scholarly journals Intraarticular Adeno‐Associated Virus Serotype AAV‐PHP.S–Mediated Chemogenetic Targeting of Knee‐Innervating Dorsal Root Ganglion Neurons Alleviates Inflammatory Pain in Mice

2020 ◽  
Vol 72 (10) ◽  
pp. 1749-1758 ◽  
Author(s):  
Sampurna Chakrabarti ◽  
Luke A. Pattison ◽  
Balint Doleschall ◽  
Rebecca H. Rickman ◽  
Helen Blake ◽  
...  
Neuroreport ◽  
1998 ◽  
Vol 9 (6) ◽  
pp. 967-972 ◽  
Author(s):  
Masaki Tanaka ◽  
Theodore R. Cummins ◽  
Kuniko Ishikawa ◽  
Sulayman D. Dib-Hajj ◽  
Joel A. Black ◽  
...  

2021 ◽  
Vol 22 (19) ◽  
pp. 10360
Author(s):  
Ling Chen ◽  
Qiguo Hu ◽  
Huaicun Liu ◽  
Yan Zhao ◽  
Sun-On Chan ◽  
...  

The microtubule, a major constituent of cytoskeletons, was shown to bind and interact with transient receptor potential vanilloid subfamily member 1 (TRPV1), and serves a pivotal role to produce thermal hyperalgesia in inflammatory pain. Nogo-A is a modulator of microtubule assembly and plays a key role in maintaining the function of TRPV1 in inflammatory heat pain. However, whether the microtubule dynamics modulated by Nogo-A in dorsal root ganglion (DRG) neurons participate in the inflammatory pain is not elucidated. Here we reported that the polymerization of microtubules in the DRG neurons, as indicated by the acetylated α-tubulin, tubulin polymerization-promoting protein 3 (TPPP3), and microtubule numbers, was significantly elevated in the complete Freund’s adjuvant (CFA) induced inflammatory pain. Consistent with our previous results, knock-out (KO) of Nogo-A protein significantly attenuated the heat hyperalgesia 72 h after CFA injection and decreased the microtubule polymerization via up-regulation of phosphorylation of collapsin response mediator protein 2 (CRMP2) in DRG. The colocalization of acetylated α-tubulin and TRPV1 in DRG neurons was also reduced dramatically in Nogo-A KO rats under inflammatory pain. Moreover, the down-regulation of TRPV1 in DRG of Nogo-A KO rats after injection of CFA was reversed by intrathecal injection of paclitaxel, a microtubule stabilizer. Furthermore, intrathecal injection of nocodazole (a microtubule disruptor) attenuated significantly the CFA-induced inflammatory heat hyperalgesia and the mechanical pain in a rat model of spared nerve injury (SNI). In these SNI cases, the Nogo-A and acetylated α-tubulin in DRG were also significantly up-regulated. We conclude that the polymerization of microtubules promoted by Nogo-A in DRG contributes to the development of inflammatory heat hyperalgesia mediated by TRPV1.


Pain ◽  
2015 ◽  
Vol 156 (4) ◽  
pp. 597-608 ◽  
Author(s):  
Xing Xiao ◽  
Xiao-Tao Zhao ◽  
Ling-Chi Xu ◽  
Lu-Peng Yue ◽  
Feng-Yu Liu ◽  
...  

2010 ◽  
Vol 21 (15) ◽  
pp. 2568-2577 ◽  
Author(s):  
Masako Nakanishi ◽  
Kenji Hata ◽  
Tomotaka Nagayama ◽  
Teruhisa Sakurai ◽  
Toshihiko Nishisho ◽  
...  

Increased production of calcitonin gene-related peptide (CGRP) in sensory neurons is implicated in inflammatory pain. The inflammatory site is acidic due to proton release from infiltrating inflammatory cells. Acid activation of peripheral nociceptors relays pain signals to the CNS. Here, we examined whether acid activated the transient receptor potential vanilloid subtype 1 (Trpv1), a widely recognized acid-sensing nociceptor and subsequently increased CGRP expression. Chemically induced inflammation was associated with thermal hyperalgesia and increased CGRP expression in dorsal root ganglion (DRG) in rats. In organ cultures of DRG, acid (pH 5.5) elevated CGRP expression and the selective Trpv1 antagonist 5′-Iodoresiniferatoxin decreased it. Trpv1-deficient DRG showed reduced CGRP increase by acid. Of note, many of CGRP/Trpv1-positive DRG neurons exhibited the phosphorylation of cAMP response element-binding protein (CREB), a nociceptive transcription factor. Knockdown of CREB by small interfering RNA or a dominant-negative form of CREB diminished acid-elevated CGRP expression. Acid elevated the transcriptional activity of CREB, which in turn stimulated CGRP gene promoter activity. These effects were inhibited by a Ca2+/calmodulin-dependent protein kinase (CaMK) inhibitor KN-93. In conclusion, our results suggest that inflammatory acidic environments activate Trpv1, leading to an up-regulation of CGRP expression via CaMK-CREB cascade, a series of events that may be associated with inflammatory pain.


Sign in / Sign up

Export Citation Format

Share Document