当前位置: X-MOL 学术Proc. Natl. Acad. Sci. U.S.A. › 论文详情
Our official English website, www.x-mol.net, welcomes your feedback! (Note: you will need to create a separate account there.)
Endosomal signaling of delta opioid receptors is an endogenous mechanism and therapeutic target for relief from inflammatory pain.
Proceedings of the National Academy of Sciences of the United States of America ( IF 11.1 ) Pub Date : 2020-06-30 , DOI: 10.1073/pnas.2000500117
Nestor N Jimenez-Vargas 1 , Jing Gong 2 , Matthew J Wisdom 3 , Dane D Jensen 3, 4 , Rocco Latorre 3 , Alan Hegron 3 , Shavonne Teng 3 , Jesse J DiCello 5 , Pradeep Rajasekhar 5 , Nicholas A Veldhuis 5, 6 , Simona E Carbone 5 , Yang Yu 1 , Cintya Lopez-Lopez 1 , Josue Jaramillo-Polanco 1 , Meritxell Canals 7 , David E Reed 1 , Alan E Lomax 1 , Brian L Schmidt 4 , Kam W Leong 2 , Stephen J Vanner 1 , Michelle L Halls 8 , Nigel W Bunnett 9 , Daniel P Poole 6, 8
Affiliation  

Whether G protein-coupled receptors signal from endosomes to control important pathophysiological processes and are therapeutic targets is uncertain. We report that opioids from the inflamed colon activate δ-opioid receptors (DOPr) in endosomes of nociceptors. Biopsy samples of inflamed colonic mucosa from patients and mice with colitis released opioids that activated DOPr on nociceptors to cause a sustained decrease in excitability. DOPr agonists inhibited mechanically sensitive colonic nociceptors. DOPr endocytosis and endosomal signaling by protein kinase C (PKC) and extracellular signal-regulated kinase (ERK) pathways mediated the sustained inhibitory actions of endogenous opioids and DOPr agonists. DOPr agonists stimulated the recruitment of Gαi/o and β-arrestin1/2 to endosomes. Analysis of compartmentalized signaling revealed a requirement of DOPr endocytosis for activation of PKC at the plasma membrane and in the cytosol and ERK in the nucleus. We explored a nanoparticle delivery strategy to evaluate whether endosomal DOPr might be a therapeutic target for pain. The DOPr agonist DADLE was coupled to a liposome shell for targeting DOPr-positive nociceptors and incorporated into a mesoporous silica core for release in the acidic and reducing endosomal environment. Nanoparticles activated DOPr at the plasma membrane, were preferentially endocytosed by DOPr-expressing cells, and were delivered to DOPr-positive early endosomes. Nanoparticles caused a long-lasting activation of DOPr in endosomes, which provided sustained inhibition of nociceptor excitability and relief from inflammatory pain. Conversely, nanoparticles containing a DOPr antagonist abolished the sustained inhibitory effects of DADLE. Thus, DOPr in endosomes is an endogenous mechanism and a therapeutic target for relief from chronic inflammatory pain.



中文翻译:

δ阿片受体的内体信号传导是减轻炎症性疼痛的内源性机制和治疗目标。

G蛋白偶联受体是否从内体发出信号来控制重要的病理生理过程,是否为治疗靶点尚不确定。我们报告说,来自发炎结肠的阿片类药物可激活伤害感受器内体中的δ-阿片类药物受体(DOPr)。结肠炎患者和小鼠发炎的结肠粘膜的活检样品释放出阿片样物质,该阿片样物质激活了伤害感受器上的DOPr,导致兴奋性持续下降。DOPr激动剂抑制机械敏感的结肠伤害感受器。DOPr通过蛋白激酶C(PKC)和细胞外信号调节激酶(ERK)途径的胞吞和内体信号传导介导了内源性阿片类药物和DOPr激动剂的持续抑制作用。DOPr激动剂刺激了Gαi / o的募集和β-arrestin1/ 2对内体。对间隔信号的分析表明,DOPr内吞作用需要激活质膜以及细胞质和核中ERK的PKC。我们探索了一种纳米颗粒递送策略,以评估内体DOPr是否可能是疼痛的治疗靶标。DOPr激动剂DADLE与脂质体壳偶联以靶向DOPr阳性伤害感受器,并掺入中孔二氧化硅核中,以在酸性和还原性内体环境中释放。纳米颗粒在质膜上激活了DOPr,优先被DOPr表达细胞内吞,并被递送至DOPr阳性的早期内体。纳米颗粒引起内体中DOPr的持久活化,从而持续抑制伤害感受器兴奋性并减轻炎症性疼痛。相反,含有DOPr拮抗剂的纳米颗粒消除了DADLE的持续抑制作用。因此,内体中的DOPr是缓解慢性炎症性疼痛的内源性机制和治疗靶标。

更新日期:2020-06-30
down
wechat
bug