Mas-related G protein-coupled receptor D exacerbates inflammatory hyperalgesia by promoting NF-κB activation in dorsal root ganglia

2020 ◽  
Author(s):  
Lei Lan ◽  
Miao Xu ◽  
Jia Li ◽  
Lin Liu ◽  
Min Xu ◽  
...  

Abstract Background Mas-related G protein–coupled receptor D (MrgprD) is mainly expressed in small-diameter sensory neurons of the dorsal root ganglion (DRG). Results from previous studies suggest that MrgprD participates in mechanical allodynia and nerve injury-induced neuropathic pain. However, it remains elusive whether and how MrgprD is involved in inflammatory pain. Methods we used a mouse model of inflammatory hyperalgesia established by intraperitoneal administration of lipopolysaccharide (LPS). The generation of neuroinflammation and inflammatory hyperalgesia were determined using animal behavioral tests, Western blotting, quantitative real-time PCR and ELISA assay. The expression levels of MrgprD were assessed by Western blotting. The involvements of MrgprD in the LPS-triggered neuroinflammation and inflammation-related signaling pathways were assessed by animal behavioral tests, Western blotting, immunohistochemistry, ELISA, and co-immunoprecipitation assays. Results The LPS injection induced an evident peripheral neuroinflammation and mechanical allodynia in the mice and increased MrgprD expression in the DRG. The LPS administration also augmented the proportion of MrgprD-expressing neurons in the lumbar 4 DRG. The LPS-induced hypersensitivities to mechanical and cold stimuli, but not to a heat stimulus, were substantially attenuated in Mrgprd- knockout mice compared with wildtype littermates. Mrgprd deletion suppressed the LPS-triggered activation of the NF-κB signaling pathway and attenuated LPS-induced up-regulation of proinflammatory cytokines. Moreover, ectopic overexpression of MrgprD in HEK293 cells stably expressing mouse toll-like receptor 4 (TLR4) markedly promoted the LPS-induced NF-κB activation and enhanced NF-κB’s DNA-binding activity. Furthermore, MrgprD physically interacted with TGF-β-activated kinase 1 (TAK1) and I-kappa-B-kinase (IKK) complexes in DRGs and macrophage-like RAW 264.7 cells, and MrgprD agonist treatment was sufficient to elicit the activation of NF-κB signaling, but not of mitogen-activated protein kinases (MAPKs) in RAW 264.7 cells. Conclusions Our findings demonstrate that MrgprD exacerbates LPS-induced inflammatory hyperalgesia by promoting canonical NF-κB activation, indicating that MrgprD could be a potential therapeutic target for managing NF-κB-mediated inflammation and inflammatory pain.

2015 ◽  
Vol 2015 ◽  
pp. 1-7
Author(s):  
Huan Liu ◽  
Shen-Bin Liu ◽  
Qian Li ◽  
Huijing Wang ◽  
Yan-Qing Wang ◽  
...  

Acupuncture or electroacupuncture (EA) has been demonstrated to have a powerful antihypernociceptive effect on inflammatory pain. The attenuation of G protein-coupled receptor kinase 2 (GRK2) in spinal cord and peripheral nociceptor has been widely acknowledged to promote the transition from acute to chronic pain and to facilitate the nociceptive progress. This study was designed to investigate the possible role of spinal GRK2 in EA antiallodynic in a rat model with complete Freund’s adjuvant (CFA) induced inflammatory pain. EA was applied to ST36 (“Zusanli”) and BL60 (“Kunlun”) one day after CFA injection. Single EA treatment at day 1 after CFA injection remarkably alleviated CFA induced mechanical allodynia two hours after EA. Repeated EA displayed significant antiallodynic effect from 2nd EA treatment and a persistent effect was observed during the rest of treatments. However, downregulation of spinal GRK2 by intrathecal exposure of GRK2 antisense 30 mins after EA treatment completely eliminated both the transient and persistent antiallodynic effect by EA treatment. These pieces of data demonstrated that the spinal GRK2 played an important role in EA antiallodynia on inflammatory pain.


2009 ◽  
Vol 425 (1) ◽  
pp. 169-180 ◽  
Author(s):  
Sonika Patial ◽  
Jiansong Luo ◽  
Katie J. Porter ◽  
Jeffrey L. Benovic ◽  
Narayanan Parameswaran

TNFα (tumour necrosis factor α) is a multifunctional cytokine involved in the pathophysiology of many chronic inflammatory diseases. TNFα activation of the NF-κB (nuclear factor κB) signalling pathway particularly in macrophages has been implicated in many diseases. We demonstrate in the present study that GRK2 and GRK5 (G-protein-coupled-receptor kinases 2 and 5) regulate TNFα-induced NF-κB signalling in Raw 264.7 macrophages. RNAi (RNA interference) knockdown of GRK2 or GRK5 in macrophages significantly inhibited TNFα-induced IκBα (inhibitory κBα) phosphorylation and degradation, NF-κB activation and expression of the NF-κB-regulated gene MIP1β (macrophage inflammatory protein 1β). Consistent with these results, overexpression of GRK2 or GRK5 enhanced TNFα-induced NF-κB activity. In addition, we show that GRK2 and GRK5 interacted with IκBα via the N-terminal domain of IκBα and that IκBα is a substrate for GRK2 and GRK5 in vitro. Furthermore, we also found that GRK5, but not GRK2, phosphorylated IκBα at the same amino acid residues (Ser32/Ser36) as that of IKKβ (IκB kinase β). Interestingly, associated with these results, knockdown of IKKβ in Raw 264.7 macrophages did not affect TNFα-induced IκBα phosphorylation. Taken together, these results demonstrate that both GRK2 and GRK5 are important and novel mediators of a non-traditional IκBα/NF-κB signalling pathway.


2007 ◽  
Vol 25 (6) ◽  
pp. 1696-1704 ◽  
Author(s):  
Wendy Kleibeuker ◽  
Annemarie Ledeboer ◽  
Niels Eijkelkamp ◽  
Linda R. Watkins ◽  
Steven F. Maier ◽  
...  

2009 ◽  
Vol 5 ◽  
pp. 1744-8069-5-39 ◽  
Author(s):  
Ying-Ju Chen ◽  
Chia-Wei Huang ◽  
Chih-Shin Lin ◽  
Wen-Han Chang ◽  
Wei-Hsin Sun

2019 ◽  
Author(s):  
Alexandra Hogea ◽  
Shihab Shah ◽  
Frederick Jones ◽  
Chase M Carver ◽  
Han Hao ◽  
...  

AbstractJunctions of endoplasmic reticulum and plasma membrane (ER-PM junctions) serve as signaling hubs in prokaryotic cells. ER-PM junctions are present in peripheral sensory neurons and are necessary for pro-inflammatory G protein coupled receptor signalling and for inflammatory pain generation. Yet, the principles of ER-PM junctions assembly and maintenance, as well as their role in inflammatory signaling in sensory neurons are only beginning to emerge. Here we discovered that a member of the junctophilin family of proteins, JPH4, is abundantly expressed in rat dorsal root ganglion (DRG) neurons and is necessary for the formation of store operated Ca2+ entry (SOCE) complex at the ER-PM junctions in response to the G-protein induced ER Ca2+ store depletion. Furthermore, we demonstrate a key role of the JPH4 and ER Ca2+ stores in the maintenance of inflammatory pain. Indeed, knockdown of JPH4 expression in DRG in vivo significantly reduced the duration of pain produced by inflammatory mediator bradykinin. Since the ER supplies Ca2+ for the excitatory action of multiple inflammatory mediators, we suggest that junctional Ca2+ signalling maintained by JPH4 is an important contributor to the inflammatory pain mechanisms.


Sign in / Sign up

Export Citation Format

Share Document