Cargando…

DNA Methylation Modulates Nociceptive Sensitization after Incision

DNA methylation is a key epigenetic mechanism controlling DNA accessibility and gene expression. Blockade of DNA methylation can significantly affect pain behaviors implicated in neuropathic and inflammatory pain. However, the role of DNA methylation with regard to postoperative pain has not yet bee...

Descripción completa

Detalles Bibliográficos
Autores principales: Sun, Yuan, Sahbaie, Peyman, Liang, DeYong, Li, Wenwu, Shi, Xiaoyou, Kingery, Paige, Clark, J. David
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Public Library of Science 2015
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4633178/
https://www.ncbi.nlm.nih.gov/pubmed/26535894
http://dx.doi.org/10.1371/journal.pone.0142046
_version_ 1782399166661525504
author Sun, Yuan
Sahbaie, Peyman
Liang, DeYong
Li, Wenwu
Shi, Xiaoyou
Kingery, Paige
Clark, J. David
author_facet Sun, Yuan
Sahbaie, Peyman
Liang, DeYong
Li, Wenwu
Shi, Xiaoyou
Kingery, Paige
Clark, J. David
author_sort Sun, Yuan
collection PubMed
description DNA methylation is a key epigenetic mechanism controlling DNA accessibility and gene expression. Blockade of DNA methylation can significantly affect pain behaviors implicated in neuropathic and inflammatory pain. However, the role of DNA methylation with regard to postoperative pain has not yet been explored. In this study we sought to investigate the role of DNA methylation in modulating incisional pain and identify possible targets under DNA methylation and contributing to incisional pain. DNA methyltranferase (DNMT) inhibitor 5-Aza-2′-deoxycytidine significantly reduced incision-induced mechanical allodynia and thermal sensitivity. Aza-2′-deoxycytidine also reduced hindpaw swelling after incision, suggesting an anti-inflammatory effect. Global DNA methylation and DNMT3b expression were increased in skin after incision, but none of DNMT1, DNMT3a or DNMT3b was altered in spinal cord or DRG. The expression of proopiomelanocortin Pomc encoding β-endorphin and Oprm1 encoding the mu-opioid receptor were upregulated peripherally after incision; moreover, Oprm1 expression was further increased under DNMT inhibitor treatment. Finally, local peripheral injection of the opioid receptor antagonist naloxone significantly exacerbated incision-induced mechanical hypersensitivity. These results suggest that DNA methylation is functionally relevant to incisional nociceptive sensitization, and that mu-opioid receptor signaling might be one methylation regulated pathway controlling sensitization after incision.
format Online
Article
Text
id pubmed-4633178
institution National Center for Biotechnology Information
language English
publishDate 2015
publisher Public Library of Science
record_format MEDLINE/PubMed
spelling pubmed-46331782015-11-13 DNA Methylation Modulates Nociceptive Sensitization after Incision Sun, Yuan Sahbaie, Peyman Liang, DeYong Li, Wenwu Shi, Xiaoyou Kingery, Paige Clark, J. David PLoS One Research Article DNA methylation is a key epigenetic mechanism controlling DNA accessibility and gene expression. Blockade of DNA methylation can significantly affect pain behaviors implicated in neuropathic and inflammatory pain. However, the role of DNA methylation with regard to postoperative pain has not yet been explored. In this study we sought to investigate the role of DNA methylation in modulating incisional pain and identify possible targets under DNA methylation and contributing to incisional pain. DNA methyltranferase (DNMT) inhibitor 5-Aza-2′-deoxycytidine significantly reduced incision-induced mechanical allodynia and thermal sensitivity. Aza-2′-deoxycytidine also reduced hindpaw swelling after incision, suggesting an anti-inflammatory effect. Global DNA methylation and DNMT3b expression were increased in skin after incision, but none of DNMT1, DNMT3a or DNMT3b was altered in spinal cord or DRG. The expression of proopiomelanocortin Pomc encoding β-endorphin and Oprm1 encoding the mu-opioid receptor were upregulated peripherally after incision; moreover, Oprm1 expression was further increased under DNMT inhibitor treatment. Finally, local peripheral injection of the opioid receptor antagonist naloxone significantly exacerbated incision-induced mechanical hypersensitivity. These results suggest that DNA methylation is functionally relevant to incisional nociceptive sensitization, and that mu-opioid receptor signaling might be one methylation regulated pathway controlling sensitization after incision. Public Library of Science 2015-11-04 /pmc/articles/PMC4633178/ /pubmed/26535894 http://dx.doi.org/10.1371/journal.pone.0142046 Text en © 2015 Sun et al http://creativecommons.org/licenses/by/4.0/ This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are properly credited.
spellingShingle Research Article
Sun, Yuan
Sahbaie, Peyman
Liang, DeYong
Li, Wenwu
Shi, Xiaoyou
Kingery, Paige
Clark, J. David
DNA Methylation Modulates Nociceptive Sensitization after Incision
title DNA Methylation Modulates Nociceptive Sensitization after Incision
title_full DNA Methylation Modulates Nociceptive Sensitization after Incision
title_fullStr DNA Methylation Modulates Nociceptive Sensitization after Incision
title_full_unstemmed DNA Methylation Modulates Nociceptive Sensitization after Incision
title_short DNA Methylation Modulates Nociceptive Sensitization after Incision
title_sort dna methylation modulates nociceptive sensitization after incision
topic Research Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4633178/
https://www.ncbi.nlm.nih.gov/pubmed/26535894
http://dx.doi.org/10.1371/journal.pone.0142046
work_keys_str_mv AT sunyuan dnamethylationmodulatesnociceptivesensitizationafterincision
AT sahbaiepeyman dnamethylationmodulatesnociceptivesensitizationafterincision
AT liangdeyong dnamethylationmodulatesnociceptivesensitizationafterincision
AT liwenwu dnamethylationmodulatesnociceptivesensitizationafterincision
AT shixiaoyou dnamethylationmodulatesnociceptivesensitizationafterincision
AT kingerypaige dnamethylationmodulatesnociceptivesensitizationafterincision
AT clarkjdavid dnamethylationmodulatesnociceptivesensitizationafterincision