Mitochondrial VDAC1 Silencing Leads to Metabolic Rewiring and the Reprogramming of Tumour Cells into Advanced Differentiated States

Oncogenic properties, along with the metabolic reprogramming necessary for tumour growth and motility, are acquired by cancer cells. Thus, tumour metabolism is becoming a target for cancer therapy. Here, cancer cell metabolism was tackled by silencing the expression of voltage-dependent anion channe...

Full description

Bibliographic Details
Main Authors: Tasleem Arif, Avijit Paul, Yakov Krelin, Anna Shteinfer-Kuzmine, Varda Shoshan-Barmatz
Format: Article
Language:English
Published: MDPI AG 2018-12-01
Series:Cancers
Subjects:
Online Access:https://www.mdpi.com/2072-6694/10/12/499
id doaj-7dcf4c6d6f604153a14dc52a232658c3
record_format Article
spelling doaj-7dcf4c6d6f604153a14dc52a232658c32020-11-25T01:28:28ZengMDPI AGCancers2072-66942018-12-01101249910.3390/cancers10120499cancers10120499Mitochondrial VDAC1 Silencing Leads to Metabolic Rewiring and the Reprogramming of Tumour Cells into Advanced Differentiated StatesTasleem Arif0Avijit Paul1Yakov Krelin2Anna Shteinfer-Kuzmine3Varda Shoshan-Barmatz4Department of Life Sciences, The National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva 84105, IsraelDepartment of Life Sciences, The National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva 84105, IsraelDepartment of Life Sciences, The National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva 84105, IsraelDepartment of Life Sciences, The National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva 84105, IsraelDepartment of Life Sciences, The National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva 84105, IsraelOncogenic properties, along with the metabolic reprogramming necessary for tumour growth and motility, are acquired by cancer cells. Thus, tumour metabolism is becoming a target for cancer therapy. Here, cancer cell metabolism was tackled by silencing the expression of voltage-dependent anion channel 1 (VDAC1), a mitochondrial protein that controls cell energy, as well as metabolic and survival pathways and that is often over-expressed in many cancers. We demonstrated that silencing VDAC1 expression using human-specific siRNA (si-hVDAC1) inhibited cancer cell growth, both in vitro and in mouse xenograft models of human glioblastoma (U-87MG), lung cancer (A549), and triple negative breast cancer (MDA-MB-231). Importantly, treatment with si-hVDAC1 induced metabolic rewiring of the cancer cells, reversing their oncogenic properties and diverting them towards differentiated-like cells. The si-hVDAC1-treated residual “tumour„ showed reprogrammed metabolism, decreased proliferation, inhibited stemness and altered expression of genes and proteins, leading to cell differentiation toward less malignant lineages. These VDAC1 depletion-mediated effects involved alterations in master transcription factors associated with cancer hallmarks, such as highly increased expression of p53 and decreased expression of HIF-1a and c-Myc that regulate signalling pathways (e.g., AMPK, mTOR). High expression of p53 and the pro-apoptotic proteins cytochrome c and caspases without induction of apoptosis points to functions for these proteins in promoting cell differentiation. These results clearly show that VDAC1 depletion similarly leads to a rewiring of cancer cell metabolism in breast and lung cancer and glioblastoma, regardless of origin or mutational status. This metabolic reprogramming results in cell growth arrest and inhibited tumour growth while encouraging cell differentiation, thus generating cells with decreased proliferation capacity. These results further suggest VDAC1 to be an innovative and markedly potent therapeutic target.https://www.mdpi.com/2072-6694/10/12/499cancer stem cellsdifferentiationmitochondriasi-RNAVDAC1
collection DOAJ
language English
format Article
sources DOAJ
author Tasleem Arif
Avijit Paul
Yakov Krelin
Anna Shteinfer-Kuzmine
Varda Shoshan-Barmatz
spellingShingle Tasleem Arif
Avijit Paul
Yakov Krelin
Anna Shteinfer-Kuzmine
Varda Shoshan-Barmatz
Mitochondrial VDAC1 Silencing Leads to Metabolic Rewiring and the Reprogramming of Tumour Cells into Advanced Differentiated States
Cancers
cancer stem cells
differentiation
mitochondria
si-RNA
VDAC1
author_facet Tasleem Arif
Avijit Paul
Yakov Krelin
Anna Shteinfer-Kuzmine
Varda Shoshan-Barmatz
author_sort Tasleem Arif
title Mitochondrial VDAC1 Silencing Leads to Metabolic Rewiring and the Reprogramming of Tumour Cells into Advanced Differentiated States
title_short Mitochondrial VDAC1 Silencing Leads to Metabolic Rewiring and the Reprogramming of Tumour Cells into Advanced Differentiated States
title_full Mitochondrial VDAC1 Silencing Leads to Metabolic Rewiring and the Reprogramming of Tumour Cells into Advanced Differentiated States
title_fullStr Mitochondrial VDAC1 Silencing Leads to Metabolic Rewiring and the Reprogramming of Tumour Cells into Advanced Differentiated States
title_full_unstemmed Mitochondrial VDAC1 Silencing Leads to Metabolic Rewiring and the Reprogramming of Tumour Cells into Advanced Differentiated States
title_sort mitochondrial vdac1 silencing leads to metabolic rewiring and the reprogramming of tumour cells into advanced differentiated states
publisher MDPI AG
series Cancers
issn 2072-6694
publishDate 2018-12-01
description Oncogenic properties, along with the metabolic reprogramming necessary for tumour growth and motility, are acquired by cancer cells. Thus, tumour metabolism is becoming a target for cancer therapy. Here, cancer cell metabolism was tackled by silencing the expression of voltage-dependent anion channel 1 (VDAC1), a mitochondrial protein that controls cell energy, as well as metabolic and survival pathways and that is often over-expressed in many cancers. We demonstrated that silencing VDAC1 expression using human-specific siRNA (si-hVDAC1) inhibited cancer cell growth, both in vitro and in mouse xenograft models of human glioblastoma (U-87MG), lung cancer (A549), and triple negative breast cancer (MDA-MB-231). Importantly, treatment with si-hVDAC1 induced metabolic rewiring of the cancer cells, reversing their oncogenic properties and diverting them towards differentiated-like cells. The si-hVDAC1-treated residual “tumour„ showed reprogrammed metabolism, decreased proliferation, inhibited stemness and altered expression of genes and proteins, leading to cell differentiation toward less malignant lineages. These VDAC1 depletion-mediated effects involved alterations in master transcription factors associated with cancer hallmarks, such as highly increased expression of p53 and decreased expression of HIF-1a and c-Myc that regulate signalling pathways (e.g., AMPK, mTOR). High expression of p53 and the pro-apoptotic proteins cytochrome c and caspases without induction of apoptosis points to functions for these proteins in promoting cell differentiation. These results clearly show that VDAC1 depletion similarly leads to a rewiring of cancer cell metabolism in breast and lung cancer and glioblastoma, regardless of origin or mutational status. This metabolic reprogramming results in cell growth arrest and inhibited tumour growth while encouraging cell differentiation, thus generating cells with decreased proliferation capacity. These results further suggest VDAC1 to be an innovative and markedly potent therapeutic target.
topic cancer stem cells
differentiation
mitochondria
si-RNA
VDAC1
url https://www.mdpi.com/2072-6694/10/12/499
work_keys_str_mv AT tasleemarif mitochondrialvdac1silencingleadstometabolicrewiringandthereprogrammingoftumourcellsintoadvanceddifferentiatedstates
AT avijitpaul mitochondrialvdac1silencingleadstometabolicrewiringandthereprogrammingoftumourcellsintoadvanceddifferentiatedstates
AT yakovkrelin mitochondrialvdac1silencingleadstometabolicrewiringandthereprogrammingoftumourcellsintoadvanceddifferentiatedstates
AT annashteinferkuzmine mitochondrialvdac1silencingleadstometabolicrewiringandthereprogrammingoftumourcellsintoadvanceddifferentiatedstates
AT vardashoshanbarmatz mitochondrialvdac1silencingleadstometabolicrewiringandthereprogrammingoftumourcellsintoadvanceddifferentiatedstates
_version_ 1725101448057847808