4.8 Article

LC3 subfamily in cardiolipin-mediated mitophagy: a comparison of the LC3A, LC3B and LC3C homologs

Journal

AUTOPHAGY
Volume 18, Issue 12, Pages 2985-3003

Publisher

TAYLOR & FRANCIS INC
DOI: 10.1080/15548627.2022.2062111

Keywords

Atg8; autophagosome; autophagy cargo recognition; LC3; GABARAP-protein family; lipid oxidation; lipid-protein interaction; membrane curvature; mitochondria; negatively charged phospholipids

Categories

Funding

  1. Spanish Ministerio de Ciencia e Innovacion (MCI)
  2. Agencia Estatal de Investigacion (AEI)
  3. Fondo Europeo de Desarrollo Regional (FEDER) [PGC2018-099857-B-I00]
  4. Basque Government [IT1625-22, IT1270-19]
  5. Fundacion Ramon Areces [CIVP20A6619]
  6. Fundacion Biofisica Bizkaia
  7. Basque Excellence Research Centre (BERC) program of the Basque Government
  8. Spanish Ministry of Science Innovation and Universities [FPU16/05873, FPU18/00799]
  9. University of the Basque Country
  10. Basque Government

Ask authors/readers for more resources

Externalization of cardiolipin acts as a trigger for mitophagy, and LC3A and LC3B bind to cardiolipin with different strengths. LC3A is involved in the recognition of damaged mitochondria, while LC3C does not participate in cargo recognition. LC3A may play a role in the recognition of oxidized cardiolipin as a counterbalance to excessive apoptosis activation.
Externalization of the phospholipid cardiolipin (CL) to the outer mitochondrial membrane has been proposed to act as a mitophagy trigger. CL would act as a signal for binding the LC3 macroautophagy/autophagy proteins. As yet, the behavior of the LC3-subfamily members has not been directly compared in a detailed way. In the present contribution, an analysis of LC3A, LC3B and LC3C interaction with CL-containing model membranes, and of their ability to translocate to mitochondria, is described. Binding of LC3A to CL was stronger than that of LC3B; both proteins showed a similar ability to colocalize with mitochondria upon induction of CL externalization in SH-SY5Y cells. Besides, the double silencing of LC3A and LC3B proteins was seen to decrease CCCP-induced mitophagy. Residues 14 and 18 located in the N-terminal region of LC3A were shown to be important for its recognition of damaged mitochondria during rotenone- or CCCP-induced mitophagy. Moreover, the in vitro results suggested a possible role of LC3A, but not of LC3B, in oxidized-CL recognition as a counterweight to excessive apoptosis activation. In the case of LC3C, even if this protein showed a stronger CL binding than LC3B or LC3A, the interaction was less specific, and colocalization of LC3C with mitochondria was not rotenone dependent. These results suggest that, at variance with LC3A, LC3C does not participate in cargo recognition during CL-mediated-mitophagy. The data support the notion that the various LC3-subfamily members might play different roles during autophagy initiation, identifying LC3A as a novel stakeholder in CL-mediated mitophagy.

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