitatively by using the ImageJ computer software,
edly promoted co-labeling of β-endorphin/Iba-1 (Fig. 4G, 4H), but not gabapentin-increased IL-10/Iba-1 colocalization in these groups of rats
β-endorphin/GFAP (Fig. 4I, 4 J) or β-endorphin/NeuN (Fig. 4K, 4L). was 140%, 150% and 149%, respectively (Fig. 5H).

349
K.A. Ahmad et al. Brain Behavior and Immunity 95 (2021) 344–361

Fig. 4. Stimulatory effects of gabapentin administered intrathecally on spinal β-endorphin microglial expression in neuropathy rats. Co-labeling immunostaining of
β-endorphin antibody with microglial (Iba-1) marker, astrocytic (GFAP) marker or neuronal (NeuN) marker in spinal dorsal horn (scale bar: 250 µm) and laminae I-V
(scale bar: 50 µm, A-F). Yellow co-labeling of β-endorphin and cellular markers shown by arrows. G-L. Stimulatory effect of gabapentin. The % immunolabeled
surface areas of β-endorphin/Iba-1 (M), β-endorphin/GFAP (N), and β-endorphin/NeuN (O) from spinal dorsal horn laminae I-V were quantified using ImageJ
software. The data are shown as means ± SEM (n = 6 per group). * P < 0.05 compared with the control group by unpaired and two-tailed Student t-test. (For
interpretation of the references to colour in this figure legend, the reader is referred to the web version of this article.)

3.3. Gabapentinoids specifically stimulated spinal microglial IL-10 and expression of IL-10 and POMC, whereas pretreatment of minocycline
β-endorphin expression (60 µM) for 1 h did not affect the baseline of either gene expression.
However, minocycline entirely inhibited gabapentin-stimulated IL-10
To additionally verify the specific stimulatory effects of gabapenti­ and POMC gene expression (Fig. 8A-8B). Likewise, minocycline blocked
noids on spinal microglial IL-10 and POMC gene expression, primary gabapentin-increased the protein levels of IL-10 and β-endorphin
microglial culture cells were incubated with vehicle, gabapentin, pre­ (Fig. 8C-8D).
gabalin and mirogabalin for 2 h. The cellular IL-10 and POMC mRNA
expression were measured by RT-qPCR. Treatment with gabapentin 3.4. Intrathecal microglial inhibitor, Il-10 antibody and β-endorphin
(100 µM), pregabalin (30 µM) and mirogabalin (10 µM) promoted the IL- antiserum and μ-opioid receptor (MOR) antagonist suppressed
10 gene expression of microglial, but not astrocytic or neuronal cells gabapentinoids-inhibited mechanical allodynia
(Fig. 6A-6C). The same specific stimulatory effects of gabapentinoids on
POMC expression were also observed in primary microglial but not To test the fundamental role of spinal microglial expression of IL-10
astrocytic or neuronal cells (Fig. 6D-6F). Further dose–response analysis and β-endorphin in gabapentinoids alleviated mechanical allodynia, the
indicated that gradient concentrations of gabapentin (3, 10, 30,100 and microglial inhibitor minocycline was first applied. Two groups of
300 µM), pregabalin (1, 3, 10, 30 and 100 µM) and mirogabalin (0.3, 1, neuropathic rats were firstly injected intrathecally with saline (10 μL) or
3, 10 and 30 µM) concentration-dependently upregulated the mRNA minocycline (100 μg), followed by second injection intrathecally of
expression of IL-10 and POMC in primary microglia with EC50 values of gabapentin (100 μg), pregabalin (30 μg) or mirogabalin (10 μg) 4 h later.
41.3, 11.5 and 2.5 µM and 34.7, 13.3 and 2.8 µM, respectively, both of The dose regimens used in minocycline treatment were from previous
which were overlapped (Fig. 6G-6I). reports (Mei et al., 2011; Tang et al., 2020; Shoaib et al., 2019). The
The stimulatory effects of gabapentinoids on the IL-10 and hindpaw withdrawal thresholds to mechanical stimuli were evaluated
β-endorphin levels in primary microglial cultured cells were also before and 0.5, 1, 2, and 4 h after gabapentinoids treatment. As shown in
compared with primary spinal astrocytes and neurons using commercial Fig. 9A-9C, intrathecal gabapentin, pregabalin and mirogabalin time-
fluorescent immunoassay kits. Treatment with the vehicle, gabapentin dependently inhibited mechanical allodynia. Minocycline did not
(100 µM), pregabalin (30 µM) and mirogabalin (10 µM) for 2 h specif­ significantly alter the baseline withdrawal thresholds in the contralat­
ically increased the IL-10 levels by approximately 3-fold (Fig. 7A-7C) eral or ipsilateral hindpaws, but entirely blocked gabapentinoids-
and β-endorphin by approximately 2.8-fold (Fig. 7D-7F) in the culture inhibited allodynia.
medium of microglial cells, but not of astrocytes or neurons. In addition, two groups of neuropathy rats were firstly injected
To further ascertain the stimulatory effect of gabapentin on micro­ intrathecally with saline (10 μL) or the IL-10 antibody (2 μg), followed
glial IL-10 and β-endorphin expression, the microglial inhibitor mino­ by second injection intrathecally of gabapentin (100 μg), pregabalin (30
cycline was applied in the primary microglial cells. Treatment of μg) or mirogabalin (10 μg) 0.5 h later. Intrathecal gabapentinoids time-
microglial cells with gabapentin (100 µM) for 2 h increased the gene dependently inhibited mechanical allodynia. Although it did not alter

350
K.A. Ahmad et al. Brain Behavior and Immunity 95 (2021) 344–361

Fig. 5. Effects of intrathecal gabapentin on mechanical allodynia and thermal hyperalgesia (A, B), IL-10 and POMC mRNA expression (C, D), IL-10 and β-endorphin
expression (E, F), co-labeling of IL-10 with the microglial marker Iba-1 in spinal dorsal horns laminae I-V (scale bar: 50 µm) and the % immunolabeled surface areas
of IL-10/Iba-1 (G, H) in male sham rats and male and female neuropathic rats. The data are presented as means ± SEM (n = 6 per group). * P < 0.05 compared with
saline group, by unpaired and two-tailed Student t-test.

the baseline withdrawal responses in the contralateral or ipsilateral saline (10 µL), the MOR antagonist CTAP (10 µg), κ-opioid receptor
hindpaws, IL-10 antibody completely prevented gabapentinoids from (KOR) antagonist GNTI (50 µg) or δ-opioid receptor (DOR) antagonist
countering mechanical allodynia (Fig. 9D-9F). naltrindole (5 µg) intrathecally, followed by another injection of gaba­
Furthermore, two groups of neuropathy rats were injected firstly pentin (100 µg), pregabalin (30 µg) or mirogabalin (10 µg) intrathecally
with the blank serum (1:10, 10 μL) or the β-endorphin antiserum (1:10, 0.5 h later. The doses and regimen of CTAP, GNTI and naltrindole were
10 μL) intrathecally, followed by the another injection of gabapentin based on the previous publication (Steinmiller and Young, 2008). As
(100 μg), pregabalin (30 μg) or mirogabalin (10 μg) intrathecally 0.5 h shown in Fig. 10D-10F, intrathecal pretreatment with CTAP did not have
later. The intrathecal pretreatment with the β-endorphin antiserum did significant effect on the response of withdrawal in the contralateral or
not affect the control withdrawal responses in the contralateral or ipsi­ ipsilateral hind paws, but nearly wholly attenuated spinal
lateral hindpaws, but considerably attenuated gabapentinoids-inhibited gabapentinoids-inhibited mechanical allodynia. In contrast, GNTI or
mechanical allodynia (Fig. 10A-10C). naltrindole failed to block gabapentinoids-inhibited mechanical
Lastly, four groups of neuropathy rats injected firstly with normal allodynia.

351
K.A. Ahmad et al. Brain Behavior and Immunity 95 (2021) 344–361

Fig. 6. Effects of gabapentin, pregabalin and mirogabalin on IL-10 (A-C) and POMC (D-F) mRNA expression in microglial, astrocytic and neuronal cells. G-H
Concentration-response curves of gabapentinoids on IL-10 and POMC mRNA expression. Cells were incubated with gabapentinoids for 2 h and mRNA expression of
IL-10 and POMC were evaluated by using real-time qPCR. The data are presented as means ± SEM (n = 6 per group). * P < 0.05 compared with control group, by one-
way ANOVA followed by the post-hoc Bonferroni test.

3.5. No expression of the α2δ-1 subunit of the voltage-gated calcium localized with NeuN-positive neurons (Fig. 12A, 12B). However, α2δ-1
channel in microglia was not expressed in GFAP-positive astrocytes (Fig. 12C, 12D) or Iba-1-
positive microglial cells (Fig. 12E, 12F). Moreover, treatment with
To figure out the target molecule responsible for gabapentinoids- gabapentin for 2 h did not affect the expression of α2δ-1 in neurons
induced microglial IL-10 and β-endorphin expression, specific cellular (Fig. 12G, 12H), astrocytes (Fig. 12I, 12 J) or microglial cells (Fig. 12K,
expression of the α2δ-1 subunit of voltage-gated calcium channel was 12L). Furthermore, quantitative measurement confirmed that α2δ-1 was
first assessed in the ipsilateral spinal cords frozen sections, using co- expressed in neurons but not astrocytes or microglial cells, and gaba­
labeling immunofluorescence of α2δ-1 with NeuN, GFAP or Iba-1 anti­ pentin treatment did not alter the α2δ-1 expression either in neurons,
bodies. Two groups of neuropathic rats received an intrathecal injection astrocytes or microglial cells (Fig. 12M-12O).
of normal saline (10 μL) or gabapentin (100 μg) and the spinal lumbar Finally, to test whether gabapentinoids-inhibited allodynia was
enlargements were obtained 1 h after injection. The α2δ-1 immunoflu­ through activation of the spinal GLP-1Rs, GPR40, CRF receptors or
orescence was co-localized with NeuN in the spinal dorsal horn adenosine A1 receptors, their specific antagonists were applied. The
(Fig. 11A, 11B), but not with GFAP (Fig. 11C, 11D) or Iba-1 (Fig. 11E, doses and regimen of the GLP-1R antagonist exendin9-39 (Göke et al.
11F). Intrathecal injection of gabapentin did not alter the immuno­ 1993), GPR40 antagonist GW1100 (Nakamoto et al. 2012), CRF receptor
staining of α2δ-1 with NeuN (Fig. 11G, 11H), GFAP (Fig. 11I, 11 J) or antagonist α-helical CRF(9–41) (Sawamura et al. 2003) and adenosine
Iba-1 (Fig. 11K, 11L), which was also confirmed by quantitative mea­ A1 receptor antagonist DPCPX (Park and Jun, 2008) were based on the
surement of α2δ-1/NeuN (Fig. 11M), α2δ-1/GFAP (Fig. 11N) or α2δ-1/ previous papers. Twelve groups of neuropathy rats injected firstly saline
Iba-1 (Fig. 11O) using the ImageJ Software. Additionally, western blot (10 µL), exendin9-39 (2 µg), GW1100 (300 µg) and α-helical CRF9-41
analysis also confirmed the expression of α2δ-1 in the spinal homoge­ (20 µg) intrathecally, followed by another injection of gabapentin
nates of neuropathic rats and intrathecal gabapentin (100 µg) did not (100 µg), pregabalin (30 µg) or mirogabalin (10 µg) 0.5 h later. In
significantly influence α2δ-1 protein expression compared to saline comparison with the control, intrathecal pretreatment with exendin9-
control (Fig. 11P). 39, GW1100 or α-helical CRF9-41 did not significantly reduce the
Likewise, double immunofluorescence staining was performed in inhibitory effects of gabapentin (Fig. 13A), pregabalin (Fig. 13B) or
primary cultured spinal cells. Primarily cultured cells were incubated mirogabalin (Fig. 13C) on mechanical allodynia. In addition, six groups
with the vehicle or gabapentin (100 µM) for 2 h. As shown, α2δ-1 was co- of neuropathy rats injected firstly saline (10 µL) or DPCPX (10 µg)

352
K.A. Ahmad et al. Brain Behavior and Immunity 95 (2021) 344–361

Fig. 7. Effects of gabapentin, pregabalin and mirogabalin on IL-10 (A-C) and β-endorphin (D-F) protein expression in microglial, astrocytic and neuronal primary
culture cells. The cultured medium was obtained 2 h after gabapentinoids incubation. The data are presented as means ± SEM (n = 6 per group). * P < 0.05 compared
with the control group, by one-way ANOVA followed by the post-hoc Bonferroni test.

intrathecally, followed by another injection of gabapentin (100 µg), shown that microglia release antiinflammatory cytokines such as IL-10,
pregabalin (30 µg) or mirogabalin (10 µg) 0.5 h later. As shown in IL-4, NGF, GDNF, and opioid peptides such as β-endorphin and dynor­
Fig. 13D-13F, intrathecal prior-treatment with DPCPX failed to inhibit phin A, which inhibit sensory transmission and central sensitization
gabapentinoids-inhibited mechanical allodynia. leading to pain relieve in neuropathic pain (Wu et al., 2017a, 2017b,
2018a; Gong et al., 2014; Franco and Fernandez-Suarez, 2015; Carniglia
4. Discussion et al., 2017). IL-10 is considered as the most prominent antiin­
flammatory and immunosuppressive cytokine so far, through binding to
Gabapentinoids have been extensively tested in various animal IL-10 receptor-α (Sabat et al., 2010; Glocker et al., 2011). IL-10 displays
models of nociceptive pain and pain hypersensitivity, particularly in marked antinociceptive activities in spinal nerve pain, chronic
neuropathic pain (Cheng and Chiou 2006). However, there have been constriction injury, chemotherapy, peripheral diabetic pain, bone can­
few reports that compared their dose–response curves head to head. Our cer pain and CFA-induced inflammatory pain (Wu et al., 2018a; Milligan
results demonstrated that a single intrathecal injection of each gaba­ et al., 2005; Ledeboer et al., 2007; Zhou et al., 2008; Kim et al., 2011;
pentinoid quickly inhibited mechanical allodynia and thermal hyper­ Thakur et al., 2016). It has been documented that IL-10 relieves nerve
algesia in the ipsilateral hind paws of neuropathic rats. Gabapentinoids pain via promotion of microglial β-endorphin expression (Wu et al.
exhibited time-dependent inhibition of allodynia, with peak 1 h after 2018a).
intrathecal administration. Their dose-dependent inhibitions of me­ Our current data showed that treatment with gabapentin, pregabalin
chanical allodynia were projected with Emax values 66%, 61% and 65% and mirogabalin for 2 h specifically stimulated IL-10 mRNA and protein
MPE, and ED50 values 30.3 µg (176.9 nmol), 6.2 µg (38.9 nmol) and 1.5 expression of microglial cells. Their stimulation on IL-10 expression was
µg (7.2 nmol) for gabapentin, pregabalin and mirogabalin respectively. not observed in astrocytic or neuronal cells. It is known that IL-10
The results indicate that mirogabalin is the most potent compound with treatment or gene transfer stimulated microglial β-endorphin expres­
the ED50 value being 4.4- and 23.6-fold less than that of pregabalin and sion and not vice versa (Wu et al. 2017b), or upregulated POMC in
gabapentin respectively, in agreement with the previous findings that arcuate nucleus in hypothalamus in leptin-substituted rats (Nakata et al.
mirogabalin produced more potent analgesic effects than pregabalin in 2016). Indeed, treatment of microglial cells with gabapentinoids spe­
the rat models of peripheral diabetic neuropathy and spinal cord injury cifically stimulated expression of β-endorphin but not dynorphin A.
(Domon et al., 2018a, 2018b; Domon, Kitano, and Makino 2018) and Additionally, concentration–response analysis showed the EC50 values
pregabalin exhibited higher potency analgesia than gabapentin in a for gabapentin, pregabalin and mirogabalin to promote IL-10 gene were
clinical trial (Bockbrader et al. 2010). 41.3, 11.5 and 2.5 µM respectively, which were overlapped with their
Remarkably, our study illustrated that gabapentinoids alleviate EC50 values 34.7, 13.3 and 2.8 µM of POMC gene upregulation. These
mechanical allodynia through de novo IL-10 and subsequent β-endor­ overlapped EC50 values were much greater than their α2δ-1 binding
phin microglial expression in neuropathic pain. Microglia are resident affinities in nanomolar range (Domon et al., 2018a, 2018b; Li et al.,
macrophages in the spinal cord and involved pain process. It has been 2011; Suman-Chauhan et al., 1993) but close to those of physiological

353
K.A. Ahmad et al. Brain Behavior and Immunity 95 (2021) 344–361

Fig. 8. Inhibitory effect of the microglial


inhibitor minocycline on gabapentin-
stimulated gene (A, B) and protein expres­
sion (C, D) of IL-10 and β-endorphin in pri­
mary cultures of microglia. Minocycline was
applied 1 h before gabapentin treatment,
and microglial cells and the cultured me­
dium were obtained 2 h later. The data are
presented as means ± SEM (n = 6 per
group). *, # P < 0.05 compared with the
control or gabapentin groups respectively,
by one-way ANOVA followed by the post-
hoc Bonferroni test.

Fig. 9. Inhibitory effects of intrathecally microglial inhibitor minocycline (A-C) and specific IL-10 antibody (D-F) on spinal gabapentinoids-inhibited mechanical
allodynia in neuropathic rats. Minocycline was given 4 h before gabapentinoids treatment, while the IL-10 antibody was injected 0.5 h prior to gabapentinoids
treatment. The data are shown as means ± SEM (n = 6 per group). * P < 0.05 vs. the saline + gabapentin, pregabalin or mirogabalin group, by repeated measures
two-way ANOVA followed by the post-hoc Bonferroni test.

354
K.A. Ahmad et al. Brain Behavior and Immunity 95 (2021) 344–361

Fig. 10. Blockage effects of intrathecally β-endorphin antiserum (A-C) and opioid receptor subtype antagonists (D-F) on spinal gabapentinoids-inhibited mechanical
allodynia in neuropathic rats. The β-endorphin antiserum and opioid receptor antagonists CTAP, GNTI, and naltrindole were administrated intrathecally 0.5 h before
spinal gabapentinoids administration. The data are presented as means ± SEM (n = 6 per group). * P < 0.05 vs. the saline + gabapentin, pregabalin or mirogabalin
group, by repeated multiple measures two-way ANOVA followed by the post-hoc Bonferroni test.

actions, which are usually in the low micromolar range (Stahl et al. 2017; Huang et al., 2017). Remarkably, gabapentinoids stimulated IL-10
2013). Further, the EC50 values of these three gabapentinoids on IL-10 or and β-endorphin expression in primary microglial cells in resting states
POMC gene expression were significantly correlated to their ED50 values (without LPS stimulation). Also, pretreatment with minocycline entirely
on mechanical allodynia (r = 0.999 or 0.988, n = 3 each, P = 0.002 or inhibited gabapentin-stimulated IL-10 and β-endorphin expression in
0.024). The positive correlation for gabapentinoids to stimulate IL-10/ primary cultures of microglia. Furthermore, intrathecal gabapentin
β-endorphin and to inhibit mechanical allodynia was further supported induced expression of spinal IL-10 and β-endorphin in sham and
by their spinal stimulation with different doses. Intrathecal injection of neuropathic rats by nearly the same degrees. However, spinal gaba­
gabapentin (100 µg), pregabalin (30 µg) and mirogabalin (10 µg), by the pentin did not significantly affect nociceptive behaviors in sham rats,
same degrees, stimulated fast gene and protein expression (within 1 h of conceivably due to lower peripheral neuronal sensitivity to endogenous
injection) of β-endorphin and IL-10 (but not dynorphin A) in spinal opioid peptides in normal nociceptive circumstances in which central
neuropathy rats, which was parallel to their time-course of allodynia. sensitization is not present (Gong et al. 2014). This observation is
Co-labeling immunostaining further indicated that gabapentin upregu­ consistent with previous findings where GLP-1 receptor agonists,
lated β-endorphin and IL-10 in spinal microglial, but was not observed in α7nAChR agonists, GPR40 agonists and electroacupuncture induced
astrocytic or neuronal cells. Our outcomes are in harmony with previous microglial IL-10 and β-endorphin expression and aconitines stimulated
reports that intrathecal gabapentin significantly increased spinal IL-10 dynorphin A expression by almost same degrees in primary microglial
expression and subsequently decreased proinflammatory cytokines cells in the presence and absence of LPS, and in both contralateral and
expression in neuropathic rats and morphine tolerant rats (Lee et al., ipsilateral spinal cords of neuropathic rats (Mao et al., 2019; Apryani
2013; Bao et al., 2014b, 2014a). et al., 2020; Gong et al., 2014; Ali et al., 2020; Fan et al., 2015; Huang
Microglia activated by nerve injury, trauma or lipopolysaccharide et al., 2016; Li et al., 2016). In addition, sexual dimorphism has been
(LPS) treatment react quickly by promoting the classical proin­ suggested in nociceptive role of microglia, where nerve injury-induced
flammatory system (M1 phenotype). M1 polarized microglia secret large spinal proinflammatory cytokine expression and mechanical allodynia
amounts of inflammatory factors implicated in the progress of neuro­ hypersensitivity were involved only in male animals (Sorge et al. 2015).
pathic pain (Milligan and Watkins, 2009; Perry and Holmes, 2014). However, intrathecal gabapentin produced nearly similar pain anti­
Reactive microglia undergo transformations in phenotype, including hypersensitivity and spinal microglial IL-10 and β-endorphin expression
activation of the p38-MAPK (probably p38α-MAPK subtype) signaling irrespective of the sex. Our findings are in agreement with previous
pathway in neuropathic pain, which can be inhibited by minocycline studies where no sexual dimorphism is found in microglia-related anti­
(Hua et al., 2005; Wu et al., 2017a, 2017b, 2018a). In contrast, microglia hypersensitive effects on peripheral neuropathic pain, e.g., IL-10 (Wu
can develop an alternative beneficial system (M2 phenotype), charac­ et al. 2018a), α7nAChR agonists (Apryani et al. 2020) and electro­
terized by the expression of the anti-inflammatory and neuroprotective acupuncture (Ali et al. 2020), or genetic deficiency of the microglial
factors such as IL-10, TGF-β, IL-4, and endogenous opioids peptides selective molecules including P2Y12 (Gu et al. 2016), CX3CR1 (Stani­
(Franco and Fernandez-Suarez, 2015; Wu et al., 2017a, 2017b, 2018b; land et al. 2010) and TMEN16F (Batti et al. 2016). Furthermore, various
Huang et al., 2016; Li et al., 2016). Expression of the anti-inflammatory female chronic pain models have shown pain inhibitory effects of
and neuroprotective factors may not require microglial activation microglial TLR4 knockdown (Lan et al. 2010), CX3CR1 antibody (Hu
(p38α-MAPK), but needs p38β-MAPK activation which can also be et al. 2012), cathepsin S inhibitor (Nieto et al. 2016), and P2X7 antag­
inhibited by minocycline (Wu et al., 2017a, 2017b, 2018b; Li et al., onists (Yang et al., 2015; Nieto et al., 2016) similar to males. Our results

355
K.A. Ahmad et al. Brain Behavior and Immunity 95 (2021) 344–361

Fig. 11. Effects of gabapentin (100 µg) administered intrathecally on spinal α2δ-1 expression in neuropathic rats. Co-labeling immunostaining of α2δ-1 with the
neuronal (NeuN) marker, astrocytic (GFAP) marker or microglial (Iba-1) marker in the spinal dorsal horn (scale bar: 250 µm) and the I-V laminate (scale bar: 50 µm,
A-F). Yellow co-labeling of α2δ-1 and cellular markers shown by arrows. G-L. Effect of gabapentin (100 µg), given intrathecally for 1 h, on α2δ-1 expression in
neurons, astrocytes or microglial cells. The % immunolabeled surface areas of α2δ-1/NeuN (M), α2δ-1/GFAP (N), and α2δ-1/Iba-1 (O) from spinal dorsal horn
laminae I-V were determined by the ImageJ software. Western blot of α2δ-1 protein expression (P). Data are shown as means ± SEM (n = 5–6 per group). (For
interpretation of the references to colour in this figure legend, the reader is referred to the web version of this article.)

also support the clinical practice that gabapentinoids do not exhibit agonists glycine, gelsemine and koumine (Huang et al., 2016; Shoaib
gender deference in anti-neuropathic pain efficacies. et al., 2019; Li et al., 2016; Gong et al., 2014; Apryani et al., 2020; Mao
Given the existed correlation between IL and 10/β-endorphin et al., 2019; Fan et al., 2016). In our current study, intrathecal pre­
microglial stimulation and mechanical allodynia inhibition, the causal treatment with minocycline completely blocked gabapentinoids-
relationship of gabapentinoids was further explored. Minocycline is an inhibited mechanical allodynia. Additionally, intrathecal pretreatment
effective microglial activation inhibitor, although it may have minor with the IL-10 antibody totally attenuated each gabapentinoid
effects on astrocytes or neurons (Yoon et al., 2012; Jackson-Lewis et al., compound-inhibited mechanical allodynia, which are supported by that
2002; Jin et al., 2014). It has been accepted that minocycline may intrathecal IL-10 antibody blocked gabapentin antinociception in
prevent the initiation or early development of neuropathy pain, but does neuropathic rats and morphine tolerant rats (Lee et al., 2013; Bao et al.,
not produce antinociception in established neuropathy (Raghavendra 2014b, 2014a). Furthermore, prior-treatment intrathecally with
et al., 2003; Li et al., 2016; Gong et al., 2014; Mei et al., 2011; Fan et al., β-endorphin antiserum and MOR antagonist, but not KOR or DOR an­
2016). In addition, minocycline has been successfully proved to block tagonists, entirely alleviated spinal gabapentinoids-inhibited mechani­
microglia-mediated antinociceptive effects of GLP-1R agonist exenatide, cal allodynia, suggesting a subsequent role of endogenous opioid role in
GPR40 agonist GW9508, α7nAChR agonist cinobufagin, aconitum- gabapentinoids analgesia, in agreement with that the opioid-receptor
derived bullatine A, aconitine, lappaconitine and bulleyaconitine A but antagonist naloxone alleviated gabapentin-induced antinociception in
not neurons-mediated antinociceptive effects of MOR agonists morphine the rat orofacial formalin test (Miranda et al. 2015).
and β-endorphin, KOR agonist dynorphin A, and glycine receptor The importance of the microglial IL-10/β-endorphin pain

356
K.A. Ahmad et al. Brain Behavior and Immunity 95 (2021) 344–361

Fig. 12. Representative photomicrographs of the specific α2δ-1 expression in primarily cultured spinal neuronal cells (A, B), but not astrocytic cells (C, D) or
microglial cells (E, F). DAPI staining was used to identify cell nuclei. G-L. Effects of gabapentin treatment (100 µM) for 2 h on α2δ-1 expression in neurons, astrocytes
or microglial cells. The % immunolabeled surface areas of α2δ-1/NeuN (M), α2δ-1/GFAP (N), and α2δ-1/Iba-1 (O) were determined by the ImageJ software. Scale
bars: A, C, E, G, I, K, 50 µm; B, D, F, H, J, L, 10 µm. Data are shown as means ± SEM (n = 5–6 per group).

Fig. 13. Effects of the intrathecally GLP-1 receptor antagonist exendin-9–39, GPR40 antagonist GW1100, CRF receptor antagonist α-helical CRF (9–41) (A-C) and
adenosine A1 receptor antagonist DPCPX (D-F) on spinal gabapentinoids-inhibited mechanical allodynia in neuropathic rats. Exendin9-39, GW1100, α-helical CRF9-
41 or DPCPX was injected intrathecally 0.5 h before spinal gabapentinoids administration. The data are presented as means ± SEM (n = 6 per group).

357
K.A. Ahmad et al. Brain Behavior and Immunity 95 (2021) 344–361

Fig. 14. Schematic of the proposed mechanisms of gabapentinoids to inhibit pain hypersensitivity in neuropathic rats following activation of microglial spinal IL-10/
β-endorphin signaling pathway. Gabapentinoids stimulate IL-10, which subsequently expresses β-endorphin following activation of IL-10 receptors on microglial cells
by an autocrine signaling. β-Endorphin passes the microglial-neuronal synapse to activate μ-opioid receptor (MOR) in neurons and produce antinociception.

antihypersensitivity signal transduction has been selectively addressed. GPR40 and α7nAChRs, cynandione A, and aconitines, are specific in
The GLP-1R agonist exenatide and morroniside, GPR40 agonist pain hypersensitivity (not nociceptive pain) with efficacies of around
GW9508, α7nAChRs agonists PHA-543613, cinobufagin and lemairamin 60–70% MPE in mechanical allodynia (Gong et al., 2014; Wu et al.,
inhibited mechanical allodynia and thermal hyperalgesia in various pain 2017b, 2018a; Li et al., 2016; Huang et al., 2016, 2017; Apryani et al.,
models through spinal microglial expression of IL-10 and subsequent 2020; Sun et al., 2018).
β-endorphin expression (Gong et al. 2014; Wu et al. 2017; Mao et al., The target molecules for gabapentinoids to fast induce IL-10/
2019; Apryani et al., 2020; Wang et al., 2020; Tang et al., 2020). Low β-endorphin microglial expression have not been illustrated. It has been
frequency electroacupuncture stimulation produced pain anti­ reported gabapentinoids did not displace radioligands from widely-
hypersensitivity by spinal microglial IL-10/β-endorphin upregulation studied target sites, such as adrenergic, adenosine, dopamine, seroto­
(Ali et al. 2020), probably by activation of α7nAChR (Wang et al. 2018). nin, histamine, GABA, acetylcholine, glutamate and opioid receptors,
Early life-induced constitutive suppression of neuropathic pain is calcium and potassium channels, and dopamine, norepinephrine and
mediated by the IL-10/β-endorphin microglial signaling pathway serotonin transmitters up to 50 or 100 μM (Domon et al., 2018a, 2018b;
(McKelvey et al., 2015; Tang Xue-qi et al., 2019). Thus the discovery of Li et al., 2011). In addition, GLP-1Rs and GRP40 are expressed in
mechanism of gabapentinoids analgesia expands biological implications microglia and their agonists stimulated microglial IL-10/β-endorphin
of the IL-10/β-endorphin spinal microglial pain antihypersensitivity expression (Mao et al., 2019; Wu et al., 2017a, 2017b). Alternatively,
pathway. In addition, the pathway might be a promising human- CRF receptors have been associated with the microglial β-endorphin
validated target for discovery and development of analgesics in neuro­ expression (Iwaszkiewicz et al. 2013) and adenosine A1 receptors
pathic pain and other pain hypersensitivity states, as gabapentinoids are antagonism has been shown to prevent gabapentin behavioral effects
clinically effective analgesics. Furthermore, the effects of gabapenti­ (Martins et al. 2015). However, our results demonstrated that the an­
noids may characterize microglia-mediated analgesics. Our results tagonists of GLP-1Rs, GPR40, CRF receptors and adenosine A1 receptors
showed that gabapentinoids specifically attenuated ipsilateral mechan­ were unable to block each gabapentinoid-inhibited mechanical allody­
ical allodynia without influencing the contralateral paws thresholds nia, suggesting that their spinal IL-10/β-endorphin expression is not
significantly. The results are in concurrence with previous reports in mediated by these receptors. Moreover, specific colocalization of α2δ-1
which gabapentinoids were efficient in attenuating pain hypersensitiv­ was observed with the neuronal biomarker, but not with microglial or
ity in models of neuropathy, bone-cancer, inflammatory and post­ astrocytic biomarker in the spinal dorsal horns from neuropathy rats and
operative pain, but not nociceptive pain such as hot-plate, tail- primary cultures of spinal cells originated from neonatal rats. In addi­
immersion or paw-pressure test (Cheng and Chiou 2006). In addition, all tion, gabapentin treatment for 2 h did not affect α2δ-1 expression either
of gabapentinoids exhibit the same efficacy of around 60–70% MPE in in neurons or even in microglia and astrocytes. Our findings are in
attenuating mechanical allodynia, which is much lower than 100% MPE agreement with the previous studies that α2δ-1 was expressed only in
or greater (changing thresholds in normal nociceptive pain states) pro­ neurons but not microglia or astrocytes (Yang et al. 2012). Thus, our
duced by morphine or other opioid analgesics (Wang et al., 2017; Mao results reveal that gabapentinoids-stimulated microglial IL-10/
et al., 2020). All these results provide the basis for gabapentinoids β-endorphin expression was not through α2δ-1 binding or interaction.
clinical indications of postherpetic neuralgia, peripheral diabetic neu­ Consequently, additional researches are warranted to search for the
ropathy and fibromyalgia, as well as postoperative pain, headache, target molecules of gabapentinoids.
chronic low back pain and visceral pain, but not nociceptive pain, and To sum up, intrathecal gabapentinoids quickly inhibited mechanical
probably define their relatively limited clinical efficacy. Indeed, our allodynia and thermal hyperalgesia in neuropathic rats. They also pro­
extensive dose–response studies revealed that microglia-mediated anti­ moted IL-10 and β-endorphin expression in spinal dorsal horn microglia
nociceptive effects, produced by agonists of IL-10 receptors, GLP-1Rs, and primary microglia cultures, which were absent in α2δ-1 expression.

358
K.A. Ahmad et al. Brain Behavior and Immunity 95 (2021) 344–361

In addition, gabapentinoids-inhibited allodynia was entirely reversed by novel ligand for the α2δ subunit of voltage-gated calcium channels. J. Pharmacol.
Exp. Ther. 365, 573–582.
prior-treatment with microglial metabolic inhibitor minocycline, IL-10
Fan, H., Li, T.F., Gong, N., Wang, Y.X., 2016. Shanzhiside methylester, the principle
antibody, β-endorphin antiserum, and MOR antagonist CTAP. The re­ effective iridoid glycoside from the analgesic herb Lamiophlomis rotata, reduces
sults suggest that spinal gabapentinoids inhibit neuropathic pain neuropathic pain by stimulating spinal microglial β-endorphin expression.
through microglial expression of IL-10 and subsequent β-endorphin Neuropharmacology 101, 98–109.
Fan, H., Gong, N., Li, T.-F., Ma, A.-N., Xiao-Yan, W.u., Wang, M.-W., Wang, Y.-X., 2015.
expression. The proposed schematic microglial pathway of IL-10/ The non-peptide GLP-1 receptor agonist WB 4–24 blocks inflammatory nociception
β-endorphin for gabapentinoids to alleviate neuropathic pain is shown by stimulating β-endorphin release from spinal microglia. Br. J. Pharmacol. 172,
in Fig. 14. 64–79.
Franco, R., Fernandez-Suarez, D., 2015. Alternatively activated microglia and
macrophages in the central nervous system. Prog. Neurobiol. 131, 65–86.
Funding sources Gee, N.S., Brown, J.P., Dissanayake, V.UK., Offord, J., Thurlow, R., Woodruff, G.N.,
1996. The novel anticonvulsant drug, gabapentin (Neurontin), binds to the subunit
of a calcium channel. J. Biol. Chem. 271, 5768–5776.
This study was supported by the National Natural Science Founda­ Glocker, E.-O., Kotlarz, D., Klein, C., Shah, N., Grimbacher, B., 2011. IL-10 and IL-10
tion of China (No. 81673403). receptor defects in humans. Ann. N. Y. Acad. Sci. 1246, 102–107.
Göke, R., Fehmann, H.C., Linn, T., Schmidt, H., Krause, M., Eng, J., Göke, B., 1993.
Exendin-4 is a high potency agonist and truncated exendin-(9–39)-amide an
Author contributions antagonist at the glucagon-like peptide 1-(7–36)-amide receptor of insulin-secreting
beta-cells. J Biol Chem 268, 19650–19655.
Gong, N., Xiao, Q., Zhu, B., Zhang, C.Y., Wang, Y.C., Fan, H., Ma, A.N., Wang, Y.X., 2014.
Conceived and designed the experiments: KAA and YXW; performed
Activation of spinal glucagon-like peptide-1 receptors specifically suppresses pain
the experiments: KAA, RMS, MZA, MD, ML, EA and XYL; analyzed the hypersensitivity. J Neurosci 34, 5322–5334.
data: KAA and YXW and wrote the paper: KAA and YXW. González-Bueno, J., Calvo-Cidoncha, E., Desongles-Corrales, T., Santos Rubio, M.D.,
Chamorro, E., de Vega, and FJ Bautista-Paloma., 2015. DI-024 off-label use of
gabapentin and pregabalin in a tertiary hospital. British Medical Journal Publishing
Declaration of Competing Interest Group, In.
Gu, Nan, Eyo, Ukpong B, Murugan, Madhuvika, Peng, Jiyun, Matta, Sanjana,
Dong, Hailong, Long-Jun, Wu., 2016. Microglial P2Y12 receptors regulate microglial
The authors declare that they have no known competing financial activation and surveillance during neuropathic pain. Brain Behav. Immun. 55,
interests or personal relationships that could have appeared to influence 82–92.
Hendrich, Jan, Minh, Alexandra Tran Van, Heblich, Fay, Nieto-Rostro, Manuela,
the work reported in this paper.
Watschinger, Katrin, Striessnig, Jörg, Wratten, Jack, Davies, Anthony,
Dolphin, Annette C, 2008. Pharmacological disruption of calcium channel trafficking
References by the α2δ ligand gabapentin. Proc. Natl. Acad. Sci. 105, 3628–3633.
Hoppa, Michael B, Lana, Beatrice, Margas, Wojciech, Dolphin, Annette C, Ryan, Timothy
A, 2012. α2δ expression sets presynaptic calcium channel abundance and release
Ali, U., Apryani, E., Wu, H.Y., Mao, X.F., Liu, H., Wang, Y.X., 2020. Low frequency
probability. Nature 486, 122–125.
electroacupuncture alleviates neuropathic pain by activation of spinal microglial IL-
Hu, Ji-Hua, Yang, Jian-Ping, Liu, Lei, Li, Cai-Fang, Wang, Li-Na, Ji, Fu-Hai, Cheng, Hao,
10/beta-endorphin pathway. Biomed Pharmacother 125, 109898.
2012. Involvement of CX3CR1 in bone cancer pain through the activation of
Apryani, E., Ali, U., Wang, Z.Y., Wu, H.Y., Mao, X.F., Ahmad, K.A., Li, X.Y., Wang, Y.X.,
microglia p38 MAPK pathway in the spinal cord. Brain Res. 1465, 1–9.
2020. The spinal microglial IL-10/beta-endorphin pathway accounts for
Hua, Xiao-Ying, Svensson, Camilla I, Matsui, Tomohiro, Fitzsimmons, Bethany,
cinobufagin-induced mechanical antiallodynia in bone cancer pain following
Yaksh, Tony L, Webb, Michael, 2005. Intrathecal minocycline attenuates peripheral
activation of alpha7-nicotinic acetylcholine receptors. J Neuroinflammation 17, 75.
inflammation-induced hyperalgesia by inhibiting p38 MAPK in spinal microglia. Eur.
Bannister, K., Sikandar, S., Bauer, C.S., Dolphin, A.C., Porreca, F., Dickenson, A.H., 2011.
J. Neurosci. 22, 2431–2440.
Pregabalin suppresses spinal neuronal hyperexcitability and visceral hypersensitivity
Huang, J.L., Chen, X.L., Guo, C., Wang, Y.X., 2012. Contributions of spinal D-amino acid
in the absence of peripheral pathophysiology. Anesthesiology: The Journal of the
oxidase to bone cancer pain. Amino Acids 43, 1905–1918.
American Society of Anesthesiologists 115, 144–152.
Huang, Q., Mao, X.F., Wu, H.Y., Li, T.F., Sun, M.L., Liu, H., Wang, Y.X., 2016. Bullatine A
Bao, Y.-H., Zhou, Q.-H., Chen, R., Hao, X.u., Zeng, L.-L., Zhang, X., Jiang, W., Dong-
stimulates spinal microglial dynorphin A expression to produce anti-hypersensitivity
Ping, D.u., 2014a. Gabapentin enhances the morphine anti-nociceptive effect in
in a variety of rat pain models. J Neuroinflammation 13, 214.
neuropathic pain via the interleukin-10-heme oxygenase-1 signalling pathway in
Huang, Q., Mao, X.F., Wu, H.Y., Liu, H., Sun, M.L., Wang, X., Wang, Y.X., 2017.
rats. J. Mol. Neurosci. 54, 137–146.
Cynandione A attenuates neuropathic pain through p38beta MAPK-mediated spinal
Bao, Y.-H., Zhou, Q.-H., Chen, R., Hao, X.u., Zeng, L., Zhang, X., Jiang, W., DongPing, D.
microglial expression of beta-endorphin. Brain Behav Immun 62, 64–77.
u., 2014b. Gabapentin attenuates morphine tolerance through interleukin-10.
Huang, S.N., Wei, J., Huang, L.T., Ju, P.J., Chen, J., Wang, Y.X., 2020. Bulleyaconitine A
NeuroReport 25, 71–76.
Inhibits Visceral Nociception and Spinal Synaptic Plasticity through Stimulation of
Batti, L., Sundukova, M., Murana, E., Pimpinella, S., De Castro, F., Reis, F.P., Wang, H.,
Microglial Release of Dynorphin A. Neural Plast 2020, 1484087.
Pellegrino, E., Perlas, E., Di Angelantonio, S., 2016. TMEM16F regulates spinal
Iwaszkiewicz, Katerina, Schneider, Jennifer, Hua, Susan, 2013. Targeting peripheral
microglial function in neuropathic pain states. Cell reports 15, 2608–2615.
opioid receptors to promote analgesic and anti-inflammatory actions. Front.
Bauer, C.S., Rahman, W., Tran-van-Minh, A., Lujan, R., Dickenson, A.H., Dolphin, A.C.,
Pharmacol. 4, 132.
2010. The anti-allodynic alpha(2)delta ligand pregabalin inhibits the trafficking of
Jackson-Lewis, Vernice, Vila, Miquel, Tieu, Kim, Teismann, Peter, Vadseth, Caryn,
the calcium channel alpha(2)delta-1 subunit to presynaptic terminals in vivo.
Choi, Dong-Kug, Ischiropoulos, Harry, Przedborski, Serge, 2002. Blockade of
Biochem Soc Trans 38, 525–528.
microglial activation is neuroprotective in the 1-methyl-4-phenyl-1, 2, 3, 6-tetra­
Bauer, C.S., Nieto-Rostro, M., Rahman, W., Tran-Van-Minh, A., Ferron, L., Douglas, L.,
hydropyridine mouse model of Parkinson disease. J. Neurosci. 22, 1763–1771.
Kadurin, I., Ranjan, Y.S., Fernandez-Alacid, L., Millar, N.S., 2009. The increased
Ji, R.R., Berta, T., Nedergaard, M., 2013. Glia and pain: is chronic pain a gliopathy? Pain
trafficking of the calcium channel subunit α2δ-1 to presynaptic terminals in
154 (Suppl 1), S10–S28.
neuropathic pain is inhibited by the α2δ ligand pregabalin. J. Neurosci. 29,
Jin, Wen-Jie, Feng, Shan-Wu, Feng, Zhou, Shun-Mei, Lu., Qi, Tao, Qian, Yan-Ning, 2014.
4076–4088.
Minocycline improves postoperative cognitive impairment in aged mice by
Bockbrader, H.N., Wesche, D., Miller, R., Chapel, S., Janiczek, N., Burger, P., 2010.
inhibiting astrocytic activation. NeuroReport 25, 1–6.
A comparison of the pharmacokinetics and pharmacodynamics of pregabalin and
Kawasaki, Yasuhiko, Zhang, Ling, Cheng, Jen-Kun, Ji, Ru-Rong, 2008. Cytokine
gabapentin. Clin. Pharmacokinet. 49, 661–669.
mechanisms of central sensitization: distinct and overlapping role of interleukin-1β,
Carniglia, Lila, Delia Ramírez, Daniela Durand, Julieta Saba, Juan Turati, Carla Caruso,
interleukin-6, and tumor necrosis factor-α in regulating synaptic and neuronal
Teresa N Scimonelli, and Mercedes Lasaga. 2017. ’Neuropeptides and microglial
activity in the superficial spinal cord. J. Neurosci. 28, 5189–5194.
activation in inflammation, pain, and neurodegenerative diseases’, Mediators of
Kim, S.H., Chung, J.M., 1992. An experimental model for peripheral neuropathy
Inflammation, 2017.
produced by segmental spinal nerve ligation in the rat. Pain 50, 355–363.
Chen, J., Li, L., Chen, S.-R., Chen, H., Xie, J.-D., Sirrieh, R.E., MacLean, D.M., Zhang, Y.,
Kim, Woong Mo, Jeong, Cheol Won, Lee, Seong Heon, Kim, Yeo Ok, Cui, Jin Hua,
Zhou, M.-H., Jayaraman, V., 2018. The α2δ-1-NMDA receptor complex is critically
Yoon, Myung Ha, 2011. The intrathecally administered Kappa-2 opioid agonist
involved in neuropathic pain development and gabapentin therapeutic actions. Cell
GR89696 and interleukin-10 attenuate bone cancer–induced pain through
reports 22, 2307–2321.
synergistic interaction. Anesth. Analg. 113, 934–940.
Cheng, J.K., Chiou, L.C., 2006. Mechanisms of the antinociceptive action of gabapentin.
Lan, Liu Si, Yang Jian Ping, Wang Li Na, Jiang Miao, Qiu Qiao Cheng, Ma Zhen Ni, Liu
J Pharmacol Sci 100, 471–486.
Lei, Li Cai Fang, Ren Chun Guang, and Zhou Jin. 2010. ’Down-regulation of Toll-like
Deeks, E.D., 2019. Mirogabalin: first global approval. Drugs 79, 463–468.
receptor 4 gene expression by short interfering RNA attenuates bone cancer pain in a
Domon, Y., Kitano, Y., Makino, M., 2018a. Analgesic effects of the novel α2δ ligand
rat model’, Molecular pain, 6: 1744-8069-6-2.
mirogabalin in a rat model of spinal cord injury. Die Pharmazie-An International
Ledeboer, Annemarie, Jekich, Brian M, Sloane, Evan M, Mahoney, John H,
Journal of Pharmaceutical Sciences 73, 659–661.
Langer, Stephen J, Milligan, Erin D, Martin, David, Maier, Steven F, Johnson, Kirk
Domon, Y., Arakawa, N., Inoue, T., Matsuda, F., Takahashi, M., Yamamura, N., Kai, K.,
W, Leinwand, Leslie A, 2007. Intrathecal interleukin-10 gene therapy attenuates
Kitano, Y., 2018b. Binding characteristics and analgesic effects of mirogabalin, a

359
K.A. Ahmad et al. Brain Behavior and Immunity 95 (2021) 344–361

paclitaxel-induced mechanical allodynia and proinflammatory cytokine expression spinal glycine receptor activation-induced allopregnanolone biosynthesis. Biochem
in dorsal root ganglia in rats. Brain Behav. Immun. 21, 686–698. Pharmacol 161, 136–148.
Lee, Byung-Sang, Jun, In-Gu, Kim, Sung-Hoon, Park, Jong Yeon, 2013. Intrathecal Sorge, Robert E, Mapplebeck, Josiane CS, Rosen, Sarah, Beggs, Simon, Taves, Sarah,
gabapentin increases interleukin-10 expression and inhibits pro-inflammatory Alexander, Jessica K, Martin, Loren J, Austin, Jean-Sebastien, Sotocinal, Susana G,
cytokine in a rat model of neuropathic pain. J. Korean Med. Sci. 28, 308–314. Chen, Di, 2015. Different immune cells mediate mechanical pain hypersensitivity in
Li, Chun-Ying, Zhang, Xiu-Lin, Matthews, Elizabeth A, Li, Kang-Wu, Kurwa, Ambereen, male and female mice. Nat. Neurosci. 18, 1081–1083.
Boroujerdi, Amin, Gross, Jimmy, Gold, Michael S, Dickenson, Anthony H, Stahl, Stephen M, Porreca, Frank, Taylor, Charles P, Cheung, Raymond, Thorpe, Andrew
Feng, Guoping, 2006. Calcium channel α2δ1 subunit mediates spinal J, Clair, Andrew, 2013. The diverse therapeutic actions of pregabalin: is a single
hyperexcitability in pain modulation. Pain 125, 20–34. mechanism responsible for several pharmacological activities? Trends Pharmacol.
Li, T.F., Fan, H., Wang, Y.X., 2016. Aconitum-Derived Bulleyaconitine A Exhibits Sci. 34, 332–339.
Antihypersensitivity Through Direct Stimulating Dynorphin A Expression in Spinal Staniland, Amelia A, Clark, Anna K, Wodarski, Rachel, Sasso, Oscar, Maione, Francesco,
Microglia. J Pain 17, 530–548. D’Acquisto, Fulvio, Malcangio, Marzia, 2010. Reduced inflammatory and
Li, T.-F., Wu, H.-Y., Wang, Y.-R., Li, X.-Y., Wang, Y.X., 2017. Molecular signaling neuropathic pain and decreased spinal microglial response in fractalkine receptor
underlying bulleyaconitine A (BAA)-induced microglial expression of prodynorphin. (CX3CR1) knockout mice. J. Neurochem. 114, 1143–1157.
Sci. Rep. 7, 45056. Steinmiller, C.L., Young, A.M., 2008. Pharmacological selectivity of CTAP in a warm
Li, Z., Taylor, C.P., Weber, M., Piechan, J., Prior, F., Bian, F., Cui, M., Hoffman, D., water tail-withdrawal antinociception assay in rats. Psychopharmacology 195,
Donevan, S., 2011. Pregabalin is a potent and selective ligand for α(2)δ-1 and α(2)δ- 497–507.
2 calcium channel subunits. Eur J Pharmacol 667, 80–90. Suman-Chauhan, N., Webdale, L., Hill, D.R., Woodruff, G.N., 1993. Characterisation of
Lu, J.M., Gong, N., Wang, Y.C., Wang, Y.X., 2012. D-Amino acid oxidase-mediated [3H]gabapentin binding to a novel site in rat brain: homogenate binding studies. Eur
increase in spinal hydrogen peroxide is mainly responsible for formalin-induced J Pharmacol 244, 293–301.
tonic pain. Br J Pharmacol 165, 1941–1955. Sun, M.L., Ao, J.P., Wang, Y.R., Huang, Q., Li, T.F., Li, X.Y., Wang, Y.X., 2018.
Mao, X.F., Wu, H.Y., Tang, X.Q., Ali, U., Liu, H., Wang, Y.X., 2019. Activation of GPR40 Lappaconitine, a C18-diterpenoid alkaloid, exhibits antihypersensitivity in chronic
produces mechanical antiallodynia via the spinal glial interleukin-10/β-endorphin pain through stimulation of spinal dynorphin A expression. Psychopharmacology
pathway. J Neuroinflammation 16, 84. 235, 2559–2571.
Mao, Xiao-Fang, Muhammad Zaeem Ahsan, Evhy Apryani, Xue-Qi Tang, Meng-Jing Tae, Han-Shen, Smith, Kelly M, Marie Phillips, A., Boyle, Kieran A, Li, Melody,
Zhao, Xin-Yan Li, and Yong-Xiang Wang. 2020. ’Dual μ-opioid receptor and Forster, Ian C, Hatch, Robert J, Richardson, Robert, Hughes, David I, Graham, Brett
norepinephrine reuptake mechanisms contribute to dezocine-and tapentadol- A, 2017. Gabapentin modulates HCN4 channel voltage-dependence. Front.
induced mechanical antiallodynia in cancer pain’, European Journal of Pharmacol. 8, 554.
Pharmacology: 173062. Tang, X., Wu, H., Mao, X., Li, X., Wang, Y., 2020. The GLP-1 receptor herbal agonist
Martins, Daniel F, Prado, Marcos RB, Daruge-Neto, Eduardo, Batisti, Ana P, Emer, Aline morroniside attenuates neuropathic pain via spinal microglial expression of IL-10
A, Mazzardo-Martins, Leidiane, Santos, Adair RS, Piovezan, Anna P, 2015. Caffeine and beta-endorphin. Biochem Biophys Res Commun 530, 494–499.
prevents antihyperalgesic effect of gabapentin in an animal model of CRPS-I: Thakur, Vikram, Gonzalez, Mayra, Pennington, Kristen, Chattopadhyay, Munmun, 2016.
evidence for the involvement of spinal adenosine A1 receptor. Journal of the Viral vector mediated continuous expression of interleukin-10 in DRG alleviates pain
Peripheral Nervous System 20, 403–409. in type 1 diabetic animals. Mol. Cell. Neurosci. 72, 46–53.
McKelvey, Rebecca, Berta, Temugin, Old, Elizabeth, Ji, Ru-Rong, Fitzgerald, Maria, Tiwari, Vinod, Guan, Yun, Raja, Srinivasa N, 2014. Modulating the delicate
2015. Neuropathic pain is constitutively suppressed in early life by anti- glial–neuronal interactions in neuropathic pain: Promises and potential caveats.
inflammatory neuroimmune regulation. J. Neurosci. 35, 457–466. Neurosci. Biobehav. Rev. 45, 19–27.
Mei, X.P., Xu, H., Xie, C., Ren, J., Zhou, Y., Zhang, H., Xu, L.X., 2011. Post-injury Wang, Y., Jiang, Q., Xia, Y.Y., Huang, Z.H., Huang, C., 2018. Involvement of α7nAChR in
administration of minocycline: an effective treatment for nerve-injury induced electroacupuncture relieving neuropathic pain in the spinal cord of rat with spared
neuropathic pain. Neurosci Res 70, 305–312. nerve injury. Brain Res Bull 137, 257–264.
Milligan, Erin D, Evan M Sloane, Stephen J Langer, Pedro E Cruz, Marucia Chacur, Leah Wang, Y.X., Mao, X.F., Li, T.F., Gong, N., Zhang, M.Z., 2017. Dezocine exhibits
Spataro, Julie Wieseler-Frank, Sayamwong E Hammack, Steven F Maier, and Terence antihypersensitivity activities in neuropathy through spinal μ-opioid receptor
R Flotte. 2005. ’Controlling neuropathic pain by adeno-associated virus driven activation and norepinephrine reuptake inhibition. Sci Rep 7, 43137.
production of the anti-inflammatory cytokine, interleukin-10’, Molecular pain, 1: Wang, Y.X., Pang, C.C., 1993. Functional integrity of the central and sympathetic
1744-8069-1-9. nervous systems is a prerequisite for pressor and tachycardic effects of
Milligan, Erin D, Watkins, Linda R, 2009. Pathological and protective roles of glia in diphenyleneiodonium, a novel inhibitor of nitric oxide synthase. J Pharmacol Exp
chronic pain. Nat. Rev. Neurosci. 10, 23–36. Ther 265, 263–272.
Miranda, Hugo F, Sierralta, Fernando, Lux, Sebastian, Troncoso, Rocío, Zepeda, Ramiro, Wang, Zi-Ying, Qiao-Qiao Han, Meng-Yan Deng, Meng-Jing Zhao, Evhy Apryani, Rana
Zanetta, Pilar, Noriega, Viviana, Prieto, Juan Carlos, 2015. Involvement of Muhammad Shoaib, Dong-Qing Wei, and Yong-Xiang Wang. 2020. ’Lemairamin,
nitridergic and opioidergic pathways in the antinociception of gabapentin in the isolated from the Zanthoxylum plants, alleviates pain hypersensitivity via spinal α7
orofacial formalin test in mice. Pharmacol. Rep. 67, 399–403. nicotinic acetylcholine receptors’, Biochemical and Biophysical Research
Nakamoto, K., Nishinaka, T., Matsumoto, K., Kasuya, F., Mankura, M., Koyama, Y., Communications.
Tokuyama, S., 2012. Involvement of the long-chain fatty acid receptor GPR40 as a Wodarski, R., Clark, A.K., Grist, J., Marchand, F., Malcangio, M., 2009. Gabapentin
novel pain regulatory system. Brain Res 1432, 74–83. reverses microglial activation in the spinal cord of streptozotocin-induced diabetic
Nakata, M., Yamamoto, S., Okada, T., Gantulga, D., Okano, H., Ozawa, K., Yada, T., rats. Eur J Pain 13, 807–811.
2016. IL-10 gene transfer upregulates arcuate POMC and ameliorates hyperphagia, Wu, H.Y., Mao, X.F., Tang, X.Q., Ali, U., Apryani, E., Liu, H., Li, X.Y., Wang, Y.X., 2018a.
obesity and diabetes by substituting for leptin. Int J Obes (Lond) 40, 425–433. Spinal interleukin-10 produces antinociception in neuropathy through microglial
Nieto, Francisco R, Clark, Anna K, Grist, John, Hathway, Gareth J, Chapman, Victoria, beta-endorphin expression, separated from antineuroinflammation. Brain Behav
Malcangio, Marzia, 2016. Neuron-immune mechanisms contribute to pain in early Immun 73, 504–519.
stages of arthritis. Journal of neuroinflammation 13, 1–13. Wu, H.Y., Tang, X.Q., Liu, H., Mao, X.F., Wang, Y.X., 2018b. Both classic Gs-cAMP/PKA/
Park, J.Y., Jun, I.G., Jun. 2008. The interaction of gabapentin and N6-(2- CREB and alternative Gs-cAMP/PKA/p38beta/CREB signal pathways mediate
phenylisopropyl)-adenosine R-(-)isomer (R-PIA) on mechanical allodynia in rats exenatide-stimulated expression of M2 microglial markers. J Neuroimmunol 316,
with a spinal nerve ligation. J Korean Med Sci 23, 678–684. 17–22.
Perry, V Hugh, Holmes, Clive, 2014. Microglial priming in neurodegenerative disease. Wu, H.-Y., Mao, X.-F., Fan, H., Wang, Y.-X., 2017a. p38β MAPK signaling mediates
Nature Reviews Neurology 10, 217–224. exenatide-stimulated microglial β-endorphin expression. Mol. Pharmacol 91,
Raghavendra, V., Tanga, F., DeLeo, J.A., 2003. Inhibition of microglial activation 451–463.
attenuates the development but not existing hypersensitivity in a rat model of Wu, H.Y., Tang, X.Q., Mao, X.F., Wang, Y.X., 2017b. Autocrine Interleukin-10 Mediates
neuropathy. J Pharmacol Exp Ther 306, 624–630. Glucagon-Like Peptide-1 Receptor-Induced Spinal Microglial beta-Endorphin
Rosa, A.S., Freitas, M.F., Rocha, I.R., Chacur, M., 2017. Gabapentin decreases microglial Expression. J Neurosci 37, 11701–11714.
cells and reverses bilateral hyperalgesia and allodynia in rats with chronic myositis. Tang Xue-qi, Lu., Jin-miao, Mao Xiao-fang, Yong-xiang, Wang, 2019. Spinal Interleukin-
Eur J Pharmacol 799, 111–117. 10/β-Endorphin Pathway Suppresses Neuropathic Pain in Infant Rats. Journal of Sun
Sabat, Robert, Grütz, Gerald, Warszawska, Katarzyna, Kirsch, Stefan, Witte, Ellen, Yat-sen University (Medical Sciences) 40 (3), 351–357.
Wolk, Kerstin, Geginat, Jens, 2010. Biology of interleukin-10. Cytokine Growth Yang, Fang, Whang, John, Derry, William T, Vardeh, Daniel, Scholz, Joachim, 2014.
Factor Rev. 21, 331–344. Analgesic treatment with pregabalin does not prevent persistent pain after
Sawamura, S., Obara, M., Takeda, K., Maze, M., Hanaoka, K., 2003. Corticotropin- peripheral nerve injury in the rat. PAIN® 155, 356–366.
releasing factor mediates the antinociceptive action of nitrous oxide in rats. Yang, Jia-Le, Bo, Xu., Li, Shuang-Shuang, Zhang, Wei-Shi, Hua, Xu., Deng, Xiao-Ming,
Anesthesiology 99, 708–715. Zhang, Yu-Qiu, 2012. Gabapentin reduces CX3CL1 signaling and blocks spinal
Scholz, Joachim, Woolf, Clifford J, 2007. The neuropathic pain triad: neurons, immune microglial activation in monoarthritic rats. Molecular brain 5, 18.
cells and glia. Nat. Neurosci. 10, 1361–1368. Yang, Rui-Hua, Wang, Wen-Ting, Chen, Jing-Yuan, Xie, Rou-Gang, San-Jue, Hu., 2009.
Shao, S., Xia, H., Hu, M., Chen, C., Fu, J., Shi, G., Guo, Q., Zhou, Y., Wang, W., Shi, J., Gabapentin selectively reduces persistent sodium current in injured type-A dorsal
Zhang, T., 2020. Isotalatizidine, a C(19)-diterpenoid alkaloid, attenuates chronic root ganglion neurons. Pain 143, 48–55.
neuropathic pain through stimulating ERK/CREB signaling pathway-mediated Yang, Yan, Li, Hui, Li, Ting-Ting, Luo, Hao, Xi-Yao, Gu., Lü, Ning, Ji, Ru-Rong,
microglial dynorphin A expression. J Neuroinflammation 17, 13. Zhang, Yu-Qiu, 2015. Delayed activation of spinal microglia contributes to the
Shoaib, R.M., Zhang, J.Y., Mao, X.F., Wang, Y.X., 2019. Gelsemine and koumine, maintenance of bone cancer pain in female Wistar rats via P2X7 receptor and IL-18.
principal active ingredients of Gelsemium, exhibit mechanical antiallodynia via J. Neurosci. 35, 7950–7963.

360
K.A. Ahmad et al. Brain Behavior and Immunity 95 (2021) 344–361

Yoon, S.-Y., Patel, Devang, Dougherty, Patrick M, 2012. Minocycline blocks Zhou, Z., Peng, X., Shuanglin Hao, D.J., Fink, and M Mata., 2008. HSV-mediated transfer
lipopolysaccharide induced hyperalgesia by suppression of microglia but not of interleukin-10 reduces inflammatory pain through modulation of membrane
astrocytes. Neuroscience 221, 214–224. tumor necrosis factor α in spinal cord microglia. Gene Ther. 15, 183–190.
Zhou, Chunyi, and Z David Luo. 2013. ’Sensory Pathway Modulation by Calcium
Channel α 2 δ 1 Subunit.’ in, Modulation of Presynaptic Calcium Channels
(Springer).

361

You might also like