Cargando…

Low-affinity neurotrophin receptor p75 of brain-derived neurotrophic factor contributes to cancer-induced bone pain by upregulating mTOR signaling

Crucial to the development and maintenance of pain sensations is neurotrophin receptor p75 (p75(NTR)), the low affinity receptor of brain-derived neurotrophic factor (BDNF). This receptor is widespread among dorsal root ganglion (DRG) neurons and the spinal cord. Few reports have demonstrated the sp...

Descripción completa

Detalles Bibliográficos
Autores principales: Meng, Xiao-Wen, Jin, Xiao-Hong, Wei, Xiang, Wang, Li-Na, Yang, Jian-Ping, Ji, Fu-Hai
Formato: Online Artículo Texto
Lenguaje:English
Publicado: D.A. Spandidos 2019
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6862244/
https://www.ncbi.nlm.nih.gov/pubmed/31777542
http://dx.doi.org/10.3892/etm.2019.8097
_version_ 1783471509343305728
author Meng, Xiao-Wen
Jin, Xiao-Hong
Wei, Xiang
Wang, Li-Na
Yang, Jian-Ping
Ji, Fu-Hai
author_facet Meng, Xiao-Wen
Jin, Xiao-Hong
Wei, Xiang
Wang, Li-Na
Yang, Jian-Ping
Ji, Fu-Hai
author_sort Meng, Xiao-Wen
collection PubMed
description Crucial to the development and maintenance of pain sensations is neurotrophin receptor p75 (p75(NTR)), the low affinity receptor of brain-derived neurotrophic factor (BDNF). This receptor is widespread among dorsal root ganglion (DRG) neurons and the spinal cord. Few reports have demonstrated the specific role of p75(NTR) in the development of cancer-induced bone pain (CIBP). Therefore the present study examined whether p75(NTR) contributed to CIBP by upregulating mammalian target of rapamycin (mTOR) signaling. A CIBP rat model was induced and reverse transcription-quantitative polymerase chain reaction was employed to determine p75(NTR) and mTOR mRNA expression. Immunofluorescence analysis was performed to determine the coexpression of p75(NTR) and mTOR in DRG neurons, as well as the spinal cord. Von Frey filaments were used to measure the 50% likelihood of paw withdrawal thresholds (PWTs). Spontaneous pain was assessed by ambulatory score. The results demonstrated that compared with the control group, mTOR activation in primary cultured DRG neurons was significantly increased. In addition, mTOR and p75(NTR) expression was significantly enhanced in the BDNF-treated primary DRG in the BDNF group. In vivo experiments determined that mTOR and p75(NTR) levels were increased in the CIBP rats compared with the sham group. PWT, in response to mechanical stimulation, was significantly lower compared with that in sham rats and the ambulatory score was significantly higher than that in sham rats. Finally, intrathecal injection of a p75(NTR)-targeting small interfering RNA significantly decreased mTOR and p75(NTR) expression levels in DRG neurons and the spinal cord of CIBP rats, as well as partially reversing the decline in PWTs and the increase in ambulatory score. In conclusion, the present study determined that the activation of BDNF/p75(NTR)/mTOR signaling may participate in nociceptive transmission in CIBP, suggesting a novel mechanism and potential therapeutic target for CIBP treatment and management.
format Online
Article
Text
id pubmed-6862244
institution National Center for Biotechnology Information
language English
publishDate 2019
publisher D.A. Spandidos
record_format MEDLINE/PubMed
spelling pubmed-68622442019-11-27 Low-affinity neurotrophin receptor p75 of brain-derived neurotrophic factor contributes to cancer-induced bone pain by upregulating mTOR signaling Meng, Xiao-Wen Jin, Xiao-Hong Wei, Xiang Wang, Li-Na Yang, Jian-Ping Ji, Fu-Hai Exp Ther Med Articles Crucial to the development and maintenance of pain sensations is neurotrophin receptor p75 (p75(NTR)), the low affinity receptor of brain-derived neurotrophic factor (BDNF). This receptor is widespread among dorsal root ganglion (DRG) neurons and the spinal cord. Few reports have demonstrated the specific role of p75(NTR) in the development of cancer-induced bone pain (CIBP). Therefore the present study examined whether p75(NTR) contributed to CIBP by upregulating mammalian target of rapamycin (mTOR) signaling. A CIBP rat model was induced and reverse transcription-quantitative polymerase chain reaction was employed to determine p75(NTR) and mTOR mRNA expression. Immunofluorescence analysis was performed to determine the coexpression of p75(NTR) and mTOR in DRG neurons, as well as the spinal cord. Von Frey filaments were used to measure the 50% likelihood of paw withdrawal thresholds (PWTs). Spontaneous pain was assessed by ambulatory score. The results demonstrated that compared with the control group, mTOR activation in primary cultured DRG neurons was significantly increased. In addition, mTOR and p75(NTR) expression was significantly enhanced in the BDNF-treated primary DRG in the BDNF group. In vivo experiments determined that mTOR and p75(NTR) levels were increased in the CIBP rats compared with the sham group. PWT, in response to mechanical stimulation, was significantly lower compared with that in sham rats and the ambulatory score was significantly higher than that in sham rats. Finally, intrathecal injection of a p75(NTR)-targeting small interfering RNA significantly decreased mTOR and p75(NTR) expression levels in DRG neurons and the spinal cord of CIBP rats, as well as partially reversing the decline in PWTs and the increase in ambulatory score. In conclusion, the present study determined that the activation of BDNF/p75(NTR)/mTOR signaling may participate in nociceptive transmission in CIBP, suggesting a novel mechanism and potential therapeutic target for CIBP treatment and management. D.A. Spandidos 2019-12 2019-10-14 /pmc/articles/PMC6862244/ /pubmed/31777542 http://dx.doi.org/10.3892/etm.2019.8097 Text en Copyright: © Meng et al. This is an open access article distributed under the terms of the Creative Commons Attribution-NonCommercial-NoDerivs License (https://creativecommons.org/licenses/by-nc-nd/4.0/) , which permits use and distribution in any medium, provided the original work is properly cited, the use is non-commercial and no modifications or adaptations are made.
spellingShingle Articles
Meng, Xiao-Wen
Jin, Xiao-Hong
Wei, Xiang
Wang, Li-Na
Yang, Jian-Ping
Ji, Fu-Hai
Low-affinity neurotrophin receptor p75 of brain-derived neurotrophic factor contributes to cancer-induced bone pain by upregulating mTOR signaling
title Low-affinity neurotrophin receptor p75 of brain-derived neurotrophic factor contributes to cancer-induced bone pain by upregulating mTOR signaling
title_full Low-affinity neurotrophin receptor p75 of brain-derived neurotrophic factor contributes to cancer-induced bone pain by upregulating mTOR signaling
title_fullStr Low-affinity neurotrophin receptor p75 of brain-derived neurotrophic factor contributes to cancer-induced bone pain by upregulating mTOR signaling
title_full_unstemmed Low-affinity neurotrophin receptor p75 of brain-derived neurotrophic factor contributes to cancer-induced bone pain by upregulating mTOR signaling
title_short Low-affinity neurotrophin receptor p75 of brain-derived neurotrophic factor contributes to cancer-induced bone pain by upregulating mTOR signaling
title_sort low-affinity neurotrophin receptor p75 of brain-derived neurotrophic factor contributes to cancer-induced bone pain by upregulating mtor signaling
topic Articles
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6862244/
https://www.ncbi.nlm.nih.gov/pubmed/31777542
http://dx.doi.org/10.3892/etm.2019.8097
work_keys_str_mv AT mengxiaowen lowaffinityneurotrophinreceptorp75ofbrainderivedneurotrophicfactorcontributestocancerinducedbonepainbyupregulatingmtorsignaling
AT jinxiaohong lowaffinityneurotrophinreceptorp75ofbrainderivedneurotrophicfactorcontributestocancerinducedbonepainbyupregulatingmtorsignaling
AT weixiang lowaffinityneurotrophinreceptorp75ofbrainderivedneurotrophicfactorcontributestocancerinducedbonepainbyupregulatingmtorsignaling
AT wanglina lowaffinityneurotrophinreceptorp75ofbrainderivedneurotrophicfactorcontributestocancerinducedbonepainbyupregulatingmtorsignaling
AT yangjianping lowaffinityneurotrophinreceptorp75ofbrainderivedneurotrophicfactorcontributestocancerinducedbonepainbyupregulatingmtorsignaling
AT jifuhai lowaffinityneurotrophinreceptorp75ofbrainderivedneurotrophicfactorcontributestocancerinducedbonepainbyupregulatingmtorsignaling