Cargando…

Streptozotocin-Induced Hyperglycemia Affects the Pharmacokinetics of Koumine and its Anti-Allodynic Action in a Rat Model of Diabetic Neuropathic Pain

Koumine (KM), the most abundant alkaloid in Gelsemium elegans, has anti-neuropathic, anti-inflammatory, and analgesic activities; thus, it has the potential to be developed as a broad-spectrum analgesic drug. However, factors determining the relationship between analgesic efficacy and the correspond...

Descripción completa

Detalles Bibliográficos
Autores principales: Ye, Li-Xiang, Huang, Hui-Hui, Zhang, Shui-Hua, Lu, Jing-Shan, Cao, Da-Xuan, Wu, Dan-Dan, Chi, Pei-Wang, Hong, Long-Hui, Wu, Min-Xia, Xu, Ying, Yu, Chang-Xi
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Frontiers Media S.A. 2021
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8156416/
https://www.ncbi.nlm.nih.gov/pubmed/34054521
http://dx.doi.org/10.3389/fphar.2021.640318
_version_ 1783699440237805568
author Ye, Li-Xiang
Huang, Hui-Hui
Zhang, Shui-Hua
Lu, Jing-Shan
Cao, Da-Xuan
Wu, Dan-Dan
Chi, Pei-Wang
Hong, Long-Hui
Wu, Min-Xia
Xu, Ying
Yu, Chang-Xi
author_facet Ye, Li-Xiang
Huang, Hui-Hui
Zhang, Shui-Hua
Lu, Jing-Shan
Cao, Da-Xuan
Wu, Dan-Dan
Chi, Pei-Wang
Hong, Long-Hui
Wu, Min-Xia
Xu, Ying
Yu, Chang-Xi
author_sort Ye, Li-Xiang
collection PubMed
description Koumine (KM), the most abundant alkaloid in Gelsemium elegans, has anti-neuropathic, anti-inflammatory, and analgesic activities; thus, it has the potential to be developed as a broad-spectrum analgesic drug. However, factors determining the relationship between analgesic efficacy and the corresponding plasma KM concentration are largely unclear. The pharmacokinetics and pharmacodynamics of KM and their optimization in the context of neuropathic pain have not been reported. We investigated the pharmacokinetics and pharmacodynamics of KM after oral administration in a streptozotocin-induced rat model of diabetic neuropathic pain (DNP) using a population approach. A first-order absorption and elimination pharmacokinetics model best described the plasma KM concentration. This pharmacokinetic model was then linked to a linear pharmacodynamic model with an effect compartment based on the measurement of the mechanical withdrawal threshold. KM was rapidly absorbed (time to maximum plasma concentration: 0.14–0.36 h) with similar values in both DNP and naïve rats, suggesting that DNP did not influence the KM absorption rate. However, the area under the curve (AUC(0–∞)) of KM in DNP rats was over 3-fold higher than that in naïve rats. The systemic clearance rate and volume of KM distribution were significantly lower in DNP rats than in naïve rats. Blood glucose value prior to KM treatment was a significant covariate for the systemic clearance rate of KM and baseline value of the threshold. Our results suggest that streptozotocin-induced hyperglycemia is an independent factor for decreased KM elimination and its anti-allodynic effects in a DNP rat model. To the best of our knowledge, this is the first study to investigate the role of DNP in the pharmacokinetics and pharmacokinetics-pharmacodynamics of KM in streptozotocin-induced diabetic rats.
format Online
Article
Text
id pubmed-8156416
institution National Center for Biotechnology Information
language English
publishDate 2021
publisher Frontiers Media S.A.
record_format MEDLINE/PubMed
spelling pubmed-81564162021-05-28 Streptozotocin-Induced Hyperglycemia Affects the Pharmacokinetics of Koumine and its Anti-Allodynic Action in a Rat Model of Diabetic Neuropathic Pain Ye, Li-Xiang Huang, Hui-Hui Zhang, Shui-Hua Lu, Jing-Shan Cao, Da-Xuan Wu, Dan-Dan Chi, Pei-Wang Hong, Long-Hui Wu, Min-Xia Xu, Ying Yu, Chang-Xi Front Pharmacol Pharmacology Koumine (KM), the most abundant alkaloid in Gelsemium elegans, has anti-neuropathic, anti-inflammatory, and analgesic activities; thus, it has the potential to be developed as a broad-spectrum analgesic drug. However, factors determining the relationship between analgesic efficacy and the corresponding plasma KM concentration are largely unclear. The pharmacokinetics and pharmacodynamics of KM and their optimization in the context of neuropathic pain have not been reported. We investigated the pharmacokinetics and pharmacodynamics of KM after oral administration in a streptozotocin-induced rat model of diabetic neuropathic pain (DNP) using a population approach. A first-order absorption and elimination pharmacokinetics model best described the plasma KM concentration. This pharmacokinetic model was then linked to a linear pharmacodynamic model with an effect compartment based on the measurement of the mechanical withdrawal threshold. KM was rapidly absorbed (time to maximum plasma concentration: 0.14–0.36 h) with similar values in both DNP and naïve rats, suggesting that DNP did not influence the KM absorption rate. However, the area under the curve (AUC(0–∞)) of KM in DNP rats was over 3-fold higher than that in naïve rats. The systemic clearance rate and volume of KM distribution were significantly lower in DNP rats than in naïve rats. Blood glucose value prior to KM treatment was a significant covariate for the systemic clearance rate of KM and baseline value of the threshold. Our results suggest that streptozotocin-induced hyperglycemia is an independent factor for decreased KM elimination and its anti-allodynic effects in a DNP rat model. To the best of our knowledge, this is the first study to investigate the role of DNP in the pharmacokinetics and pharmacokinetics-pharmacodynamics of KM in streptozotocin-induced diabetic rats. Frontiers Media S.A. 2021-05-13 /pmc/articles/PMC8156416/ /pubmed/34054521 http://dx.doi.org/10.3389/fphar.2021.640318 Text en Copyright © 2021 Ye, Huang, Zhang, Lu, Cao, Wu, Chi, Hong, Wu, Xu and Yu. https://creativecommons.org/licenses/by/4.0/This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.
spellingShingle Pharmacology
Ye, Li-Xiang
Huang, Hui-Hui
Zhang, Shui-Hua
Lu, Jing-Shan
Cao, Da-Xuan
Wu, Dan-Dan
Chi, Pei-Wang
Hong, Long-Hui
Wu, Min-Xia
Xu, Ying
Yu, Chang-Xi
Streptozotocin-Induced Hyperglycemia Affects the Pharmacokinetics of Koumine and its Anti-Allodynic Action in a Rat Model of Diabetic Neuropathic Pain
title Streptozotocin-Induced Hyperglycemia Affects the Pharmacokinetics of Koumine and its Anti-Allodynic Action in a Rat Model of Diabetic Neuropathic Pain
title_full Streptozotocin-Induced Hyperglycemia Affects the Pharmacokinetics of Koumine and its Anti-Allodynic Action in a Rat Model of Diabetic Neuropathic Pain
title_fullStr Streptozotocin-Induced Hyperglycemia Affects the Pharmacokinetics of Koumine and its Anti-Allodynic Action in a Rat Model of Diabetic Neuropathic Pain
title_full_unstemmed Streptozotocin-Induced Hyperglycemia Affects the Pharmacokinetics of Koumine and its Anti-Allodynic Action in a Rat Model of Diabetic Neuropathic Pain
title_short Streptozotocin-Induced Hyperglycemia Affects the Pharmacokinetics of Koumine and its Anti-Allodynic Action in a Rat Model of Diabetic Neuropathic Pain
title_sort streptozotocin-induced hyperglycemia affects the pharmacokinetics of koumine and its anti-allodynic action in a rat model of diabetic neuropathic pain
topic Pharmacology
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8156416/
https://www.ncbi.nlm.nih.gov/pubmed/34054521
http://dx.doi.org/10.3389/fphar.2021.640318
work_keys_str_mv AT yelixiang streptozotocininducedhyperglycemiaaffectsthepharmacokineticsofkoumineanditsantiallodynicactioninaratmodelofdiabeticneuropathicpain
AT huanghuihui streptozotocininducedhyperglycemiaaffectsthepharmacokineticsofkoumineanditsantiallodynicactioninaratmodelofdiabeticneuropathicpain
AT zhangshuihua streptozotocininducedhyperglycemiaaffectsthepharmacokineticsofkoumineanditsantiallodynicactioninaratmodelofdiabeticneuropathicpain
AT lujingshan streptozotocininducedhyperglycemiaaffectsthepharmacokineticsofkoumineanditsantiallodynicactioninaratmodelofdiabeticneuropathicpain
AT caodaxuan streptozotocininducedhyperglycemiaaffectsthepharmacokineticsofkoumineanditsantiallodynicactioninaratmodelofdiabeticneuropathicpain
AT wudandan streptozotocininducedhyperglycemiaaffectsthepharmacokineticsofkoumineanditsantiallodynicactioninaratmodelofdiabeticneuropathicpain
AT chipeiwang streptozotocininducedhyperglycemiaaffectsthepharmacokineticsofkoumineanditsantiallodynicactioninaratmodelofdiabeticneuropathicpain
AT honglonghui streptozotocininducedhyperglycemiaaffectsthepharmacokineticsofkoumineanditsantiallodynicactioninaratmodelofdiabeticneuropathicpain
AT wuminxia streptozotocininducedhyperglycemiaaffectsthepharmacokineticsofkoumineanditsantiallodynicactioninaratmodelofdiabeticneuropathicpain
AT xuying streptozotocininducedhyperglycemiaaffectsthepharmacokineticsofkoumineanditsantiallodynicactioninaratmodelofdiabeticneuropathicpain
AT yuchangxi streptozotocininducedhyperglycemiaaffectsthepharmacokineticsofkoumineanditsantiallodynicactioninaratmodelofdiabeticneuropathicpain