Cargando…

Tetramethylpyrazine reverses high-glucose induced hypoxic effects by negatively regulating HIF-1α induced BNIP3 expression to ameliorate H9c2 cardiomyoblast apoptosis

BACKGROUND: Diabetic patients are highly vulnerable to hypoxic injury, which is associated with hypoxia induced BNIP3 expression that subsequently activate apoptosis. Our previous research show that Tetramethylpyrazine (TMP), a food flavoring agent, represses the hypoxia induced BNIP3 expression att...

Descripción completa

Detalles Bibliográficos
Autores principales: Liu, Shih-Ping, Shibu, Marthandam Asokan, Tsai, Fuu-Jen, Hsu, Yuan-Man, Tsai, Chang-Hai, Chung, Jing-Gung, Yang, Jai-Sing, Tang, Chih-Hsin, Wang, Shulin, Li, Qiaowen, Huang, Chih-Yang
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2020
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6995207/
https://www.ncbi.nlm.nih.gov/pubmed/32021640
http://dx.doi.org/10.1186/s12986-020-0432-x
_version_ 1783493341310091264
author Liu, Shih-Ping
Shibu, Marthandam Asokan
Tsai, Fuu-Jen
Hsu, Yuan-Man
Tsai, Chang-Hai
Chung, Jing-Gung
Yang, Jai-Sing
Tang, Chih-Hsin
Wang, Shulin
Li, Qiaowen
Huang, Chih-Yang
author_facet Liu, Shih-Ping
Shibu, Marthandam Asokan
Tsai, Fuu-Jen
Hsu, Yuan-Man
Tsai, Chang-Hai
Chung, Jing-Gung
Yang, Jai-Sing
Tang, Chih-Hsin
Wang, Shulin
Li, Qiaowen
Huang, Chih-Yang
author_sort Liu, Shih-Ping
collection PubMed
description BACKGROUND: Diabetic patients are highly vulnerable to hypoxic injury, which is associated with hypoxia induced BNIP3 expression that subsequently activate apoptosis. Our previous research show that Tetramethylpyrazine (TMP), a food flavoring agent, represses the hypoxia induced BNIP3 expression attenuate myocardial apoptosis. In this study, we evaluate the effect of TMP to provide protection against hypoxia aggravated high-glucose associated cellular apoptosis. METHODS: The cytoprotective effect of TMP against high glucose induced cellular damages was determined on embryo derived H9c2 cardiomyoblast cells that were subjected to 5% hypoxia for 24 h and subjected to different duration of 33 mM high glucose challenge. Further, the involvement of HIF-1α and BNIP3 in cellular damage and the mechanism of protection of TMP were determined by overexpression and silencing HIF-1α and BNIP3 protein expression. RESULTS: The results show that hypoxic effects on cell viability aggravates with high glucose challenge and this augmentative effect is mediated through BNIP3 in H9c2 cardiomyoblast cells. However, TMP administration effectively reversed the augmented HIF-1α levels and BNIP3 elevation. TMP improved the survival of H9c2 cells and effectively suppressed apoptosis in H9c2 cells. Further comparison on the effects of TMP on H9c2 cells challenged with high glucose and those challenged with hypoxia show that TMP precisely regulated the hypoxic intensified apoptotic effects in high-glucose condition. CONCLUSION: The results clearly show that flavoring agent-TMP attenuates cytotoxicity amplified by hypoxia challenge in high glucose condition by destabilizing HIF-1α.
format Online
Article
Text
id pubmed-6995207
institution National Center for Biotechnology Information
language English
publishDate 2020
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-69952072020-02-04 Tetramethylpyrazine reverses high-glucose induced hypoxic effects by negatively regulating HIF-1α induced BNIP3 expression to ameliorate H9c2 cardiomyoblast apoptosis Liu, Shih-Ping Shibu, Marthandam Asokan Tsai, Fuu-Jen Hsu, Yuan-Man Tsai, Chang-Hai Chung, Jing-Gung Yang, Jai-Sing Tang, Chih-Hsin Wang, Shulin Li, Qiaowen Huang, Chih-Yang Nutr Metab (Lond) Research BACKGROUND: Diabetic patients are highly vulnerable to hypoxic injury, which is associated with hypoxia induced BNIP3 expression that subsequently activate apoptosis. Our previous research show that Tetramethylpyrazine (TMP), a food flavoring agent, represses the hypoxia induced BNIP3 expression attenuate myocardial apoptosis. In this study, we evaluate the effect of TMP to provide protection against hypoxia aggravated high-glucose associated cellular apoptosis. METHODS: The cytoprotective effect of TMP against high glucose induced cellular damages was determined on embryo derived H9c2 cardiomyoblast cells that were subjected to 5% hypoxia for 24 h and subjected to different duration of 33 mM high glucose challenge. Further, the involvement of HIF-1α and BNIP3 in cellular damage and the mechanism of protection of TMP were determined by overexpression and silencing HIF-1α and BNIP3 protein expression. RESULTS: The results show that hypoxic effects on cell viability aggravates with high glucose challenge and this augmentative effect is mediated through BNIP3 in H9c2 cardiomyoblast cells. However, TMP administration effectively reversed the augmented HIF-1α levels and BNIP3 elevation. TMP improved the survival of H9c2 cells and effectively suppressed apoptosis in H9c2 cells. Further comparison on the effects of TMP on H9c2 cells challenged with high glucose and those challenged with hypoxia show that TMP precisely regulated the hypoxic intensified apoptotic effects in high-glucose condition. CONCLUSION: The results clearly show that flavoring agent-TMP attenuates cytotoxicity amplified by hypoxia challenge in high glucose condition by destabilizing HIF-1α. BioMed Central 2020-01-31 /pmc/articles/PMC6995207/ /pubmed/32021640 http://dx.doi.org/10.1186/s12986-020-0432-x Text en © The Author(s). 2020 Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.
spellingShingle Research
Liu, Shih-Ping
Shibu, Marthandam Asokan
Tsai, Fuu-Jen
Hsu, Yuan-Man
Tsai, Chang-Hai
Chung, Jing-Gung
Yang, Jai-Sing
Tang, Chih-Hsin
Wang, Shulin
Li, Qiaowen
Huang, Chih-Yang
Tetramethylpyrazine reverses high-glucose induced hypoxic effects by negatively regulating HIF-1α induced BNIP3 expression to ameliorate H9c2 cardiomyoblast apoptosis
title Tetramethylpyrazine reverses high-glucose induced hypoxic effects by negatively regulating HIF-1α induced BNIP3 expression to ameliorate H9c2 cardiomyoblast apoptosis
title_full Tetramethylpyrazine reverses high-glucose induced hypoxic effects by negatively regulating HIF-1α induced BNIP3 expression to ameliorate H9c2 cardiomyoblast apoptosis
title_fullStr Tetramethylpyrazine reverses high-glucose induced hypoxic effects by negatively regulating HIF-1α induced BNIP3 expression to ameliorate H9c2 cardiomyoblast apoptosis
title_full_unstemmed Tetramethylpyrazine reverses high-glucose induced hypoxic effects by negatively regulating HIF-1α induced BNIP3 expression to ameliorate H9c2 cardiomyoblast apoptosis
title_short Tetramethylpyrazine reverses high-glucose induced hypoxic effects by negatively regulating HIF-1α induced BNIP3 expression to ameliorate H9c2 cardiomyoblast apoptosis
title_sort tetramethylpyrazine reverses high-glucose induced hypoxic effects by negatively regulating hif-1α induced bnip3 expression to ameliorate h9c2 cardiomyoblast apoptosis
topic Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6995207/
https://www.ncbi.nlm.nih.gov/pubmed/32021640
http://dx.doi.org/10.1186/s12986-020-0432-x
work_keys_str_mv AT liushihping tetramethylpyrazinereverseshighglucoseinducedhypoxiceffectsbynegativelyregulatinghif1ainducedbnip3expressiontoameliorateh9c2cardiomyoblastapoptosis
AT shibumarthandamasokan tetramethylpyrazinereverseshighglucoseinducedhypoxiceffectsbynegativelyregulatinghif1ainducedbnip3expressiontoameliorateh9c2cardiomyoblastapoptosis
AT tsaifuujen tetramethylpyrazinereverseshighglucoseinducedhypoxiceffectsbynegativelyregulatinghif1ainducedbnip3expressiontoameliorateh9c2cardiomyoblastapoptosis
AT hsuyuanman tetramethylpyrazinereverseshighglucoseinducedhypoxiceffectsbynegativelyregulatinghif1ainducedbnip3expressiontoameliorateh9c2cardiomyoblastapoptosis
AT tsaichanghai tetramethylpyrazinereverseshighglucoseinducedhypoxiceffectsbynegativelyregulatinghif1ainducedbnip3expressiontoameliorateh9c2cardiomyoblastapoptosis
AT chungjinggung tetramethylpyrazinereverseshighglucoseinducedhypoxiceffectsbynegativelyregulatinghif1ainducedbnip3expressiontoameliorateh9c2cardiomyoblastapoptosis
AT yangjaising tetramethylpyrazinereverseshighglucoseinducedhypoxiceffectsbynegativelyregulatinghif1ainducedbnip3expressiontoameliorateh9c2cardiomyoblastapoptosis
AT tangchihhsin tetramethylpyrazinereverseshighglucoseinducedhypoxiceffectsbynegativelyregulatinghif1ainducedbnip3expressiontoameliorateh9c2cardiomyoblastapoptosis
AT wangshulin tetramethylpyrazinereverseshighglucoseinducedhypoxiceffectsbynegativelyregulatinghif1ainducedbnip3expressiontoameliorateh9c2cardiomyoblastapoptosis
AT liqiaowen tetramethylpyrazinereverseshighglucoseinducedhypoxiceffectsbynegativelyregulatinghif1ainducedbnip3expressiontoameliorateh9c2cardiomyoblastapoptosis
AT huangchihyang tetramethylpyrazinereverseshighglucoseinducedhypoxiceffectsbynegativelyregulatinghif1ainducedbnip3expressiontoameliorateh9c2cardiomyoblastapoptosis