Cargando…

Bufarenogin induces intrinsic apoptosis via Bax and ANT cooperation

Toads have high medicinal value and have been used for medicinal purposes since the Tang Dynasty period (7th‐10th Century AD). Bufarenogin, an active anti‐tumor constituent of toad venom, shows anti‐tumor activity. In this study, we investigated the inhibitory effects of bufarenogin on the growth an...

Descripción completa

Detalles Bibliográficos
Autores principales: Han, Qinrui, Zhang, Chun, Zhang, Yongbin, Li, Yuan, Wu, Liyi, Sun, Xuegang
Formato: Online Artículo Texto
Lenguaje:English
Publicado: John Wiley and Sons Inc. 2021
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7796793/
https://www.ncbi.nlm.nih.gov/pubmed/33421322
http://dx.doi.org/10.1002/prp2.694
_version_ 1783634755935272960
author Han, Qinrui
Zhang, Chun
Zhang, Yongbin
Li, Yuan
Wu, Liyi
Sun, Xuegang
author_facet Han, Qinrui
Zhang, Chun
Zhang, Yongbin
Li, Yuan
Wu, Liyi
Sun, Xuegang
author_sort Han, Qinrui
collection PubMed
description Toads have high medicinal value and have been used for medicinal purposes since the Tang Dynasty period (7th‐10th Century AD). Bufarenogin, an active anti‐tumor constituent of toad venom, shows anti‐tumor activity. In this study, we investigated the inhibitory effects of bufarenogin on the growth and metastasis of colorectal cancer (CRC), particularly its effects on mediating intrinsic signaling pathways that initiate apoptosis. An orthotopic CRC model was established in nude mice via surgical orthotopic implantation to investigate tumor growth. Immunohistochemistry, immunofluorescence, and Western blotting assays were performed to evaluate protein expression. The in vitro results revealed the anti‐proliferative effect of bufarenogin against CRC cells. Bufarenogin caused cell death via apoptosis, as revealed by Annexin V/7‐amino‐actinomycin D double staining, which was verified using a pan‐caspase inhibitor. Bufarenogin induced B‐cell lymphoma 2‐associated X protein (Bax)‐dependent intrinsic apoptosis, as demonstrated by mitochondrial translocation of Bax and cytoplasm release of HCT116 wild‐type cells and cytochrome C (soluble pro‐apoptotic factors). Additionally, we showed that adenine‐nucleotide translocator interacted with Bax. Bufarenogin induced intrinsic apoptosis through the cooperation of Bax and adenine‐nucleotide translocator and inhibited the metastasis and growth of orthotopical CRC cells.
format Online
Article
Text
id pubmed-7796793
institution National Center for Biotechnology Information
language English
publishDate 2021
publisher John Wiley and Sons Inc.
record_format MEDLINE/PubMed
spelling pubmed-77967932021-01-15 Bufarenogin induces intrinsic apoptosis via Bax and ANT cooperation Han, Qinrui Zhang, Chun Zhang, Yongbin Li, Yuan Wu, Liyi Sun, Xuegang Pharmacol Res Perspect Original Articles Toads have high medicinal value and have been used for medicinal purposes since the Tang Dynasty period (7th‐10th Century AD). Bufarenogin, an active anti‐tumor constituent of toad venom, shows anti‐tumor activity. In this study, we investigated the inhibitory effects of bufarenogin on the growth and metastasis of colorectal cancer (CRC), particularly its effects on mediating intrinsic signaling pathways that initiate apoptosis. An orthotopic CRC model was established in nude mice via surgical orthotopic implantation to investigate tumor growth. Immunohistochemistry, immunofluorescence, and Western blotting assays were performed to evaluate protein expression. The in vitro results revealed the anti‐proliferative effect of bufarenogin against CRC cells. Bufarenogin caused cell death via apoptosis, as revealed by Annexin V/7‐amino‐actinomycin D double staining, which was verified using a pan‐caspase inhibitor. Bufarenogin induced B‐cell lymphoma 2‐associated X protein (Bax)‐dependent intrinsic apoptosis, as demonstrated by mitochondrial translocation of Bax and cytoplasm release of HCT116 wild‐type cells and cytochrome C (soluble pro‐apoptotic factors). Additionally, we showed that adenine‐nucleotide translocator interacted with Bax. Bufarenogin induced intrinsic apoptosis through the cooperation of Bax and adenine‐nucleotide translocator and inhibited the metastasis and growth of orthotopical CRC cells. John Wiley and Sons Inc. 2021-01-09 /pmc/articles/PMC7796793/ /pubmed/33421322 http://dx.doi.org/10.1002/prp2.694 Text en © 2021 The Authors. Pharmacology Research & Perspectives published by John Wiley & Sons Ltd, British Pharmacological Society and American Society for Pharmacology and Experimental Therapeutics. This is an open access article under the terms of the http://creativecommons.org/licenses/by-nc-nd/4.0/ License, which permits use and distribution in any medium, provided the original work is properly cited, the use is non‐commercial and no modifications or adaptations are made.
spellingShingle Original Articles
Han, Qinrui
Zhang, Chun
Zhang, Yongbin
Li, Yuan
Wu, Liyi
Sun, Xuegang
Bufarenogin induces intrinsic apoptosis via Bax and ANT cooperation
title Bufarenogin induces intrinsic apoptosis via Bax and ANT cooperation
title_full Bufarenogin induces intrinsic apoptosis via Bax and ANT cooperation
title_fullStr Bufarenogin induces intrinsic apoptosis via Bax and ANT cooperation
title_full_unstemmed Bufarenogin induces intrinsic apoptosis via Bax and ANT cooperation
title_short Bufarenogin induces intrinsic apoptosis via Bax and ANT cooperation
title_sort bufarenogin induces intrinsic apoptosis via bax and ant cooperation
topic Original Articles
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7796793/
https://www.ncbi.nlm.nih.gov/pubmed/33421322
http://dx.doi.org/10.1002/prp2.694
work_keys_str_mv AT hanqinrui bufarenogininducesintrinsicapoptosisviabaxandantcooperation
AT zhangchun bufarenogininducesintrinsicapoptosisviabaxandantcooperation
AT zhangyongbin bufarenogininducesintrinsicapoptosisviabaxandantcooperation
AT liyuan bufarenogininducesintrinsicapoptosisviabaxandantcooperation
AT wuliyi bufarenogininducesintrinsicapoptosisviabaxandantcooperation
AT sunxuegang bufarenogininducesintrinsicapoptosisviabaxandantcooperation