Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 91
Filter
1.
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
2.
J Neurosci ; 43(8): 1334-1347, 2023 02 22.
Article in English | MEDLINE | ID: mdl-36653189

ABSTRACT

Itch is an uncomfortable and complex sensation that elicits the desire to scratch. The nucleus accumbens (NAc) activity is important in driving sensation, motivation, and emotion. Excitatory afferents from the medial prefrontal cortex (mPFC), amygdala, and hippocampus are crucial in tuning the activity of dopamine receptor D1-expressing and D2-expressing medium spiny neurons (Drd1-MSN and Drd2-MSN) in the NAc. However, a cell-type and neural circuity-based mechanism of the NAc underlying acute itch remains unclear. We found that acute itch induced by compound 48/80 (C48/80) decreased the intrinsic membrane excitability in Drd1-MSNs, but not in Drd2-MSNs, in the NAc core of male mice. Chemogenetic activation of Drd1-MSNs alleviated C48/80-induced scratching behaviors but not itch-related anxiety-like behaviors. In addition, C48/80 enhanced the frequency of spontaneous EPSCs (sEPSCs) and reduced the paired-pulse ratio (PPR) of electrical stimulation-evoked EPSCs in Drd1-MSNs. Furthermore, C48/80 increased excitatory synaptic afferents to Drd1-MSNs from the mPFC, not from the basolateral amygdala (BLA) or ventral hippocampus (vHipp). Consistently, the intrinsic excitability of mPFC-NAc projecting pyramidal neurons was increased after C48/80 treatment. Chemogenetic inhibition of mPFC-NAc excitatory synaptic afferents relieved the scratching behaviors. Moreover, pharmacological activation of κ opioid receptor (KOR) in the NAc core suppressed C48/80-induced scratching behaviors, and the modulation of KOR activity in the NAc resulted in the changes of presynaptic excitatory inputs to Drd1-MSNs in C48/80-treated mice. Together, these results reveal the neural plasticity in synapses of NAc Drd1-MSNs from the mPFC underlying acute itch and indicate the modulatory role of the KOR in itch-related scratching behaviors.SIGNIFICANCE STATEMENT Itch stimuli cause strongly scratching desire and anxiety in patients. However, the related neural mechanisms remain largely unclear. In the present study, we demonstrated that the pruritogen compound 48/80 (C48/80) shapes the excitability of dopamine receptor D1-expressing medium spiny neurons (Drd1-MSNs) in the nucleus accumbens (NAc) core and the glutamatergic synaptic afferents from medial prefrontal cortex (mPFC) to these neurons. Chemogenetic activation of Drd1-MSNs or inhibition of mPFC-NAc excitatory synaptic afferents relieves the scratching behaviors. In addition, pharmacological activation of κ opioid receptor (KOR) in the NAc core alleviates C48/80-induced itch. Thus, targeting mPFC-NAc Drd1-MSNs or KOR may provide effective treatments for itch.


Subject(s)
Nucleus Accumbens , Receptors, Opioid, kappa , Mice , Male , Animals , Nucleus Accumbens/physiology , Hippocampus/physiology , Neurons/physiology , Receptors, Dopamine D1/metabolism , Prefrontal Cortex/metabolism
3.
Mol Pain ; 19: 17448069231169373, 2023.
Article in English | MEDLINE | ID: mdl-36998150

ABSTRACT

BACKGROUND: Chemokine-mediated neuroinflammation plays an important role in the pathogenesis of neuropathic pain. The chemokine CC motif ligand 7 (CCL7) and its receptor CCR2 have been reported to contribute to neuropathic pain via astrocyte-microglial interaction in the spinal cord. Whether CCL7 in the trigeminal ganglion (TG) involves in trigeminal neuropathic pain and the involved mechanism remain largely unknown. METHODS: The partial infraorbital nerve transection (pIONT) was used to induce trigeminal neuropathic pain in mice. The expression of Ccl7, Ccr1, Ccr2, and Ccr3 was examined by real-time quantitative polymerase chain reaction. The distribution of CCL7, CCR2, and CCR3 was detected by immunofluorescence double-staining. The activation of extracellular signal-regulated kinase (ERK) was examined by Western blot and immunofluorescence. The effect of CCL7 on neuronal excitability was tested by whole-cell patch clamp recording. The effect of selective antagonists for CCR1, CCR2, and CCR3 on pain hypersensitivity was checked by behavioral testing. RESULTS: Ccl7 was persistently increased in neurons of TG after pIONT, and specific inhibition of CCL7 in the TG effectively relieved pIONT-induced orofacial mechanical allodynia. Intra-TG injection of recombinant CCL7 induced mechanical allodynia and increased the phosphorylation of ERK in the TG. Incubation of CCL7 with TG neurons also dose-dependently enhanced the neuronal excitability. Furthermore, pIONT increased the expression of CCL7 receptors Ccr1, Ccr2, and Ccr3. The intra-TG injection of the specific antagonist of CCR2 or CCR3 but not of CCR1 alleviated pIONT-induced orofacial mechanical allodynia and reduced ERK activation. Immunostaining showed that CCR2 and CCR3 are expressed in TG neurons, and CCL7-induced hyperexcitability of TG neurons was decreased by antagonists of CCR2 or CCR3. CONCLUSION: CCL7 activates ERK in TG neurons via CCR2 and CCR3 to enhance neuronal excitability, which contributes to the maintenance of trigeminal neuropathic pain. CCL7-CCR2/CCR3-ERK pathway may be potential targets for treating trigeminal neuropathic pain.


Subject(s)
Chemokine CCL7 , Extracellular Signal-Regulated MAP Kinases , Neuralgia , Trigeminal Neuralgia , Animals , Mice , Chemokine CCL2/metabolism , Chemokine CCL7/metabolism , Chemokine CCL7/pharmacology , Extracellular Signal-Regulated MAP Kinases/metabolism , Hyperalgesia/metabolism , Ligands , MAP Kinase Signaling System , Neuralgia/metabolism , Trigeminal Ganglion/metabolism , Trigeminal Neuralgia/metabolism , Receptors, CCR2/metabolism , Receptors, CCR3/metabolism
4.
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
5.
Mol Pain ; 18: 17448069211053255, 2022.
Article in English | MEDLINE | ID: mdl-35057644

ABSTRACT

N-methyl-d-aspartate receptors (NMDARs) dysfunction in the nucleus accumbens (NAc) participates in regulating many neurological and psychiatric disorders such as drug addiction, chronic pain, and depression. NMDARs are heterotetrameric complexes generally composed of two NR1 and two NR2 subunits (NR2A, NR2B, NR2C and NR2D). Much attention has been focused on the role of NR2A and NR2B-containing NMDARs in a variety of neurological disorders; however, the function of NR2C/2D subunits at NAc in chronic pain remains unknown. In this study, spinal nerve ligation (SNL) induced a persistent sensory abnormity and depressive-like behavior. The whole-cell patch clamp recording on medium spiny neurons (MSNs) in the NAc showed that the amplitude of NMDAR-mediated excitatory postsynaptic currents (EPSCs) was significantly increased when membrane potential held at -40 to 0 mV in mice after 14 days of SNL operation. In addition, selective inhibition of NR2C/2D-containing NMDARs with PPDA caused a larger decrease on peak amplitude of NMDAR-EPSCs in SNL than that in sham-operated mice. Appling of selective potentiator of NR2C/2D, CIQ, markedly enhanced the evoked NMDAR-EPSCs in SNL-operated mice, but no change in sham-operated mice. Finally, intra-NAc injection of PPDA significantly attenuated SNL-induced mechanical allodynia and depressive-like behavior. These results for the first time showed that the functional change of NR2C/2D subunits-containing NMDARs in the NAc might contribute to the sensory and affective components in neuropathic pain.


Subject(s)
Neuralgia , Peripheral Nerve Injuries , Animals , Depression/etiology , Humans , Mice , Nucleus Accumbens , Peripheral Nerve Injuries/complications , Receptors, N-Methyl-D-Aspartate/metabolism
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
7.
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
8.
J Neuroinflammation ; 16(1): 209, 2019 Nov 10.
Article in English | MEDLINE | ID: mdl-31707979

ABSTRACT

BACKGROUND: Paclitaxel is a widely used and potent chemotherapeutic agent for the treatment of cancer. However, patients receiving paclitaxel often develop an acute pain syndrome for which there are few treatment options. Astrocytes play an important role in the pathogenesis of pain in multiple preclinical models, as well as in paclitaxel-treated rodents. However, it is still unclear what the exact contribution of astrocytes may be in paclitaxel-associated acute pain syndrome (P-APS). METHODS: P-APS was modeled by a single systemic or intrathecal injection of paclitaxel and astrocyte contribution tested by immunohistochemical, pharmacological, and behavioral approaches. Cell cultures were also prepared to assess whether paclitaxel treatment directly activates astrocytes and whether intrathecal injection of paclitaxel-treated astrocytes produces pain that is reminiscent of P-APS. RESULTS: Systemic injection of paclitaxel resulted in increased expression of glial fibrillary acidic protein (a common marker of astrocytic activation), as well as both systemic or intrathecal injection of paclitaxel induced pain hypersensitivity indicated by the development of mechanical allodynia, which was significantly reversed by the astrocytic inhibitor L-α-AA. Cultured astrocytes were activated by paclitaxel with significant increases in protein levels for tumor necrosis factor-α (TNF-α) and stromal-derived cell factor 1 (SDF-1). Importantly, intrathecal injection of paclitaxel-activated astrocytes produced mechanical allodynia that was reversed by TNF-α and SDF-1 neutralizing antibodies. CONCLUSION: Our results suggest for the first time that paclitaxel can directly activate astrocytes, which are sufficient to produce acute pain by releasing TNF-α and SDF-1. Targeting astrocytes and these cytokines may offer new treatments for P-APS.


Subject(s)
Antineoplastic Agents, Phytogenic/toxicity , Astrocytes/metabolism , Chemokine CXCL12/metabolism , Hyperalgesia/metabolism , Paclitaxel/toxicity , Tumor Necrosis Factor-alpha/metabolism , Animals , Astrocytes/drug effects , Female , Hyperalgesia/chemically induced , Male , Mice
9.
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
10.
Mol Pain ; 14: 1744806918777401, 2018.
Article in English | MEDLINE | ID: mdl-29712506

ABSTRACT

Chemokines-mediated neuroinflammation in the spinal cord plays a critical role in the pathogenesis of neuropathic pain. Chemokine CXCL9, CXCL10, and CXCL11 have been identified as a same subfamily chemokine which bind to CXC chemokine receptor 3 to exert functions. Our recent work found that CXCL10 is upregulated in spinal astrocytes after spinal nerve ligation (SNL) and acts on chemokine receptor CXCR3 on neurons to contribute to central sensitization and neuropathic pain, but less is known about CXCL9 and CXCL11 in the maintenance of neuropathic pain. Here, we report that CXCL9 and CXCL11, same as CXCL10, were increased in spinal astrocytes after SNL. Surprisingly, inhibition of CXCL9 or CXCL11 by spinal injection of shRNA lentivirus did not attenuate SNL-induced neuropathic pain. In addition, intrathecal injection of CXCL9 and CXCL11 did not produce hyperalgesia or allodynia behaviors, and neither of them induced ERK activation, a marker of central sensitization. Whole-cell patch clamp recording on spinal neurons showed that CXCL9 and CXCL11 enhanced both excitatory synaptic transmission and inhibitory synaptic transmission, whereas CXCL10 only produced an increase in excitatory synaptic transmission. These results suggest that, although the expression of CXCL9 and CXCL11 are increased after SNL, they may not contribute to the maintenance of neuropathic pain.


Subject(s)
Chemokine CXCL11/genetics , Chemokine CXCL9/genetics , Neuralgia/genetics , Spinal Cord/metabolism , Spinal Nerves/injuries , Spinal Nerves/metabolism , Up-Regulation/genetics , Animals , Astrocytes/metabolism , Astrocytes/pathology , Chemokine CXCL11/metabolism , Chemokine CXCL9/metabolism , Enzyme Activation , Excitatory Postsynaptic Potentials , Extracellular Signal-Regulated MAP Kinases/metabolism , Ligation , Male , Mice, Inbred ICR , Neural Inhibition , Neuralgia/enzymology , Neuralgia/pathology , Neurons/metabolism , Neurons/pathology , Spinal Cord/pathology , Spinal Cord/physiopathology , Spinal Nerves/pathology , Spinal Nerves/physiopathology
11.
Cell Mol Life Sci ; 74(18): 3275-3291, 2017 09.
Article in English | MEDLINE | ID: mdl-28389721

ABSTRACT

Neuropathic pain resulting from damage or dysfunction of the nervous system is a highly debilitating chronic pain state and is often resistant to currently available treatments. It has become clear that neuroinflammation, mainly mediated by proinflammatory cytokines and chemokines, plays an important role in the establishment and maintenance of neuropathic pain. Chemokines were originally identified as regulators of peripheral immune cell trafficking and were also expressed in neurons and glial cells in the central nervous system. In recent years, accumulating studies have revealed the expression, distribution and function of chemokines in the spinal cord under chronic pain conditions. In this review, we provide evidence showing that several chemokines are upregulated after peripheral nerve injury and contribute to the pathogenesis of neuropathic pain via different forms of neuron-glia interaction in the spinal cord. First, chemokine CX3CL1 is expressed in primary afferents and spinal neurons and induces microglial activation via its microglial receptor CX3CR1 (neuron-to-microglia signaling). Second, CCL2 and CXCL1 are expressed in spinal astrocytes and act on CCR2 and CXCR2 in spinal neurons to increase excitatory synaptic transmission (astrocyte-to-neuron signaling). Third, we recently identified that CXCL13 is highly upregulated in spinal neurons after spinal nerve ligation and induces spinal astrocyte activation via receptor CXCR5 (neuron-to-astrocyte signaling). Strategies that target chemokine-mediated neuron-glia interactions may lead to novel therapies for the treatment of neuropathic pain.


Subject(s)
Chemokines/metabolism , Neuralgia/pathology , Neuroglia/metabolism , Neurons/metabolism , Animals , Neuralgia/metabolism , Receptors, Chemokine/metabolism , Signal Transduction , Spinal Cord/metabolism
12.
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
13.
Cell Mol Neurobiol ; 36(1): 143-9, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26115624

ABSTRACT

Ligustilide is a major component of Radix Angelica Sinensis and reported to have anti-inflammatory and anti-nociceptive effects. Toll-like receptor 4 (TLR4) has been shown to be expressed in the spinal cord and be involved in inflammatory pain and neuropathic pain. Whether ligustilide can inhibit spinal TLR4 expression in inflammatory pain is still unknown. In the present study, we intravenously injected ligustilide daily for 4 days, with the first injection given at 1 h before complete Freund's adjuvant (CFA) injection. We tested the analgesic effect of ligustilide by behavioral test and checked the expression and distribution of TLR4 in the spinal cord by real-time quantitative PCR, Western blot, and immunofluorescence. Our data showed that repeated daily intravenous treatment with ligustilide alleviated CFA-induced heat hyperalgesia and mechanical allodynia. The same treatment also inhibited CFA-induced TLR4 mRNA and protein increase in the spinal cord. Immunofluorescence double staining showed that TLR4 was predominantly expressed in spinal astrocytes. In primary cultured astrocytes, ligustilide dose-dependently reduced lipopolysaccharide-induced upregulation of TLR4 mRNA expression. These data indicate that ligustilide treatment reduces TLR4 expression in spinal astrocytes and is an effective therapy for inflammatory pain.


Subject(s)
4-Butyrolactone/analogs & derivatives , Astrocytes/metabolism , Inflammation/drug therapy , Pain/drug therapy , Spinal Cord/pathology , Toll-Like Receptor 4/genetics , Up-Regulation/drug effects , 4-Butyrolactone/pharmacology , 4-Butyrolactone/therapeutic use , Animals , Astrocytes/drug effects , Cells, Cultured , Freund's Adjuvant/administration & dosage , Hot Temperature , Hyperalgesia/complications , Hyperalgesia/drug therapy , Hyperalgesia/genetics , Inflammation/genetics , Inflammation/pathology , Male , Mice, Inbred ICR , Pain/complications , Pain/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Toll-Like Receptor 4/metabolism
14.
Neurobiol Dis ; 79: 100-10, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25956228

ABSTRACT

Chemotherapy drugs such as vincristine can produce painful peripheral neuropathy for which is still lack of effective treatment. Recent studies have demonstrated that neuroinflammation plays an important role in the pathogenesis of neuropathic pain. Heme oxygenase 1 (HO-1) was shown to mediate the resolution of inflammation. In this study, we investigated the contribution of HO-1 in the modulation of vincristine-induced pain and the mechanisms implicated. Injection of vincristine induced persistent mechanical allodynia and thermal hyperalgesia in mice. The expression of HO-1 mRNA and protein was increased in 2 weeks in the spinal cord. Immunostaining showed that HO-1 was mainly expressed in neurons of spinal cord dorsal horn in naïve animals, but induced in astrocytes and microglia after vincristine injection. Intraperitoneal injection of HO-1 inducer increased HO-1 expression in the spinal cord and attenuated vincristine-induced pain. Persistent induction of HO-1 by intraspinal injection of HO-1-expressing lentivirus alleviated vincristine-induced pain for more than 2 weeks. Furthermore, vincristine induced activation of glial cells (astrocytes and microglia), phosphorylation of MAPKs (JNK, ERK, and p38), and production of TNF-α and monocyte chemoattractant protein-1 in the spinal cord, which were all reduced by intrathecal injection of HO-1 inducer. Taken together, our data provide the first evidence that induction of HO-1 attenuates vincristine-induced neuropathic pain via inhibition of glia-mediated neuroinflammation in the spinal cord. This suggests that exogenously induced HO-1 may have potential as therapy in chemotherapy-induced neuropathic pain.


Subject(s)
Antineoplastic Agents, Phytogenic/toxicity , Heme Oxygenase-1/metabolism , Membrane Proteins/metabolism , Neuralgia/therapy , Neuroglia/enzymology , Spinal Cord/enzymology , Vincristine/toxicity , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Disease Models, Animal , Enzyme Activators/pharmacology , Genetic Therapy , Heme Oxygenase-1/genetics , Hyperalgesia/chemically induced , Hyperalgesia/enzymology , Hyperalgesia/immunology , Hyperalgesia/therapy , Lentivirus/genetics , Male , Membrane Proteins/genetics , Mice, Inbred ICR , Neuralgia/chemically induced , Neuralgia/enzymology , Neuralgia/immunology , Neuroglia/drug effects , Neuroglia/immunology , Neurons/drug effects , Neurons/enzymology , Neurons/immunology , Pain/chemically induced , Pain/drug therapy , Pain/enzymology , Pain/immunology , Peripheral Nervous System Diseases/chemically induced , Peripheral Nervous System Diseases/drug therapy , Peripheral Nervous System Diseases/enzymology , Peripheral Nervous System Diseases/immunology , Protoporphyrins/pharmacology , RNA, Messenger/metabolism , Spinal Cord/drug effects , Spinal Cord/immunology
15.
Mol Pain ; 11: 43, 2015 Jul 17.
Article in English | MEDLINE | ID: mdl-26184882

ABSTRACT

BACKGROUND: Neuropathic pain that caused by lesion or dysfunction of the nervous system is associated with gene expression changes in the sensory pathway. Long noncoding RNAs (lncRNAs) have been reported to be able to regulate gene expression. Identifying lncRNA expression patterns in the spinal cord under normal and neuropathic pain conditions is essential for understanding the genetic mechanisms behind the pathogenesis of neuropathic pain. RESULTS: Spinal nerve ligation (SNL) induced rapid and persistent pain hypersensitivity, characterized by mechanical allodynia and heat hyperalgesia. Meanwhile, astrocytes and microglia were dramatically activated in the ipsilateral spinal cord dorsal horn at 10 days after SNL. Further lncRNA microarray and mRNA microarray analysis showed that the expression profiles of lncRNA and mRNA between SNL and sham-operated mice were greatly changed at 10 days. The 511 differentially expressed (>2 fold) lncRNAs (366 up-regulated, 145 down-regulated) and 493 mRNAs (363 up-regulated, 122 down-regulated) were finally identified. The expression patterns of several lncRNAs and mRNAs were further confirmed by qPCR. Functional analysis of differentially expressed (DE) mRNAs showed that the most significant enriched biological processes of up-regulated genes in SNL include immune response, defense response, and inflammation response, which are important pathogenic mechanisms underlying neuropathic pain. 35 DE lncRNAs have neighboring or overlapping DE mRNAs in genome, which is related to Toll-like receptor signaling, cytokine-cytokine receptor interaction, and peroxisome proliferator-activated receptor signaling pathway. CONCLUSION: Our findings uncovered the expression pattern of lncRNAs and mRNAs in the mice spinal cord under neuropathic pain condition. These lncRNAs and mRNAs may represent new therapeutic targets for the treatment of neuropathic pain.


Subject(s)
Gene Expression Profiling , Neuralgia/genetics , Spinal Cord/metabolism , Spinal Nerves/pathology , Animals , Disease Models, Animal , Down-Regulation/genetics , Ligation , Male , Mice, Inbred ICR , Neuralgia/pathology , Neuroglia/metabolism , Neuroglia/pathology , Oligonucleotide Array Sequence Analysis , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Reproducibility of Results , Spinal Cord/pathology , Spinal Nerves/metabolism , Up-Regulation/genetics
16.
Brain Behav Immun ; 49: 119-29, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25957028

ABSTRACT

Glia-mediated neuroinflammation plays an important role in the pathogenesis of neuropathic pain. Our recent study demonstrated that TNF receptor associated factor-6 (TRAF6) is expressed in spinal astrocytes and contributes to the maintenance of spinal nerve ligation (SNL)-induced neuropathic pain. MicroRNA (miR)-146a is a key regulator of the innate immune response and was shown to target TRAF6 and reduce inflammation. In this study, we found that in cultured astrocytes, TNF-α, IL-1ß, or lipopolysaccharide (LPS) induced rapid TRAF6 upregulation and delayed miR-146a-5p upregulation. In addition, miR-146a-5p mimic blocked LPS-induced TRAF6 upregulation, as well as LPS-induced c-Jun N-terminal kinase (JNK) activation and chemokine CCL2 expression in astrocytes. Notably, LPS incubation with astrocytes enhanced the DNA binding activity of AP-1 to the promoters of mir-146a and ccl2. TRAF6 siRNA or JNK inhibitor SP600125 significantly reduced LPS-induced miR-146a-5p increase in astrocytes. In vivo, intrathecal injection of TNF-α or LPS increased spinal TRAF6 expression. Pretreatment with miR-146a-5p mimic alleviated TNF-α- or LPS-induced mechanical allodynia and reduced TRAF6 expression. Finally, SNL induced miR-146a-5p upregulation in the spinal cord at 10 and 21days. Intrathecal injection of miR-146a-5p mimic attenuated SNL-induced mechanical allodynia and decreased spinal TRAF6 expression. Taken together, the results suggest that (1) miR-146a-5p attenuates neuropathic pain partly through inhibition of TRAF6 and its downstream JNK/CCL2 signaling, (2) miR-146a-5p is increased by the activation of TRAF6/JNK pathway. Hence, miR-146a-5p may be a novel treatment for chronic neuropathic pain.


Subject(s)
Astrocytes/metabolism , MicroRNAs/metabolism , Neuralgia/metabolism , Spinal Cord/metabolism , TNF Receptor-Associated Factor 6/metabolism , Animals , Cells, Cultured , Chemokine CCL2/metabolism , Hyperalgesia/chemically induced , Hyperalgesia/metabolism , Lipopolysaccharides , MAP Kinase Signaling System , Male , Mice, Inbred ICR , Signal Transduction , Tumor Necrosis Factor-alpha/administration & dosage
17.
Eur J Neurosci ; 39(8): 1391-402, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24521480

ABSTRACT

Ligustilide (LIG) is a major component of Radix Angelica Sinensis, and reportedly has neuroprotective and anti-inflammatory effects. Recent studies have demonstrated that spinal astrocyte-mediated neuroinflammation plays an important role in the pathogenesis of chronic pain. Here we investigated the anti-nociceptive effect of systemic treatment with LIG on chronic inflammatory pain and explored possible mechanisms. Unilateral hindpaw injection of complete Freund's adjuvant (CFA) induced persistent pain hypersensitivity. Repeated daily intravenous treatment with LIG, either before or after CFA injection, attenuated CFA-induced thermal hyperalgesia and mechanical allodynia. The same treatment also inhibited CFA-induced keratinocyte-derived chemokine (KC) and monocyte chemoattractant protein-1 (MCP-1) mRNA and protein increases in astrocytes of the spinal cord. In vitro study showed LIG dose-dependently reduced lipopolysaccharide (LPS)-induced upregulation of KC and MCP-1 mRNA in astrocyte cultures. Interestingly, LIG treatment did not affect CFA- or LPS-induced glial fibrillary acidic protein upregulation, but did inhibit CFA-induced phosphorylated nuclear factor-κB (p-NFκB) upregulation in spinal astrocytes. Furthermore, intrathecal injection of NFκB inhibitor attenuated CFA-induced pain hypersensitivity and upregulation of KC and MCP-1 in the spinal cord. Finally, single intravenous injection of LIG attenuated intrathecal injection of LPS-induced mechanical allodynia. The same treatment also decreased LPS-induced NFκB activation and KC and MCP-1 upregulation in the spinal cord. These data indicate that LIG attenuates chronic inflammatory pain potentially via inhibiting NFκB-mediated chemokines production in spinal astrocytes. These results provide direct evidence of the anti-nociceptive and anti-inflammatory effects of LIG, suggesting a new application of LIG for the treatment of chronic inflammatory pain.


Subject(s)
4-Butyrolactone/analogs & derivatives , Astrocytes/drug effects , Chemokine CCL2/metabolism , Chemokines/metabolism , NF-kappa B/metabolism , Nociceptive Pain/drug therapy , Spinal Cord/metabolism , 4-Butyrolactone/pharmacology , 4-Butyrolactone/therapeutic use , Angelica sinensis/chemistry , Animals , Astrocytes/metabolism , Cells, Cultured , Chemokine CCL2/genetics , Chemokines/genetics , Hyperalgesia/drug therapy , Male , Mice , Mice, Inbred ICR , NF-kappa B/genetics , Nociception/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Spinal Cord/drug effects , Spinal Cord/pathology
18.
J Neuroinflammation ; 11: 38, 2014 Mar 01.
Article in English | MEDLINE | ID: mdl-24580964

ABSTRACT

BACKGROUND: Bone cancer pain (BCP) is one of the most disabling factors in patients suffering from primary bone cancer or bone metastases. Recent studies show several chemokines (for example, CCL2, CXCL10) in the spinal cord are involved in the pathogenesis of BCP. Here we investigated whether and how spinal CXCL1 contributes to BCP. METHODS: Mouse prostate tumor cell line, RM-1 cells were intramedullary injected into the femur to induce BCP. The mRNA expression of CXCL1 and CXCR2 was detected by quantitative real-time PCR. The protein expression and distribution of CXCL1, NFκB, and CXCR2 was examined by immunofluorescence staining and western blot. The effect of CXCL1 neutralizing antibody, NFκB antagonist, and CXCR2 antagonist on pain hypersensitivity was checked by behavioral testing. RESULTS: Intramedullary injection of RM-1 cells into the femur induced cortical bone damage and persistent (>21 days) mechanical allodynia and heat hyperalgesia. Tumor cell inoculation also produced CXCL1 upregulation in activated astrocytes in the spinal cord for more than 21 days. Inhibition of CXCL1 by intrathecal administration of CXCL1 neutralizing antibody at 7 days after inoculation attenuated mechanical allodynia and heat hyperalgesia. In cultured astrocytes, TNF-α induced robust CXCL1 expression, which was dose-dependently decreased by NFκB inhibitor. Furthermore, inoculation induced persistent NFκB phosphorylation in spinal astrocytes. Intrathecal injection of NFκB inhibitor attenuated BCP and reduced CXCL1 increase in the spinal cord. Finally, CXCR2, the primary receptor of CXCL1, was upregulated in dorsal horn neurons after inoculation. Inhibition of CXCR2 by its selective antagonist SB225002 attenuated BCP. CONCLUSION: NFκB mediates CXCL1 upregulation in spinal astrocytes in the BCP model. In addition, CXCL1 may be released from astrocytes and act on CXCR2 on neurons in the spinal cord and be involved in the maintenance of BCP. Inhibition of the CXCL1 signaling may provide a new therapy for BCP management.


Subject(s)
Astrocytes/physiology , Bone Neoplasms/complications , Chemokine CXCL1/metabolism , NF-kappa B/metabolism , Pain/etiology , Pain/pathology , Spinal Cord/pathology , Animals , Animals, Newborn , Antibodies/therapeutic use , Bone Neoplasms/secondary , Cell Line, Tumor , Cells, Cultured , Cerebral Cortex/cytology , Chemokine CXCL1/genetics , Chemokine CXCL1/immunology , Disease Models, Animal , Enzyme Inhibitors/therapeutic use , Gene Expression Regulation, Neoplastic/physiology , Male , Mice , Mice, Inbred C57BL , NF-kappa B/genetics , Neoplasm Transplantation , Nitriles/pharmacology , Pain/drug therapy , Prostatic Neoplasms/pathology , Sulfones/pharmacology
19.
Brain Res Bull ; 214: 111008, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38866373

ABSTRACT

The infralimbic (IL) cortex dysfunction has been implicated in major depressive disorder (MDD), yet the precise cellular and molecular mechanisms remain poorly understood. In this study, we investigated the role of layer V pyramidal neurons in a mouse model of MDD induced by repeated lipopolysaccharide (LPS) administration. Our results demonstrate that three days of systemic LPS administration induced depressive-like behavior and upregulated mRNA levels of interleukin-1ß (IL-1ß), tumor necrosis factor-alpha (TNF-α), and transforming growth factor-ß (TGF-ß) in the IL cortex. Electrophysiological recordings revealed a significant decrease in the intrinsic excitability of layer V pyramidal neurons in the IL following systemic LPS exposure. Importantly, chemogenetic activation of IL pyramidal neurons ameliorated LPS-induced depressive-like behavior. Additionally, LPS administration significantly increased microglial activity in the IL, as evidenced by a greater number of Ionized calcium binding adaptor molecule-1 (IBA-1)-positive cells. Morphometric analysis further unveiled enlarged soma, decreased branch numbers, and shorter branch lengths of microglial cells in the IL cortex following LPS exposure. Moreover, the activation of pyramidal neurons by clozapine-N-oxide increased the microglia branch length but did not change branch number or cytosolic area. These results collectively suggest that targeted activation of pyramidal neurons in the IL cortex mitigates microglial response and ameliorates depressive-like behaviors induced by systemic LPS administration. Therefore, our findings offer potential therapeutic targets for the development of interventions aimed at alleviating depressive symptoms by modulating IL cortical circuitry and microglial activity.


Subject(s)
Lipopolysaccharides , Microglia , Pyramidal Cells , Animals , Pyramidal Cells/drug effects , Pyramidal Cells/metabolism , Lipopolysaccharides/pharmacology , Mice , Male , Microglia/drug effects , Microglia/metabolism , Mice, Inbred C57BL , Depression/chemically induced , Depression/metabolism , Depression/drug therapy , Clozapine/pharmacology , Clozapine/analogs & derivatives , Disease Models, Animal , Depressive Disorder, Major/metabolism
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL