4.8 Article

Involvement of mitophagy in oncogenic K-Ras-induced transformation Overcoming a cellular energy deficit from glucose deficiency

Journal

AUTOPHAGY
Volume 7, Issue 10, Pages 1187-1198

Publisher

TAYLOR & FRANCIS INC
DOI: 10.4161/auto.7.10.16643

Keywords

cell transformation; energy deficit; K-Ras; mitophagy; mitochondrial loss

Categories

Funding

  1. Korean Science and Engineering Foundation (KOSEF)
  2. Korean government (MEST) [R13-2003-019-01007-0]
  3. NRF
  4. MEST [2009-0079076]
  5. Korea Research Foundation
  6. Korean Government [KRF-2008-314-C00286]
  7. National Research Foundation of Korea [2009-0079076, R13-2003-019-01007-0] Funding Source: Korea Institute of Science & Technology Information (KISTI), National Science & Technology Information Service (NTIS)

Ask authors/readers for more resources

Although mitochondrial impairment has often been implicated in carcinogenesis, the mechanisms of its development in cancer remain unknown. We report here that autophagy triggered by oncogenic K-Ras mediates functional loss of mitochondria during cell transformation to overcome an energy deficit resulting from glucose deficiency. When Rat2 cells were infected with a retrovirus harboring constitutively active K-Ras(V12), mitochondrial respiration significantly declined in parallel with the acquisition of transformation characteristics. Decreased respiration was not related to mitochondrial biogenesis but was inversely associated with the increased formation of acidic vesicles enclosing mitochondria, during which autophagy-related proteins such as Beclin 1, Atg5, LC3-II and vacuolar ATPases were induced. Interestingly, blocking autophagy with conventional inhibitors (bafilomycin A, 3-methyladenin) and siRNA-mediated knockdown of autophagy-related genes recovered respiratory protein expression and respiratory activity; JNK was involved in these phenomena as an upstream regulator. The cells transformed by K-Ras(V12) maintained cellular ATP level mainly through glycolytic ATP production without induction of GLUT1, the low K-m glucose transporter. Finally, K-Ras(V12)-triggered LC3-II formation was modulated by extracellular glucose levels, and LC3-II formation increased only in hepatocellular carcinoma tissues exhibiting low glucose uptake and increased K-Ras expression. Taken together, our observations suggest that mitochondrial functional loss may be mediated by oncogenic K-Ras-induced mitophagy during early tumorigenesis even in the absence of hypoxia, and that this mitophagic process may be an important strategy to overcome the cellular energy deficit triggered by insufficient glucose.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.8
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available