Cargando…

Porric acid E, a natural compound from Rhytidhysteron sp. BZM-9, suppresses colorectal cancer growth via an autophagy-dependent pathway

Colorectal cancer (CRC) is one of the major killer diseases worldwide, and more effective therapeutic compounds for CRC treatment are urgently needed. Although bioactive natural products derived from endophytic fungi have been extensively employed as antibiotics and anticancer agents, little is know...

Descripción completa

Detalles Bibliográficos
Autores principales: Tang, Da, Zhang, Wei, Zou, Zhenxing, Wang, Yikun, Yan, Shichao, Zhang, Sha, Cai, Wenwu, Li, Daming, Li, Qiuguo, Li, Wenbo
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Ivyspring International Publisher 2022
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9723991/
https://www.ncbi.nlm.nih.gov/pubmed/36484011
http://dx.doi.org/10.7150/jca.77588
_version_ 1784844313537019904
author Tang, Da
Zhang, Wei
Zou, Zhenxing
Wang, Yikun
Yan, Shichao
Zhang, Sha
Cai, Wenwu
Li, Daming
Li, Qiuguo
Li, Wenbo
author_facet Tang, Da
Zhang, Wei
Zou, Zhenxing
Wang, Yikun
Yan, Shichao
Zhang, Sha
Cai, Wenwu
Li, Daming
Li, Qiuguo
Li, Wenbo
author_sort Tang, Da
collection PubMed
description Colorectal cancer (CRC) is one of the major killer diseases worldwide, and more effective therapeutic compounds for CRC treatment are urgently needed. Although bioactive natural products derived from endophytic fungi have been extensively employed as antibiotics and anticancer agents, little is known about the effect of Rhytidhysteron sp. BZM-9 (an endophytic fungus)-derived compounds on CRC. Herein, a natural molecule porric acid E was isolated from Rhytidhysteron sp. BZM-9. Alamar Blue cell viability assay, Western blotting, transmission electron microscopy, flow cytometry analysis, and fluorescence image examination were employed to evaluate the antitumor effects of porric acid E on CRC cell lines. To establish the xenograft tumor model, nude mice received subcutaneous implants consisting of CRC cells on their flanks. Then the mice were treated with porric acid E or vehicle to assess the tumor-killing effects. The results revealed that porric acid E exhibited cytotoxicity by inhibiting proliferation and promoting apoptosis in CRC cells in vitro. Additionally, compared with fluorouracil (5-FU), porric acid E exhibited a more potent inhibitory effect on CRC HT29 cells. Importantly, extensive autophagy induced by porric acid E was detected in CRC cells, whereas inhibition of autophagy could significantly ameliorate porric acid E-mediated cytotoxic effect on CRC cells. Moreover, porric acid E treatment could markedly suppress subcutaneous HT29 xenograft tumor growth in vivo. Bioinformatics prediction indicated that Beclin-1 might be the potential target of porric acid E. These findings might afford a useful and important method for the treatment of CRC through fungal endophyte-derived natural compounds.
format Online
Article
Text
id pubmed-9723991
institution National Center for Biotechnology Information
language English
publishDate 2022
publisher Ivyspring International Publisher
record_format MEDLINE/PubMed
spelling pubmed-97239912022-12-07 Porric acid E, a natural compound from Rhytidhysteron sp. BZM-9, suppresses colorectal cancer growth via an autophagy-dependent pathway Tang, Da Zhang, Wei Zou, Zhenxing Wang, Yikun Yan, Shichao Zhang, Sha Cai, Wenwu Li, Daming Li, Qiuguo Li, Wenbo J Cancer Research Paper Colorectal cancer (CRC) is one of the major killer diseases worldwide, and more effective therapeutic compounds for CRC treatment are urgently needed. Although bioactive natural products derived from endophytic fungi have been extensively employed as antibiotics and anticancer agents, little is known about the effect of Rhytidhysteron sp. BZM-9 (an endophytic fungus)-derived compounds on CRC. Herein, a natural molecule porric acid E was isolated from Rhytidhysteron sp. BZM-9. Alamar Blue cell viability assay, Western blotting, transmission electron microscopy, flow cytometry analysis, and fluorescence image examination were employed to evaluate the antitumor effects of porric acid E on CRC cell lines. To establish the xenograft tumor model, nude mice received subcutaneous implants consisting of CRC cells on their flanks. Then the mice were treated with porric acid E or vehicle to assess the tumor-killing effects. The results revealed that porric acid E exhibited cytotoxicity by inhibiting proliferation and promoting apoptosis in CRC cells in vitro. Additionally, compared with fluorouracil (5-FU), porric acid E exhibited a more potent inhibitory effect on CRC HT29 cells. Importantly, extensive autophagy induced by porric acid E was detected in CRC cells, whereas inhibition of autophagy could significantly ameliorate porric acid E-mediated cytotoxic effect on CRC cells. Moreover, porric acid E treatment could markedly suppress subcutaneous HT29 xenograft tumor growth in vivo. Bioinformatics prediction indicated that Beclin-1 might be the potential target of porric acid E. These findings might afford a useful and important method for the treatment of CRC through fungal endophyte-derived natural compounds. Ivyspring International Publisher 2022-10-31 /pmc/articles/PMC9723991/ /pubmed/36484011 http://dx.doi.org/10.7150/jca.77588 Text en © The author(s) https://creativecommons.org/licenses/by/4.0/This is an open access article distributed under the terms of the Creative Commons Attribution License (https://creativecommons.org/licenses/by/4.0/). See http://ivyspring.com/terms for full terms and conditions.
spellingShingle Research Paper
Tang, Da
Zhang, Wei
Zou, Zhenxing
Wang, Yikun
Yan, Shichao
Zhang, Sha
Cai, Wenwu
Li, Daming
Li, Qiuguo
Li, Wenbo
Porric acid E, a natural compound from Rhytidhysteron sp. BZM-9, suppresses colorectal cancer growth via an autophagy-dependent pathway
title Porric acid E, a natural compound from Rhytidhysteron sp. BZM-9, suppresses colorectal cancer growth via an autophagy-dependent pathway
title_full Porric acid E, a natural compound from Rhytidhysteron sp. BZM-9, suppresses colorectal cancer growth via an autophagy-dependent pathway
title_fullStr Porric acid E, a natural compound from Rhytidhysteron sp. BZM-9, suppresses colorectal cancer growth via an autophagy-dependent pathway
title_full_unstemmed Porric acid E, a natural compound from Rhytidhysteron sp. BZM-9, suppresses colorectal cancer growth via an autophagy-dependent pathway
title_short Porric acid E, a natural compound from Rhytidhysteron sp. BZM-9, suppresses colorectal cancer growth via an autophagy-dependent pathway
title_sort porric acid e, a natural compound from rhytidhysteron sp. bzm-9, suppresses colorectal cancer growth via an autophagy-dependent pathway
topic Research Paper
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9723991/
https://www.ncbi.nlm.nih.gov/pubmed/36484011
http://dx.doi.org/10.7150/jca.77588
work_keys_str_mv AT tangda porricacideanaturalcompoundfromrhytidhysteronspbzm9suppressescolorectalcancergrowthviaanautophagydependentpathway
AT zhangwei porricacideanaturalcompoundfromrhytidhysteronspbzm9suppressescolorectalcancergrowthviaanautophagydependentpathway
AT zouzhenxing porricacideanaturalcompoundfromrhytidhysteronspbzm9suppressescolorectalcancergrowthviaanautophagydependentpathway
AT wangyikun porricacideanaturalcompoundfromrhytidhysteronspbzm9suppressescolorectalcancergrowthviaanautophagydependentpathway
AT yanshichao porricacideanaturalcompoundfromrhytidhysteronspbzm9suppressescolorectalcancergrowthviaanautophagydependentpathway
AT zhangsha porricacideanaturalcompoundfromrhytidhysteronspbzm9suppressescolorectalcancergrowthviaanautophagydependentpathway
AT caiwenwu porricacideanaturalcompoundfromrhytidhysteronspbzm9suppressescolorectalcancergrowthviaanautophagydependentpathway
AT lidaming porricacideanaturalcompoundfromrhytidhysteronspbzm9suppressescolorectalcancergrowthviaanautophagydependentpathway
AT liqiuguo porricacideanaturalcompoundfromrhytidhysteronspbzm9suppressescolorectalcancergrowthviaanautophagydependentpathway
AT liwenbo porricacideanaturalcompoundfromrhytidhysteronspbzm9suppressescolorectalcancergrowthviaanautophagydependentpathway