Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
Neurosci Bull ; 40(3): 293-309, 2024 Mar.
Article in English | MEDLINE | ID: mdl-37639183

ABSTRACT

The cytochrome P450 proteins (CYP450s) have been implicated in catalyzing numerous important biological reactions and contribute to a variety of diseases. CYP26A1, a member of the CYP450 family, carries out the oxidative metabolism of retinoic acid (RA), the active metabolite of vitamin A. Here we report that CYP26A1 was dramatically upregulated in the spinal cord after spinal nerve ligation (SNL). CYP26A1 was mainly expressed in spinal neurons and astrocytes. HPLC analysis displayed that the content of all-trans-RA (at-RA), the substrate of CYP26A1, was reduced in the spinal cord on day 7 after SNL. Inhibition of CYP26A1 by siRNA or inhibition of CYP26A1-mediated at-RA catabolism by talarozole relieved the SNL-induced mechanical allodynia during the maintenance phase of neuropathic pain. Talarozole also reduced SNL-induced glial activation and proinflammatory cytokine production but increased anti-inflammatory cytokine (IL-10) production. The RA receptors RARα, RXRß, and RXRγ were expressed in spinal neurons and glial cells. The promoter of Il-10 has several binding sites for RA receptors, and at-RA directly increased Il-10 mRNA expression in vitro. Finally, intrathecal IL-10 attenuated SNL-induced neuropathic pain and reduced the activation of astrocytes and microglia. Collectively, the inhibition of CYP26A1-mediated at-RA catabolism alleviates SNL-induced neuropathic pain by promoting the expression of IL-10 and suppressing glial activation. CYP26A1 may be a potential therapeutic target for the treatment of neuropathic pain.


Subject(s)
Interleukin-10 , Neuralgia , Humans , Interleukin-10/metabolism , Retinoic Acid 4-Hydroxylase/metabolism , Spinal Cord/metabolism , Neuralgia/metabolism , Cytokines/metabolism , Hyperalgesia/metabolism
2.
EMBO Rep ; 24(10): e56098, 2023 10 09.
Article in English | MEDLINE | ID: mdl-37522391

ABSTRACT

A11 dopaminergic neurons regulate somatosensory transduction by projecting from the diencephalon to the spinal cord, but the function of this descending projection in itch remained elusive. Here, we report that dopaminergic projection neurons from the A11 nucleus to the spinal dorsal horn (dopaminergicA11-SDH ) are activated by pruritogens. Inhibition of these neurons alleviates itch-induced scratching behaviors. Furthermore, chemogenetic inhibition of spinal dopamine receptor D1-expressing (DRD1+ ) neurons decreases acute or chronic itch-induced scratching. Mechanistically, spinal DRD1+ neurons are excitatory and mostly co-localize with gastrin-releasing peptide (GRP), an endogenous neuropeptide for itch. In addition, DRD1+ neurons form synapses with GRP receptor-expressing (GRPR+ ) neurons and activate these neurons via AMPA receptor (AMPAR). Finally, spontaneous itch and enhanced acute itch induced by activating spinal DRD1+ neurons are relieved by antagonists against AMPAR and GRPR. Thus, the descending dopaminergic pathway facilitates spinal itch transmission via activating DRD1+ neurons and releasing glutamate and GRP, which directly augments GRPR signaling. Interruption of this descending pathway may be used to treat chronic itch.


Subject(s)
Receptors, Bombesin , Spinal Cord , Humans , Receptors, Bombesin/genetics , Receptors, Bombesin/metabolism , Gastrin-Releasing Peptide/genetics , Gastrin-Releasing Peptide/metabolism , Spinal Cord/metabolism , Glutamic Acid/metabolism , Dopamine/metabolism , Pruritus/genetics , Pruritus/metabolism , Dopaminergic Neurons/metabolism , Receptors, AMPA/genetics , Receptors, AMPA/metabolism
3.
Int J Mol Sci ; 24(12)2023 Jun 15.
Article in English | MEDLINE | ID: mdl-37373296

ABSTRACT

Phosphorylation of the serine 139 of the histone variant H2AX (γH2AX) is a DNA damage marker that regulates DNA damage response and various diseases. However, whether γH2AX is involved in neuropathic pain is still unclear. We found the expression of γH2AX and H2AX decreased in mice dorsal root ganglion (DRG) after spared nerve injury (SNI). Ataxia telangiectasia mutated (ATM), which promotes γH2AX, was also down-regulated in DRG after peripheral nerve injury. ATM inhibitor KU55933 decreased the level of γH2AX in ND7/23 cells. The intrathecal injection of KU55933 down-regulated DRG γH2AX expression and significantly induced mechanical allodynia and thermal hyperalgesia in a dose-dependent manner. The inhibition of ATM by siRNA could also decrease the pain threshold. The inhibition of dephosphorylation of γH2AX by protein phosphatase 2A (PP2A) siRNA partially suppressed the down-regulation of γH2AX after SNI and relieved pain behavior. Further exploration of the mechanism revealed that inhibiting ATM by KU55933 up-regulated extracellular-signal regulated kinase (ERK) phosphorylation and down-regulated potassium ion channel genes, such as potassium voltage-gated channel subfamily Q member 2 (Kcnq2) and potassium voltage-gated channel subfamily D member 2 (Kcnd2) in vivo, and KU559333 enhanced sensory neuron excitability in vitro. These preliminary findings imply that the down-regulation of γH2AX may contribute to neuropathic pain.


Subject(s)
Neuralgia , Peripheral Nerve Injuries , Animals , Mice , Ganglia, Spinal/metabolism , Hyperalgesia/genetics , Hyperalgesia/metabolism , Neuralgia/etiology , Neuralgia/metabolism , Peripheral Nerve Injuries/metabolism , Potassium/metabolism , RNA, Small Interfering/metabolism , Sensory Receptor Cells/metabolism , Shal Potassium Channels/metabolism
4.
Brain Res Bull ; 187: 122-137, 2022 09.
Article in English | MEDLINE | ID: mdl-35781031

ABSTRACT

Trigeminal neuropathic pain (TNP) arises due to peripheral nerve injury, the mechanisms underlying which are little known. The altered gene expression profile in sensory ganglia is critical for neuropathic pain generation and maintenance. We, therefore, assessed the transcriptome of the trigeminal ganglion (TG) from mice at different periods of pain progression. Trigeminal neuropathic pain was established by partial infraorbital nerve transection (pIONT). High-throughput RNA sequencing was applied to detect the mRNA profiles of TG collected at 3 and 10 days after modeling. Injured TG displayed dramatically altered mRNA expression profiles compared to Sham. Different gene expression profiles were obtained at 3 and 10 days after pIONT. Moreover, 314 genes were significantly upregulated, and 81 were significantly downregulated at both 3 and 10 days post-pIONT. Meanwhile, enrichment analysis of these persistent differentially expressed genes (DEGs) showed that the MAPK pathway was the most significantly enriched pathway for upregulated DEGs, validated by immunostaining. In addition, TG cell populations defined by single-nuclei RNA sequencing displayed cellular localization of DEGs at a single-cell resolution. Protein-protein interaction (PPI) and sub-PPI network analyses constructed networks and identified the top 10 hub genes for DEGs at different time points. The present data provide novel information on the gene expression signatures of TG during the development and maintenance phases of TNP, and the identified hub genes and pathways may serve as potential targets for treatment.


Subject(s)
Neuralgia , Trigeminal Neuralgia , Animals , High-Throughput Nucleotide Sequencing , Mice , Neuralgia/genetics , Neuralgia/metabolism , RNA, Messenger/metabolism , Transcriptome , Trigeminal Ganglion/metabolism , Trigeminal Neuralgia/genetics , Trigeminal Neuralgia/metabolism
5.
Adv Sci (Weinh) ; 9(27): e2201300, 2022 09.
Article in English | MEDLINE | ID: mdl-35892263

ABSTRACT

Peripheral nerve injury-induced spinal microglial proliferation plays a pivotal role in neuropathic pain. So far, key intracellular druggable molecules involved in this process are not identified. The nuclear factor of activated T-cells (NFAT1) is a master regulator of immune cell proliferation. Whether and how NFAT1 modulates spinal microglial proliferation during neuropathic pain remain unknown. Here it is reported that NFAT1 is persistently upregulated in microglia after spinal nerve ligation (SNL), which is regulated by TET2-mediated DNA demethylation. Global or microglia-specific deletion of Nfat1 attenuates SNL-induced pain and decreases excitatory synaptic transmission of lamina II neurons. Furthermore, deletion of Nfat1 decreases microglial proliferation and the expression of multiple microglia-related genes, such as cytokines, transmembrane signaling receptors, and transcription factors. Particularly, SNL increases the binding of NFAT1 with the promoter of Itgam, Tnf, Il-1b, and c-Myc in the spinal cord. Microglia-specific overexpression of c-MYC induces pain hypersensitivity and microglial proliferation. Finally, inhibiting NFAT1 and c-MYC by intrathecal injection of inhibitor or siRNA alleviates SNL-induced neuropathic pain. Collectively, NFAT1 is a hub transcription factor that regulates microglial proliferation via c-MYC and guides the expression of the activated microglia genome. Thus, NFAT1 may be an effective target for treating neuropathic pain.


Subject(s)
Microglia , Neuralgia , Cell Proliferation , Humans , Microglia/metabolism , Neuralgia/genetics , Neuralgia/metabolism , RNA, Small Interfering/metabolism , RNA, Small Interfering/pharmacology , Transcription Factors/metabolism , Transcription Factors/pharmacology
6.
Cell Mol Neurobiol ; 42(5): 1543-1555, 2022 Jul.
Article in English | MEDLINE | ID: mdl-33694132

ABSTRACT

Tumor necrosis factor receptor-associated factor 6 (TRAF6) has been reported to be expressed in spinal astrocytes and is involved in neuropathic pain. In this study, we investigated the role and mechanism of TRAF6 in complete Freund's adjuvant (CFA)-evoked chronic inflammatory hypersensitivity and the effect of docosahexaenoic acid (DHA) on TRAF6 expression and inflammatory pain. We found that TRAF6 was dominantly increased in microglia at the spinal level after intraplantar injection of CFA. Intrathecal TRAF6 siRNA alleviated CFA-triggered allodynia and reversed the upregulation of IBA-1 (microglia marker). In addition, intrathecal administration of DHA inhibited CFA-induced upregulation of TRAF6 and IBA-1 in the spinal cord and attenuated CFA-evoked mechanical allodynia. Furthermore, DHA prevented lipopolysaccharide (LPS)-caused increase of TRAF6 and IBA-1 in both BV2 cell line and primary cultured microglia. Finally, intrathecal DHA reduced LPS-induced upregulation of spinal TRAF6 and IBA-1, and alleviated LPS-induced mechanical allodynia. Our findings indicate that TRAF6 contributes to pain hypersensitivity via regulating microglial activation in the spinal dorsal horn. Direct inhibition of TRAF6 by siRNA or indirect inhibition by DHA may have therapeutic effects on chronic inflammatory pain.


Subject(s)
Chronic Pain , Neuralgia , Animals , Chronic Pain/metabolism , Freund's Adjuvant/metabolism , Freund's Adjuvant/toxicity , Hyperalgesia/metabolism , Inflammation/pathology , Lipopolysaccharides/pharmacology , Mice , Microglia/metabolism , Neuralgia/metabolism , RNA, Small Interfering/metabolism , Spinal Cord/metabolism , Spinal Cord Dorsal Horn/metabolism , TNF Receptor-Associated Factor 6/metabolism , TNF Receptor-Associated Factor 6/pharmacology
8.
Neurosci Bull ; 37(4): 550-562, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33355900

ABSTRACT

Trigeminal neuropathic pain (TNP) is a significant health problem but the involved mechanism has not been completely elucidated. Toll-like receptors (TLRs) have recently been demonstrated to be expressed in the dorsal root ganglion and involved in chronic pain. Here, we show that TLR8 was persistently increased in the trigeminal ganglion (TG) neurons in model of TNP induced by partial infraorbital nerve ligation (pIONL). In addition, deletion or knockdown of Tlr8 in the TG attenuated pIONL-induced mechanical allodynia, reduced the activation of ERK and p38-MAPK, and decreased the expression of pro-inflammatory cytokines in the TG. Furthermore, intra-TG injection of the TLR8 agonist VTX-2337 induced pain hypersensitivity. VTX-2337 also increased the intracellular Ca2+ concentration, induced the activation of ERK and p38, and increased the expression of pro-inflammatory cytokines in the TG. These data indicate that TLR8 contributes to the maintenance of TNP through increasing MAPK-mediated neuroinflammation. Targeting TLR8 signaling may be effective for the treatment of TNP.


Subject(s)
Neuralgia , Toll-Like Receptor 8 , Trigeminal Neuralgia , Animals , Hyperalgesia , Mice , Trigeminal Ganglion
9.
J Exp Med ; 215(12): 3019-3037, 2018 12 03.
Article in English | MEDLINE | ID: mdl-30455267

ABSTRACT

Toll-like receptors (TLRs) are nucleic acid-sensing receptors and have been implicated in mediating pain and itch. Here we report that Tlr8 -/- mice show normal itch behaviors, but have defects in neuropathic pain induced by spinal nerve ligation (SNL) in mice. SNL increased TLR8 expression in small-diameter IB4+ DRG neurons. Inhibition of TLR8 in the DRG attenuated SNL-induced pain hypersensitivity. Conversely, intrathecal or intradermal injection of TLR8 agonist, VTX-2337, induced TLR8-dependent pain hypersensitivity. Mechanistically, TLR8, localizing in the endosomes and lysosomes, mediated ERK activation, inflammatory mediators' production, and neuronal hyperexcitability after SNL. Notably, miR-21 was increased in DRG neurons after SNL. Intrathecal injection of miR-21 showed the similar effects as VTX-2337 and inhibition of miR-21 in the DRG attenuated neuropathic pain. The present study reveals a previously unknown role of TLR8 in the maintenance of neuropathic pain, suggesting that miR-21-TLR8 signaling may be potential new targets for drug development against this type of chronic pain.


Subject(s)
Chronic Pain/immunology , Ganglia, Spinal/immunology , MicroRNAs/immunology , Neuralgia/immunology , Signal Transduction/immunology , Toll-Like Receptor 8/immunology , Animals , Chronic Pain/drug therapy , Chronic Pain/genetics , Chronic Pain/pathology , Endosomes/immunology , Endosomes/pathology , Ganglia, Spinal/pathology , Lysosomes/immunology , Lysosomes/pathology , Mice , Mice, Inbred ICR , Mice, Knockout , MicroRNAs/genetics , Neuralgia/drug therapy , Neuralgia/genetics , Neuralgia/pathology , Neurons/immunology , Neurons/pathology , Signal Transduction/genetics , Toll-Like Receptor 8/genetics
10.
J Neurosci ; 38(49): 10535-10551, 2018 12 05.
Article in English | MEDLINE | ID: mdl-30373770

ABSTRACT

G-protein-coupled receptors are considered to be cell-surface sensors of extracellular signals, thereby having a crucial role in signal transduction and being the most fruitful targets for drug discovery. G-protein-coupled receptor 151 (GPR151) was reported to be expressed specifically in the habenular area. Here we report the expression and the epigenetic regulation of GRP151 in the spinal cord after spinal nerve ligation (SNL) and the contribution of GPR151 to neuropathic pain in male mice. SNL dramatically increased GPR151 expression in spinal neurons. GPR151 mutation or spinal inhibition by shRNA alleviated SNL-induced mechanical allodynia and heat hyperalgesia. Interestingly, the CpG island in the GPR151 gene promoter region was demethylated, the expression of DNA methyltransferase 3b (DNMT3b) was decreased, and the binding of DNMT3b with GPR151 promoter was reduced after SNL. Overexpression of DNMT3b in the spinal cord decreased GPR151 expression and attenuated SNL-induced neuropathic pain. Furthermore, Krüppel-like factor 5 (KLF5), a transcriptional factor of the KLF family, was upregulated in spinal neurons, and the binding affinity of KLF5 with GPR151 promoter was increased after SNL. Inhibition of KLF5 reduced GPR151 expression and attenuated SNL-induced pain hypersensitivity. Further mRNA microarray analysis revealed that mutation of GPR151 reduced the expression of a variety of pain-related genes in response to SNL, especially mitogen-activated protein kinase (MAPK) signaling pathway-associated genes. This study reveals that GPR151, increased by DNA demethylation and the enhanced interaction with KLF5, contributes to the maintenance of neuropathic pain via increasing MAPK pathway-related gene expression.SIGNIFICANCE STATEMENT G-protein-coupled receptors (GPCRs) are targets of various clinically approved drugs. Here we report that SNL increased GPR151 expression in the spinal cord, and mutation or inhibition of GPR151 alleviated SNL-induced neuropathic pain. In addition, SNL downregulated the expression of DNMT3b, which caused demethylation of GPR151 gene promoter, facilitated the binding of transcriptional factor KLF5 with the GPR151 promoter, and further increased GPR151 expression in spinal neurons. The increased GPR151 may contribute to the pathogenesis of neuropathic pain via activating MAPK signaling and increasing pain-related gene expression. Our study reveals an epigenetic mechanism underlying GPR151 expression and suggests that targeting GPR151 may offer a new strategy for the treatment of neuropathic pain.


Subject(s)
Demethylation , Kruppel-Like Transcription Factors/metabolism , Neuralgia/metabolism , Promoter Regions, Genetic/physiology , Receptors, G-Protein-Coupled/metabolism , Spinal Cord/metabolism , Animals , Base Sequence , Kruppel-Like Transcription Factors/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Mice, Transgenic , Neuralgia/genetics , Neuralgia/pathology , Protein Binding/physiology , Receptors, G-Protein-Coupled/genetics , Spinal Cord/pathology
11.
Neuropsychopharmacology ; 43(11): 2320-2330, 2018 10.
Article in English | MEDLINE | ID: mdl-29993042

ABSTRACT

Patients with neuropathic pain are usually accompanied by depression. Chemokine-mediated neuroinflammation is involved in a variety of diseases, including neurodegenerative diseases, depression, and chronic pain. The nucleus accumbens (NAc) is an important area in mediating pain sensation and depression. Here we report that spinal nerve ligation (SNL) induced upregulation of chemokine CCL2 and its major receptor CCR2 in both dopamine D1 and D2 receptor (D1R and D2R)-containing neurons in the NAc. Inhibition of CCR2 by shRNA lentivirus in the NAc shell attenuated SNL-induced pain hypersensitivity and depressive behaviors. Conversely, intra-NAc injection of CCL2-expressing lentivirus-induced nociceptive and depressive behaviors in naïve mice. Whole-cell patch clamp recording of D1R-positive or D2R-positive medium spiny neurons (MSNs) showed that SNL increased NMDA receptor (NMDAR)-mediated currents that are induced by stimulation of prefrontal cortical afferents to MSNs, which was inhibited by a CCR2 antagonist. Furthermore, Ccr2 shRNA also reduced NMDAR-mediated currents, and this reduction was mainly mediated via NR2B subunit. Consistently, NR2B, colocalized with CCR2 in the NAc, was phosphorylated after SNL and was inhibited by intra-NAc injection of Ccr2 shRNA. Furthermore, SNL or CCL2 induced ERK activation in the NAc. Inhibition of ERK by a MEK inhibitor reduced NR2B phosphorylation induced by SNL or CCL2. Finally, intra-NAc injection of NR2B antagonist or MEK inhibitor attenuated SNL-induced pain hypersensitivity and depressive behaviors. Collectively, these results suggest that CCL2/CCR2 signaling in the NAc shell is important in mediating neuropathic pain and depression via regulating NR2B-mediated NMDAR function in D1R- and D2R-containing neurons following peripheral nerve injury.


Subject(s)
Depression/metabolism , Neuralgia/metabolism , Nucleus Accumbens/metabolism , Receptors, CCR2/metabolism , Receptors, Dopamine D1/metabolism , Receptors, Dopamine D2/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Depression/physiopathology , Depression/psychology , Excitatory Amino Acid Antagonists/pharmacology , Male , Mice , Mice, Inbred ICR , Neuralgia/physiopathology , Neuralgia/psychology , Neurons/drug effects , Neurons/metabolism , Nucleus Accumbens/drug effects , Organ Culture Techniques , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors
12.
Brain Res Bull ; 139: 235-242, 2018 05.
Article in English | MEDLINE | ID: mdl-29550454

ABSTRACT

Visceral pain, observed in inflammatory bowel disease (IBD) patients, is a challenging medical problem and remains poorly understood because the mechanisms underlying it are unclear. Emerging evidence indicates that microRNAs (miRNAs) play a crucial role in the pathogenesis of acute and chronic pain. In this study, we aimed to explore the potential role of miR-146a-5p (the mature form of miR-146a) in a mouse model of colitis induced by intracolonic injection of trinitrobenzene sulfonic acid (TNBS). We found that induction of colitis resulted in visceral hyperalgesia manifested by a decreased pain threshold to colorectal distension and upregulation of miR-146a-5p expression in the lumbosacral spinal cord. In situ hybridization and immunohistochemistry results showed that miR-146a-5p was colocalized with neuronal marker NeuN, but not with astrocytic marker GFAP or microglial marker IBA-1. Dual-luciferase reporter assay showed that miR-146a-5p directly targeted the 3'-untranslated region (UTR) of CCL8, which was previously identified as an important regulator of visceral pain. In cultured Neuro-2a cells, TNF-α-induced CCL8 upregulation was decreased by transfection of miR-146a-5p mimic dose-dependently. In vivo, exogenous supplementation of miR-146a-5p by intrathecal miR-146a-5p agomir significantly alleviated visceral pain and decreased CCL8 expression in colitis mice. Furthermore, inhibition of CCL8 expression by CCL8 siRNA relieved colitis-induced visceral nociception. Finally, in naïve mice intrathecal miR-146a-5p antagomir upregulated CCL8 expression and induced visceral pain hypersensitivity, which could be partially rescued by neutralization of CCL8. Taken together, the present findings indicate that miR-146a-5p may be an endogenous suppressor of visceral pain and exogenous supplementation of miR-146a-5p could exert an analgesic effect at least partly by targeting spinal CCL8 expression. Thus, miR-146a-5p may serve as a novel therapeutic target for visceral pain intervention in the context of colitis.


Subject(s)
Chemokine CCL8/metabolism , Colitis/complications , Gene Expression Regulation/genetics , MicroRNAs/therapeutic use , Spinal Cord/metabolism , Visceral Pain , Animals , Antagomirs/therapeutic use , Antibodies/therapeutic use , Cells, Cultured , Chemokine CCL8/chemistry , Chemokine CCL8/genetics , Chemokine CCL8/immunology , Colitis/chemically induced , Colitis/pathology , Disease Models, Animal , Gene Expression Regulation/drug effects , Humans , Hyperalgesia/metabolism , Male , Mice , Mice, Inbred C57BL , MicroRNAs/chemistry , MicroRNAs/metabolism , Peroxidase/metabolism , RNA, Small Interfering/therapeutic use , Spinal Cord/drug effects , Trinitrobenzenesulfonic Acid/toxicity , Tumor Necrosis Factor-alpha/pharmacology , Up-Regulation/drug effects , Visceral Pain/etiology , Visceral Pain/pathology , Visceral Pain/therapy
13.
Neurosci Bull ; 34(1): 54-63, 2018 Feb.
Article in English | MEDLINE | ID: mdl-28401489

ABSTRACT

Recent studies have shown that the chemokine receptor CXCR3 and its ligand CXCL10 in the dorsal root ganglion mediate itch in experimental allergic contact dermatitis (ACD). CXCR3 in the spinal cord also contributes to the maintenance of neuropathic pain. However, whether spinal CXCR3 is involved in acute or chronic itch remains unclear. Here, we report that Cxcr3 -/- mice showed normal scratching in acute itch models but reduced scratching in chronic itch models of dry skin and ACD. In contrast, both formalin-induced acute pain and complete Freund's adjuvant-induced chronic inflammatory pain were reduced in Cxcr3 -/- mice. In addition, the expression of CXCR3 and CXCL10 was increased in the spinal cord in the dry skin model induced by acetone and diethyl ether followed by water (AEW). Intrathecal injection of a CXCR3 antagonist alleviated AEW-induced itch. Furthermore, touch-elicited itch (alloknesis) after compound 48/80 or AEW treatment was suppressed in Cxcr3 -/- mice. Finally, AEW-induced astrocyte activation was inhibited in Cxcr3 -/- mice. Taken together, these data suggest that spinal CXCR3 mediates chronic itch and alloknesis, and targeting CXCR3 may provide effective treatment for chronic pruritus.


Subject(s)
Pruritus/pathology , Receptors, CXCR3/metabolism , Spinal Cord/metabolism , Acetamides/therapeutic use , Animals , Chemokine CXCL10/metabolism , Chloroquine/toxicity , Chronic Disease , Cyclopropanes/adverse effects , Dehydration/complications , Dinitrofluorobenzene/adverse effects , Disease Models, Animal , Formaldehyde/toxicity , Freund's Adjuvant/toxicity , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/drug effects , Pain/chemically induced , Pruritus/chemically induced , Pyrimidines/therapeutic use , Receptors, CXCR3/antagonists & inhibitors , Receptors, CXCR3/genetics , Skin/pathology , Spinal Cord/drug effects , Spinal Cord/pathology , Time Factors , p-Methoxy-N-methylphenethylamine/toxicity
14.
Brain Res Bull ; 135: 170-178, 2017 Oct.
Article in English | MEDLINE | ID: mdl-29037608

ABSTRACT

Visceral hypersensitivity induced by inflammatory bowel disease (IBD) is a clinical challenge since the underlying mechanisms remain elusive. Chemokines and their receptors have been suggested to modulate inflammatory pain and neuropathic pain. However, the exact chemokines involved in visceral pain remain to be determined. Here, we investigated the effects of spinal chemokine CCL8 and its major receptor CCR5 on the development of visceral hyperalgesia. We showed that intracolonic injection of 2,4,6-trinitrobenzene sulfonic acid (TNBS) in mice produced significant colonic inflammation and visceral hypersensitivity to colorectal distension. Moreover, the mRNA and protein expression of CCL8 and CCR5 in the lumbosacral spinal cord were significantly upregulated. Both of CCL8 and CCR5 were expressed in spinal neurons. Furthermore, TNBS induced the activation of extracellular signal-regulated kinase (ERK) in the spinal cord. The induction of visceral pain by TNBS was attenuated by injection of ERK upstream kinase (MEK) inhibitor PD98059. Finally, intrathecal CCL8 neutralizing antibody or CCR5 antagonist DAPTA dose-dependently suppressed TNBS-evoked visceral hyperalgesia and spinal ERK activation. Taken together, these data demonstrated that CCL8 and CCR5, expressed and upregulated in spinal neurons after colonic inflammation, are involved in the maintenance of visceral hyperalgesia via the activation of spinal ERK. Targeting CCL8/CCR5/ERK pathway in the spinal cord might provide a novel treatment for the relief of visceral pain.


Subject(s)
Chemokine CCL8/metabolism , Colitis/physiopathology , Visceral Pain/metabolism , Animals , Chemokine CCL8/physiology , Colitis/chemically induced , Disease Models, Animal , Gene Expression Regulation/genetics , Hyperalgesia/metabolism , Hyperalgesia/pathology , Male , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinases/metabolism , Neuralgia/metabolism , Neurons/metabolism , Receptors, CCR5/metabolism , Receptors, CCR5/physiology , Spinal Cord/metabolism , Spinal Cord/physiopathology , Up-Regulation , Visceral Pain/physiopathology
15.
Inflammation ; 40(3): 762-769, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28155010

ABSTRACT

Recent data demonstrated that chemokine CXCL13 mediates neuroinflammation and contributes to the maintenance of neuropathic pain after nerve injury in the spinal cord. Pro-nociceptive chemokines activate mitogen-activated protein kinases (MAPKs) which are potential signaling pathways contributing to the nociceptive behavior in inflammatory or neuropathic pain. However, whether activation of p38 and JNK MAPK signaling pathway in the trigeminal ganglion (TG) are involved in CXCL13 and its receptor CXCR5-mediated orofacial pain has not yet been clarified. Here, we show that the unilateral partial infraorbital nerve ligation (pIONL) induced a profound orofacial pain in wild-type (WT) mice. Western blot results showed that pIONL induced p38 but not JNK activation in the TG of WT mice. However, the orofacial pain induced by pIONL was alleviated in Cxcr5 -/- mice, and the activation of p38 was also abrogated in Cxcr5 -/- mice. Furthermore, intra-TG injection of CXCL13 evoked mechanical hypersensitivity and increased p-p38 expression in WT mice. But CXCL13 had no effect on pain behavior or p-p38 expression in Cxcr5 -/- mice. Finally, pretreatment with p38 inhibitor, SB203580, attenuated the pIONL-induced mechanical allodynia and decreased the mRNA expression of pro-inflammatory cytokines including tumor necrosis factor-α (TNF-α) and interleukin-1ß (IL-1ß) in the TG. Taken together, our data suggest that CXCL13 acts on CXCR5 to increase p38 activation and further contributes to the pathogenesis of orofacial neuropathic pain.


Subject(s)
Chemokine CXCL13/physiology , Eye Injuries/metabolism , Facial Pain/etiology , Trigeminal Ganglion/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Eye/innervation , Eye Injuries/pathology , Mice , Neurogenic Inflammation/etiology , Receptors, CXCR5/physiology
16.
J Neurosci ; 37(3): 685-700, 2017 01 18.
Article in English | MEDLINE | ID: mdl-28100749

ABSTRACT

DNA methylation has been implicated in the pathogenesis of chronic pain. However, the specific genes regulated by DNA methylation under neuropathic pain condition remain largely unknown. Here we investigated how chemokine receptor CXCR3 is regulated by DNA methylation and how it contributes to neuropathic pain induced by spinal nerve ligation (SNL) in mice. SNL increased Cxcr3 mRNA and protein expression in the neurons of the spinal cord. Meanwhile, the CpG (5'-cytosine-phosphate-guanine-3') island in the Cxcr3 gene promoter region was demethylated, and the expression of DNA methyltransferase 3b (DNMT3b) was decreased. SNL also increased the binding of CCAAT (cytidine-cytidine-adenosine-adenosine-thymidine)/enhancer binding protein α (C/EBPα) with Cxcr3 promoter and decreased the binding of DNMT3b with Cxcr3 promoter in the spinal cord. C/EBPα expression was increased in spinal neurons after SNL, and inhibition of C/EBPα by intrathecal small interfering RNA attenuated SNL-induced pain hypersensitivity and reduced Cxcr3 expression. Furthermore, SNL-induced mechanical allodynia and heat hyperalgesia were markedly reduced in Cxcr3-/- mice. Spinal inhibition of Cxcr3 by shRNA or CXCR3 antagonist also attenuated established neuropathic pain. Moreover, CXCL10, the ligand of CXCR3, was increased in spinal neurons and astrocytes after SNL. Superfusing spinal cord slices with CXCL10 enhanced spontaneous EPSCs and potentiated NMDA-induced and AMPA-induced currents of lamina II neurons. Finally, intrathecal injection of CXCL10 induced CXCR3-dependent pain hypersensitivity in naive mice. Collectively, our results demonstrated that CXCR3, increased by DNA demethylation and the enhanced interaction with C/EBPα, can be activated by CXCL10 to facilitate excitatory synaptic transmission and contribute to the maintenance of neuropathic pain. SIGNIFICANCE STATEMENT: Peripheral nerve injury induces changes of gene expression in the spinal cord that may contribute to the pathogenesis of neuropathic pain. CXCR3 is a chemokine receptor. Whether it is involved in neuropathic pain and how it is regulated after nerve injury remain largely unknown. Our study demonstrates that spinal nerve ligation downregulates the expression of DNMT3b, which may cause demethylation of Cxcr3 gene promoter and facilitate the binding of CCAAT/enhancer binding protein α with Cxcr3 promoter and further increase CXCR3 expression in spinal neurons. The upregulated CXCR3 may contribute to neuropathic pain by facilitating central sensitization. Our study reveals an epigenetic mechanism underlying CXCR3 expression and also suggests that targeting the expression or activation of CXCR3 signaling may offer new therapeutics for neuropathic pain.


Subject(s)
CCAAT-Enhancer-Binding Protein-alpha/metabolism , Neuralgia/metabolism , Promoter Regions, Genetic/physiology , Receptors, CXCR3/metabolism , Animals , Base Sequence , CCAAT-Enhancer-Binding Protein-alpha/genetics , Methylation , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Neuralgia/genetics , Protein Binding/physiology , Receptors, CXCR3/genetics
17.
Bio Protoc ; 7(1): e2098, 2017 Jan 05.
Article in English | MEDLINE | ID: mdl-34458428

ABSTRACT

Primary afferents of sensory neurons mainly terminate in the spinal cord dorsal horn, which has an important role in the integration and modulation of sensory-related signals. Primary culture of mouse spinal dorsal horn neuron (SDHN) is useful for studying signal transmission from peripheral nervous system to the brain, as well as for developing cellular disease models, such as pain and itch. Because of the specific features of SDHN, it is necessary to establish a reliable culture method that is suitable for testing neural response to various external stimuli in vitro.

18.
Sci Rep ; 6: 34836, 2016 10 06.
Article in English | MEDLINE | ID: mdl-27708397

ABSTRACT

CXCL13 is a B lymphocyte chemoattractant and activates CXCR5 receptor in the immune system. Here we investigated whether CXCL13/CXCR5 mediates inflammatory pain in dorsal root ganglia (DRG) and the underlying mechanisms. Peripheral injection of complete Freund's Adjuvant (CFA) increased the expression of CXCL13 and CXCR5 in DRG neurons. In Cxcr5-/- mice, CFA-induced pain hypersensitivity were attenuated. Whole-cell patch-clamp recording showed that the excitability of dissociated DRG neurons was increased after CFA injection or CXCL13 incubation from wild-type (WT) mice, but not from Cxcr5-/- mice. Additionally, sodium channel Nav1.8 was co-expressed with CXCR5 in dissociated DRG neurons, and the increased neuronal excitability induced by CFA or CXCL13 was reduced by Nav1.8 blocker. Intrathecal injection of Nav1.8 blocker also attenuated intrathecal injection of CXCL13-induced pain hypersensitivity. Furthermore, CXCL13 increased Nav1.8 current density in DRG neurons, which was inhibited by p38 MAP kinase inhibitor. CFA and CXCL13 increased p38 phosphorylation in the DRG of WT mice but not Cxcr5-/- mice. Finally, intrathecal p38 inhibitor alleviated CXCL13-induced pain hypersensitivity. Taken together, these results demonstrated that CXCL13, upregulated by peripheral inflammation, acts on CXCR5 on DRG neurons and activates p38, which increases Nav1.8 current density and further contributes to the maintenance of inflammatory pain.


Subject(s)
Inflammation/metabolism , NAV1.8 Voltage-Gated Sodium Channel/metabolism , Pain/metabolism , Receptors, CXCR5/metabolism , Sensory Receptor Cells/metabolism , Animals , Cell Count , Chemokine CXCL13/genetics , Chemokine CXCL13/metabolism , Disease Models, Animal , Freund's Adjuvant/toxicity , Ganglia, Spinal/cytology , Ganglia, Spinal/metabolism , Inflammation/physiopathology , Male , Mice, Inbred ICR , Mice, Knockout , NAV1.8 Voltage-Gated Sodium Channel/genetics , Receptors, CXCR5/genetics , Sensory Receptor Cells/pathology , p38 Mitogen-Activated Protein Kinases/metabolism
19.
Brain Res Bull ; 127: 219-225, 2016 10.
Article in English | MEDLINE | ID: mdl-27697507

ABSTRACT

Chemokines and their receptors have been demonstrated to be important contributors to the development and maintenance of chronic pain. Recent studies showed that chemokine (C-X-C motif) ligand 1 (CXCL1) and its major receptor CXCR2 are respectively expressed in astrocytes and neurons in the spinal cord and are involved in the maintenance of neuropathic pain and inflammatory pain via astrocytic-neuronal interaction. Here we investigated how CXCL1 and CXCR2 are regulated in the dorsal root ganglion (DRG) after peripheral injection of complete Freund's adjuvant (CFA) and its implication in inflammatory pain. CFA induced rapid increase of CXCL1 mRNA and protein in the DRG. Double immunostaining showed that CXCL1 was colocalized with calcitonin gene-related peptide (CGRP), isolectin B4 (IB4), and neurofilament 200 (NF200). Furthermore, CXCR2 mRNA was increased 3h after CFA injection and maintained for more than 3 days. CXCR2 protein was also increased and colocalized with CGRP, IB4, and NF200. Finally, perisciatic nerve injection of CXCR2 siRNA to specifically knock down CXCR2 in the DRG effectively attenuated CFA-induced mechanical allodynia and heat hyperalgesia, and the effect maintained for more than 5 days. Taken together, our results demonstrated that CXCL1 and CXCR2 may regulate the maintenance of inflammatory pain via an autocrine/paracrine way in DRG neurons.


Subject(s)
Ganglia, Spinal/metabolism , Inflammation/metabolism , Pain/metabolism , Receptors, Interleukin-8B/metabolism , Animals , Calcitonin Gene-Related Peptide/metabolism , Chemokine CXCL1/metabolism , Freund's Adjuvant , Ganglia, Spinal/pathology , Glycoproteins/metabolism , Inflammation/pathology , Male , Mice, Inbred ICR , Neurofilament Proteins/metabolism , Pain/pathology , RNA, Messenger/metabolism , Receptors, Interleukin-8B/antagonists & inhibitors , Time Factors
20.
J Neuroinflammation ; 13(1): 183, 2016 07 11.
Article in English | MEDLINE | ID: mdl-27401148

ABSTRACT

BACKGROUND: Trigeminal nerve damage-induced neuropathic pain is a severely debilitating chronic orofacial pain syndrome. Spinal chemokine CXCL13 and its receptor CXCR5 were recently demonstrated to play a pivotal role in the pathogenesis of spinal nerve ligation-induced neuropathic pain. Whether and how CXCL13/CXCR5 in the trigeminal ganglion (TG) mediates orofacial pain are unknown. METHODS: The partial infraorbital nerve ligation (pIONL) was used to induce trigeminal neuropathic pain in mice. The expression of ATF3, CXCL13, CXCR5, and phosphorylated extracellular signal-regulated kinase (pERK) in the TG was detected by immunofluorescence staining and western blot. The effect of shRNA targeting on CXCL13 or CXCR5 on pain hypersensitivity was checked by behavioral testing. RESULTS: pIONL induced persistent mechanical allodynia and increased the expression of ATF3, CXCL13, and CXCR5 in the TG. Inhibition of CXCL13 or CXCR5 by shRNA lentivirus attenuated pIONL-induced mechanical allodynia. Additionally, pIONL-induced neuropathic pain and the activation of ERK in the TG were reduced in Cxcr5 (-/-) mice. Furthermore, MEK inhibitor (PD98059) attenuated mechanical allodynia and reduced TNF-α and IL-1ß upregulation induced by pIONL. TNF-α inhibitor (Etanercept) and IL-1ß inhibitor (Diacerein) attenuated pIONL-induced orofacial pain. Finally, intra-TG injection of CXCL13 induced mechanical allodynia, increased the activation of ERK and the production of TNF-α and IL-1ß in the TG of WT mice, but not in Cxcr5 (-/-) mice. Pretreatment with PD98059, Etanercept, or Diacerein partially blocked CXCL13-induced mechanical allodynia, and PD98059 also reduced CXCL13-induced TNF-α and IL-1ß upregulation. CONCLUSIONS: CXCL13 and CXCR5 contribute to orofacial pain via ERK-mediated proinflammatory cytokines production. Targeting CXCL13/CXCR5/ERK/TNF-α and IL-1ß pathway in the trigeminal ganglion may offer effective treatment for orofacial neuropathic pain.


Subject(s)
Chemokine CXCL13/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Facial Pain/metabolism , Facial Pain/pathology , Motor Neurons/metabolism , Trigeminal Ganglion/pathology , Activating Transcription Factor 3/metabolism , Animals , Chemokine CXCL13/genetics , Disease Models, Animal , Facial Pain/genetics , Hyperalgesia/metabolism , Male , Mice , Mice, Inbred ICR , Mice, Transgenic , Pain Measurement , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Receptors, CXCR5/genetics , Receptors, CXCR5/metabolism , Signal Transduction/genetics , Time Factors , Tubulin/metabolism , Up-Regulation/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...