Nano-flow cytometry unveils mitochondrial permeability transition process and multi-pathway cell death induction for cancer therapy

Abstract Mitochondrial permeability transition (mPT)-mediated mitochondrial dysfunction plays a pivotal role in various human diseases. However, the intricate details of its mechanisms and the sequence of events remain elusive, primarily due to the interference caused by Bax/Bak-induced mitochondria...

Full description

Bibliographic Details
Main Authors: Liyun Su, Jingyi Xu, Cheng Lu, Kaimin Gao, Yunyun Hu, Chengfeng Xue, Xiaomei Yan
Format: Article
Language:English
Published: Nature Publishing Group 2024-04-01
Series:Cell Death Discovery
Online Access:https://doi.org/10.1038/s41420-024-01947-y
_version_ 1797199657746563072
author Liyun Su
Jingyi Xu
Cheng Lu
Kaimin Gao
Yunyun Hu
Chengfeng Xue
Xiaomei Yan
author_facet Liyun Su
Jingyi Xu
Cheng Lu
Kaimin Gao
Yunyun Hu
Chengfeng Xue
Xiaomei Yan
author_sort Liyun Su
collection DOAJ
description Abstract Mitochondrial permeability transition (mPT)-mediated mitochondrial dysfunction plays a pivotal role in various human diseases. However, the intricate details of its mechanisms and the sequence of events remain elusive, primarily due to the interference caused by Bax/Bak-induced mitochondrial outer membrane permeabilization (MOMP). To address these, we have developed a methodology that utilizes nano-flow cytometry (nFCM) to quantitatively analyze the opening of mitochondrial permeability transition pore (mPTP), dissipation of mitochondrial membrane potential ( $$\Delta$$ Δ Ψm), release of cytochrome c (Cyt c), and other molecular alternations of isolated mitochondria in response to mPT induction at the single-mitochondrion level. It was identified that betulinic acid (BetA) and antimycin A can directly induce mitochondrial dysfunction through mPT-mediated mechanisms, while cisplatin and staurosporine cannot. In addition, the nFCM analysis also revealed that BetA primarily induces mPTP opening through a reduction in Bcl-2 and Bcl-xL protein levels, along with an elevation in ROS content. Employing dose and time-dependent strategies of BetA, for the first time, we experimentally verified the sequential occurrence of mPTP opening and $$\Delta$$ Δ Ψm depolarization prior to the release of Cyt c during mPT-mediated mitochondrial dysfunction. Notably, our study uncovers a simultaneous release of cell-death-associated factors, including Cyt c, AIF, PNPT1, and mtDNA during mPT, implying the initiation of multiple cell death pathways. Intriguingly, BetA induces caspase-independent cell death, even in the absence of Bax/Bak, thereby overcoming drug resistance. The presented findings offer new insights into mPT-mediated mitochondrial dysfunction using nFCM, emphasizing the potential for targeting such dysfunction in innovative cancer therapies and interventions.
first_indexed 2024-04-24T07:19:15Z
format Article
id doaj.art-1fa458a1dfd34cd9af37cb77e0393991
institution Directory Open Access Journal
issn 2058-7716
language English
last_indexed 2024-04-24T07:19:15Z
publishDate 2024-04-01
publisher Nature Publishing Group
record_format Article
series Cell Death Discovery
spelling doaj.art-1fa458a1dfd34cd9af37cb77e03939912024-04-21T11:08:38ZengNature Publishing GroupCell Death Discovery2058-77162024-04-0110111010.1038/s41420-024-01947-yNano-flow cytometry unveils mitochondrial permeability transition process and multi-pathway cell death induction for cancer therapyLiyun Su0Jingyi Xu1Cheng Lu2Kaimin Gao3Yunyun Hu4Chengfeng Xue5Xiaomei Yan6Department of Chemical Biology, MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, Key Laboratory for Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Collaborative Innovation Center of Chemistry for Energy Materials, College of Chemistry and Chemical Engineering, Xiamen UniversityDepartment of Chemical Biology, MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, Key Laboratory for Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Collaborative Innovation Center of Chemistry for Energy Materials, College of Chemistry and Chemical Engineering, Xiamen UniversityDepartment of Chemical Biology, MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, Key Laboratory for Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Collaborative Innovation Center of Chemistry for Energy Materials, College of Chemistry and Chemical Engineering, Xiamen UniversityDepartment of Chemical Biology, MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, Key Laboratory for Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Collaborative Innovation Center of Chemistry for Energy Materials, College of Chemistry and Chemical Engineering, Xiamen UniversityDepartment of Chemical Biology, MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, Key Laboratory for Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Collaborative Innovation Center of Chemistry for Energy Materials, College of Chemistry and Chemical Engineering, Xiamen UniversityDepartment of Chemical Biology, MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, Key Laboratory for Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Collaborative Innovation Center of Chemistry for Energy Materials, College of Chemistry and Chemical Engineering, Xiamen UniversityDepartment of Chemical Biology, MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, Key Laboratory for Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Collaborative Innovation Center of Chemistry for Energy Materials, College of Chemistry and Chemical Engineering, Xiamen UniversityAbstract Mitochondrial permeability transition (mPT)-mediated mitochondrial dysfunction plays a pivotal role in various human diseases. However, the intricate details of its mechanisms and the sequence of events remain elusive, primarily due to the interference caused by Bax/Bak-induced mitochondrial outer membrane permeabilization (MOMP). To address these, we have developed a methodology that utilizes nano-flow cytometry (nFCM) to quantitatively analyze the opening of mitochondrial permeability transition pore (mPTP), dissipation of mitochondrial membrane potential ( $$\Delta$$ Δ Ψm), release of cytochrome c (Cyt c), and other molecular alternations of isolated mitochondria in response to mPT induction at the single-mitochondrion level. It was identified that betulinic acid (BetA) and antimycin A can directly induce mitochondrial dysfunction through mPT-mediated mechanisms, while cisplatin and staurosporine cannot. In addition, the nFCM analysis also revealed that BetA primarily induces mPTP opening through a reduction in Bcl-2 and Bcl-xL protein levels, along with an elevation in ROS content. Employing dose and time-dependent strategies of BetA, for the first time, we experimentally verified the sequential occurrence of mPTP opening and $$\Delta$$ Δ Ψm depolarization prior to the release of Cyt c during mPT-mediated mitochondrial dysfunction. Notably, our study uncovers a simultaneous release of cell-death-associated factors, including Cyt c, AIF, PNPT1, and mtDNA during mPT, implying the initiation of multiple cell death pathways. Intriguingly, BetA induces caspase-independent cell death, even in the absence of Bax/Bak, thereby overcoming drug resistance. The presented findings offer new insights into mPT-mediated mitochondrial dysfunction using nFCM, emphasizing the potential for targeting such dysfunction in innovative cancer therapies and interventions.https://doi.org/10.1038/s41420-024-01947-y
spellingShingle Liyun Su
Jingyi Xu
Cheng Lu
Kaimin Gao
Yunyun Hu
Chengfeng Xue
Xiaomei Yan
Nano-flow cytometry unveils mitochondrial permeability transition process and multi-pathway cell death induction for cancer therapy
Cell Death Discovery
title Nano-flow cytometry unveils mitochondrial permeability transition process and multi-pathway cell death induction for cancer therapy
title_full Nano-flow cytometry unveils mitochondrial permeability transition process and multi-pathway cell death induction for cancer therapy
title_fullStr Nano-flow cytometry unveils mitochondrial permeability transition process and multi-pathway cell death induction for cancer therapy
title_full_unstemmed Nano-flow cytometry unveils mitochondrial permeability transition process and multi-pathway cell death induction for cancer therapy
title_short Nano-flow cytometry unveils mitochondrial permeability transition process and multi-pathway cell death induction for cancer therapy
title_sort nano flow cytometry unveils mitochondrial permeability transition process and multi pathway cell death induction for cancer therapy
url https://doi.org/10.1038/s41420-024-01947-y
work_keys_str_mv AT liyunsu nanoflowcytometryunveilsmitochondrialpermeabilitytransitionprocessandmultipathwaycelldeathinductionforcancertherapy
AT jingyixu nanoflowcytometryunveilsmitochondrialpermeabilitytransitionprocessandmultipathwaycelldeathinductionforcancertherapy
AT chenglu nanoflowcytometryunveilsmitochondrialpermeabilitytransitionprocessandmultipathwaycelldeathinductionforcancertherapy
AT kaimingao nanoflowcytometryunveilsmitochondrialpermeabilitytransitionprocessandmultipathwaycelldeathinductionforcancertherapy
AT yunyunhu nanoflowcytometryunveilsmitochondrialpermeabilitytransitionprocessandmultipathwaycelldeathinductionforcancertherapy
AT chengfengxue nanoflowcytometryunveilsmitochondrialpermeabilitytransitionprocessandmultipathwaycelldeathinductionforcancertherapy
AT xiaomeiyan nanoflowcytometryunveilsmitochondrialpermeabilitytransitionprocessandmultipathwaycelldeathinductionforcancertherapy