Counteracting Colon Cancer by Inhibiting Mitochondrial Respiration and Glycolysis with a Selective PKCδ Activator

Metabolic reprogramming is a central hub in tumor development and progression. Therefore, several efforts have been developed to find improved therapeutic approaches targeting cancer cell metabolism. Recently, we identified the 7 -acetoxy-6 -benzoyloxy-12- -benzoylroyleanone (Roy-Bz) as a PKCδ-selec...

Full description

Saved in:
Bibliographic Details
Published inInternational journal of molecular sciences Vol. 24; no. 6; p. 5710
Main Authors Bessa, Cláudia, Loureiro, Joana B, Barros, Matilde, Isca, Vera M S, Sardão, Vilma A, Oliveira, Paulo J, Bernardino, Raquel L, Herman-de-Sousa, Carina, Costa, Maria Adelina, Correia-de-Sá, Paulo, Alves, Marco G, Rijo, Patrícia, Saraiva, Lucília
Format Journal Article
LanguageEnglish
Published Switzerland MDPI AG 01.03.2023
MDPI
Subjects
Online AccessGet full text

Cover

Loading…
More Information
Summary:Metabolic reprogramming is a central hub in tumor development and progression. Therefore, several efforts have been developed to find improved therapeutic approaches targeting cancer cell metabolism. Recently, we identified the 7 -acetoxy-6 -benzoyloxy-12- -benzoylroyleanone (Roy-Bz) as a PKCδ-selective activator with potent anti-proliferative activity in colon cancer by stimulating a PKCδ-dependent mitochondrial apoptotic pathway. Herein, we investigated whether the antitumor activity of Roy-Bz, in colon cancer, could be related to glucose metabolism interference. The results showed that Roy-Bz decreased the mitochondrial respiration in human colon HCT116 cancer cells, by reducing electron transfer chain complexes I/III. Consistently, this effect was associated with downregulation of the mitochondrial markers cytochrome oxidase subunit 4 (COX4), voltage-dependent anion channel (VDAC) and mitochondrial import receptor subunit TOM20 homolog (TOM20), and upregulation of synthesis of cytochrome oxidase 2 (SCO2). Roy-Bz also dropped glycolysis, decreasing the expression of critical glycolytic markers directly implicated in glucose metabolism such as glucose transporter 1 (GLUT1), hexokinase 2 (HK2) and monocarboxylate transporter 4 (MCT4), and increasing -induced glycolysis and apoptosis regulator (TIGAR) protein levels. These results were further corroborated in tumor xenografts of colon cancer. Altogether, using a PKCδ-selective activator, this work evidenced a potential dual role of PKCδ in tumor cell metabolism, resulting from the inhibition of both mitochondrial respiration and glycolysis. Additionally, it reinforces the antitumor therapeutic potential of Roy-Bz in colon cancer by targeting glucose metabolism.
Bibliography:ObjectType-Article-1
SourceType-Scholarly Journals-1
ObjectType-Feature-2
content type line 23
ISSN:1422-0067
1661-6596
1422-0067
DOI:10.3390/ijms24065710