Cargando…

HOXC13-driven TIMM13 overexpression promotes osteosarcoma cell growth

TIMM13 (translocase of inner mitochondrial membrane 13) located at the mitochondrial intermembrane space is vital for the integrity and function of mitochondria. We found that the mitochondrial protein TIMM13 is upregulated in human OS tissues and cells. In patient-derived primary OS cells and estab...

Descripción completa

Detalles Bibliográficos
Autores principales: Han, Qicai, Yan, Penghui, Song, Ruipeng, Liu, Feifei, Tian, Qing
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Nature Publishing Group UK 2023
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10322838/
https://www.ncbi.nlm.nih.gov/pubmed/37407582
http://dx.doi.org/10.1038/s41419-023-05910-0
_version_ 1785068845194543104
author Han, Qicai
Yan, Penghui
Song, Ruipeng
Liu, Feifei
Tian, Qing
author_facet Han, Qicai
Yan, Penghui
Song, Ruipeng
Liu, Feifei
Tian, Qing
author_sort Han, Qicai
collection PubMed
description TIMM13 (translocase of inner mitochondrial membrane 13) located at the mitochondrial intermembrane space is vital for the integrity and function of mitochondria. We found that the mitochondrial protein TIMM13 is upregulated in human OS tissues and cells. In patient-derived primary OS cells and established cell lines, TIMM13 shRNA or knockout provoked mitochondrial dysfunction, causing mitochondrial depolarization, reactive oxygen species production, and oxidative injury, as well as lipid peroxidation, DNA damage, and ATP depletion. Moreover, TIMM13 depletion provoked OS cell apoptosis and inhibited cell proliferation and migration. Conversely, ectopic TIMM13 overexpression increased ATP contents, enhancing OS cell proliferation and migration. Moreover, we discovered that Akt-mTOR activation was inhibited with TIMM13 depletion in primary OS cells. Further studies revealed that HOXC13 (Homeobox C13)-dependent TIMM13 transcription was significantly increased in OS tissues and cells. Whereas TIMM13 transcription and expression were decreased following HOXC13 silencing in primary OS cells. In vivo, TIMM13 KO potently inhibited OS xenograft growth in the proximal tibia of nude mice. TIMM13 KO also induced Akt-mTOR inactivation, ATP depletion, oxidative injury, and apoptosis in the in situ OS tumors. Together, upregulation of the mitochondrial protein TIMM13 is important for OS cell growth, representing a novel and promising therapeutic target.
format Online
Article
Text
id pubmed-10322838
institution National Center for Biotechnology Information
language English
publishDate 2023
publisher Nature Publishing Group UK
record_format MEDLINE/PubMed
spelling pubmed-103228382023-07-07 HOXC13-driven TIMM13 overexpression promotes osteosarcoma cell growth Han, Qicai Yan, Penghui Song, Ruipeng Liu, Feifei Tian, Qing Cell Death Dis Article TIMM13 (translocase of inner mitochondrial membrane 13) located at the mitochondrial intermembrane space is vital for the integrity and function of mitochondria. We found that the mitochondrial protein TIMM13 is upregulated in human OS tissues and cells. In patient-derived primary OS cells and established cell lines, TIMM13 shRNA or knockout provoked mitochondrial dysfunction, causing mitochondrial depolarization, reactive oxygen species production, and oxidative injury, as well as lipid peroxidation, DNA damage, and ATP depletion. Moreover, TIMM13 depletion provoked OS cell apoptosis and inhibited cell proliferation and migration. Conversely, ectopic TIMM13 overexpression increased ATP contents, enhancing OS cell proliferation and migration. Moreover, we discovered that Akt-mTOR activation was inhibited with TIMM13 depletion in primary OS cells. Further studies revealed that HOXC13 (Homeobox C13)-dependent TIMM13 transcription was significantly increased in OS tissues and cells. Whereas TIMM13 transcription and expression were decreased following HOXC13 silencing in primary OS cells. In vivo, TIMM13 KO potently inhibited OS xenograft growth in the proximal tibia of nude mice. TIMM13 KO also induced Akt-mTOR inactivation, ATP depletion, oxidative injury, and apoptosis in the in situ OS tumors. Together, upregulation of the mitochondrial protein TIMM13 is important for OS cell growth, representing a novel and promising therapeutic target. Nature Publishing Group UK 2023-07-05 /pmc/articles/PMC10322838/ /pubmed/37407582 http://dx.doi.org/10.1038/s41419-023-05910-0 Text en © The Author(s) 2023 https://creativecommons.org/licenses/by/4.0/Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as 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 images or other third party material in this article are included in the article’s Creative Commons license, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons license and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this license, visit http://creativecommons.org/licenses/by/4.0/ (https://creativecommons.org/licenses/by/4.0/) .
spellingShingle Article
Han, Qicai
Yan, Penghui
Song, Ruipeng
Liu, Feifei
Tian, Qing
HOXC13-driven TIMM13 overexpression promotes osteosarcoma cell growth
title HOXC13-driven TIMM13 overexpression promotes osteosarcoma cell growth
title_full HOXC13-driven TIMM13 overexpression promotes osteosarcoma cell growth
title_fullStr HOXC13-driven TIMM13 overexpression promotes osteosarcoma cell growth
title_full_unstemmed HOXC13-driven TIMM13 overexpression promotes osteosarcoma cell growth
title_short HOXC13-driven TIMM13 overexpression promotes osteosarcoma cell growth
title_sort hoxc13-driven timm13 overexpression promotes osteosarcoma cell growth
topic Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10322838/
https://www.ncbi.nlm.nih.gov/pubmed/37407582
http://dx.doi.org/10.1038/s41419-023-05910-0
work_keys_str_mv AT hanqicai hoxc13driventimm13overexpressionpromotesosteosarcomacellgrowth
AT yanpenghui hoxc13driventimm13overexpressionpromotesosteosarcomacellgrowth
AT songruipeng hoxc13driventimm13overexpressionpromotesosteosarcomacellgrowth
AT liufeifei hoxc13driventimm13overexpressionpromotesosteosarcomacellgrowth
AT tianqing hoxc13driventimm13overexpressionpromotesosteosarcomacellgrowth