Hmgb1 Silencing in the Amygdala Inhibits Pain-Related Behaviors in a Rat Model of Neuropathic Pain

Chronic pain presents a therapeutic challenge due to the highly complex interplay of sensory, emotional-affective and cognitive factors. The mechanisms of the transition from acute to chronic pain are not well understood. We hypothesized that neuroimmune mechanisms in the amygdala, a brain region in...

Full description

Bibliographic Details
Main Authors: Peyton Presto, Guangchen Ji, Olga Ponomareva, Igor Ponomarev, Volker Neugebauer
Format: Article
Language:English
Published: MDPI AG 2023-07-01
Series:International Journal of Molecular Sciences
Subjects:
Online Access:https://www.mdpi.com/1422-0067/24/15/11944
_version_ 1797586656439566336
author Peyton Presto
Guangchen Ji
Olga Ponomareva
Igor Ponomarev
Volker Neugebauer
author_facet Peyton Presto
Guangchen Ji
Olga Ponomareva
Igor Ponomarev
Volker Neugebauer
author_sort Peyton Presto
collection DOAJ
description Chronic pain presents a therapeutic challenge due to the highly complex interplay of sensory, emotional-affective and cognitive factors. The mechanisms of the transition from acute to chronic pain are not well understood. We hypothesized that neuroimmune mechanisms in the amygdala, a brain region involved in the emotional-affective component of pain and pain modulation, play an important role through high motility group box 1 (Hmgb1), a pro-inflammatory molecule that has been linked to neuroimmune signaling in spinal nociception. Transcriptomic analysis revealed an upregulation of Hmgb1 mRNA in the right but not left central nucleus of the amygdala (CeA) at the chronic stage of a spinal nerve ligation (SNL) rat model of neuropathic pain. Hmgb1 silencing with a stereotaxic injection of siRNA for Hmgb1 into the right CeA of adult male and female rats 1 week after (post-treatment), but not 2 weeks before (pre-treatment) SNL induction decreased mechanical hypersensitivity and emotional-affective responses, but not anxiety-like behaviors, measured 4 weeks after SNL. Immunohistochemical data suggest that neurons are a major source of Hmgb1 in the CeA. Therefore, Hmgb1 in the amygdala may contribute to the transition from acute to chronic neuropathic pain, and the inhibition of Hmgb1 at a subacute time point can mitigate neuropathic pain.
first_indexed 2024-03-11T00:26:08Z
format Article
id doaj.art-2a5c100a172d4452b6061fc598a85c0d
institution Directory Open Access Journal
issn 1661-6596
1422-0067
language English
last_indexed 2024-03-11T00:26:08Z
publishDate 2023-07-01
publisher MDPI AG
record_format Article
series International Journal of Molecular Sciences
spelling doaj.art-2a5c100a172d4452b6061fc598a85c0d2023-11-18T22:58:16ZengMDPI AGInternational Journal of Molecular Sciences1661-65961422-00672023-07-0124151194410.3390/ijms241511944Hmgb1 Silencing in the Amygdala Inhibits Pain-Related Behaviors in a Rat Model of Neuropathic PainPeyton Presto0Guangchen Ji1Olga Ponomareva2Igor Ponomarev3Volker Neugebauer4Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USADepartment of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USADepartment of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USADepartment of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USADepartment of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USAChronic pain presents a therapeutic challenge due to the highly complex interplay of sensory, emotional-affective and cognitive factors. The mechanisms of the transition from acute to chronic pain are not well understood. We hypothesized that neuroimmune mechanisms in the amygdala, a brain region involved in the emotional-affective component of pain and pain modulation, play an important role through high motility group box 1 (Hmgb1), a pro-inflammatory molecule that has been linked to neuroimmune signaling in spinal nociception. Transcriptomic analysis revealed an upregulation of Hmgb1 mRNA in the right but not left central nucleus of the amygdala (CeA) at the chronic stage of a spinal nerve ligation (SNL) rat model of neuropathic pain. Hmgb1 silencing with a stereotaxic injection of siRNA for Hmgb1 into the right CeA of adult male and female rats 1 week after (post-treatment), but not 2 weeks before (pre-treatment) SNL induction decreased mechanical hypersensitivity and emotional-affective responses, but not anxiety-like behaviors, measured 4 weeks after SNL. Immunohistochemical data suggest that neurons are a major source of Hmgb1 in the CeA. Therefore, Hmgb1 in the amygdala may contribute to the transition from acute to chronic neuropathic pain, and the inhibition of Hmgb1 at a subacute time point can mitigate neuropathic pain.https://www.mdpi.com/1422-0067/24/15/11944amygdalaneuropathic painHmgb1neuroimmune signalingbehavior
spellingShingle Peyton Presto
Guangchen Ji
Olga Ponomareva
Igor Ponomarev
Volker Neugebauer
Hmgb1 Silencing in the Amygdala Inhibits Pain-Related Behaviors in a Rat Model of Neuropathic Pain
International Journal of Molecular Sciences
amygdala
neuropathic pain
Hmgb1
neuroimmune signaling
behavior
title Hmgb1 Silencing in the Amygdala Inhibits Pain-Related Behaviors in a Rat Model of Neuropathic Pain
title_full Hmgb1 Silencing in the Amygdala Inhibits Pain-Related Behaviors in a Rat Model of Neuropathic Pain
title_fullStr Hmgb1 Silencing in the Amygdala Inhibits Pain-Related Behaviors in a Rat Model of Neuropathic Pain
title_full_unstemmed Hmgb1 Silencing in the Amygdala Inhibits Pain-Related Behaviors in a Rat Model of Neuropathic Pain
title_short Hmgb1 Silencing in the Amygdala Inhibits Pain-Related Behaviors in a Rat Model of Neuropathic Pain
title_sort hmgb1 silencing in the amygdala inhibits pain related behaviors in a rat model of neuropathic pain
topic amygdala
neuropathic pain
Hmgb1
neuroimmune signaling
behavior
url https://www.mdpi.com/1422-0067/24/15/11944
work_keys_str_mv AT peytonpresto hmgb1silencingintheamygdalainhibitspainrelatedbehaviorsinaratmodelofneuropathicpain
AT guangchenji hmgb1silencingintheamygdalainhibitspainrelatedbehaviorsinaratmodelofneuropathicpain
AT olgaponomareva hmgb1silencingintheamygdalainhibitspainrelatedbehaviorsinaratmodelofneuropathicpain
AT igorponomarev hmgb1silencingintheamygdalainhibitspainrelatedbehaviorsinaratmodelofneuropathicpain
AT volkerneugebauer hmgb1silencingintheamygdalainhibitspainrelatedbehaviorsinaratmodelofneuropathicpain