Archives

  • 2018-07
  • 2018-10
  • 2018-11
  • 2019-04
  • 2019-05
  • 2019-06
  • 2019-07
  • 2019-08
  • 2019-09
  • 2019-10
  • 2019-11
  • 2019-12
  • 2020-01
  • 2020-02
  • 2020-03
  • 2020-04
  • 2020-05
  • 2020-06
  • 2020-07
  • 2020-08
  • 2020-09
  • 2020-10
  • 2020-11
  • 2020-12
  • 2021-01
  • 2021-02
  • 2021-03
  • 2021-04
  • 2021-05
  • 2021-06
  • 2021-07
  • 2021-08
  • 2021-09
  • 2021-10
  • 2021-11
  • 2021-12
  • 2022-01
  • 2022-02
  • 2022-03
  • 2022-04
  • 2022-05
  • 2022-06
  • 2022-07
  • 2022-08
  • 2022-09
  • 2022-10
  • 2022-11
  • 2022-12
  • 2023-01
  • 2023-02
  • 2023-03
  • 2023-04
  • 2023-05
  • 2023-06
  • 2023-07
  • 2023-08
  • 2023-09
  • 2023-10
  • 2023-11
  • 2023-12
  • 2024-01
  • 2024-02
  • 2024-03
  • Despite the weak and transient nature of E E

    2019-10-22

    Despite the weak and transient nature of E2–E3 interactions, this binding is required for the transfer of ubiquitin to the substrate. The low affinity characteristics of this interaction may be due to the fact that E2s use the same surface to interact with E1s and E3s, and hence while binding to an E3 enzyme, they cannot be recharged with ubiquitin by an E1. Thus, binding of E2 to E3 is necessarily weak in order to allow E2 to be involved in repeated cycles of interaction with E1 and E3 (Ye and Rape, 2009). In this sense, traditional interaction analysis methodologies, such as the yeast two-hybrid system, may not be sensitive enough to detect the E2–E3 interaction, and perhaps for this reason we did not detect any two-hybrid interaction between UBE2N and laforin or malin. We obtained better results when using GFP-trap, a pull-down assay based on the use of anti-GFP Terbinafine mg which has been previously employed to detect this kind of weak interactions (Hoxhaj et al., 2012). Using this technique, we observed that UBE2N interacted physically with the malin–laforin complex, and our results also suggested a stronger interaction between UBE2N and malin. The interaction also occurred when the UBE2N-C87A form was used in the assay, although with reduced intensity. This latter result may suggest that binding between UBE2N and malin is increased if ubiquitin is attached to the E2, as already reported for other E2–E3 complexes (Eddins et al., 2006). K63-linked ubiquitination of substrates has been related to autophagic degradation (Deshaies and Joazeiro, 2009, Tan et al., 2008). During this process, cargos are bound by specific adaptors which recognize both the ubiquitin chains and components in the autophagosome membrane, recruiting the substrates for their degradation (Lippai and Low, 2014). Previous publications have shown a relationship between LD and autophagy (Aguado et al., 2010, Criado et al., 2012). Here, we show that one of the specific adaptors that mediates selective autophagy, p62 (SQSTM1), interacts with the malin–laforin complex. This interaction was not dependent on the ubiquitination state of the complex, since in the presence of UBE2N-C87A, which is unable to bind ubiquitin, the interaction of p62 with the complex was maintained. Remarkably, binding of p62 to the malin–laforin complex enhanced its E3-ubiquitin ligase activity. As the malin–laforin complex generates K63-linked polyubiquitin chains (Moreno et al., 2010, Romá-Mateo et al., 2011, Rubio-Villena et al., 2013) and p62 seems to have a preference for this type of modifications (Seibenhener et al., 2004), the interaction between the malin–laforin complex and p62 is in agreement with the clearance of malin-dependent substrates by autophagy. In addition, the fact that p62 is ubiquitinated by the malin–laforin complex with K63-ubiquitin linked chains might create a positive feedback loop where the modified p62 could recruit more p62, facilitating in this way its oligomerization, which would induce autophagy, as reported in other cases (Johansen and Lamark, 2011, Shaid et al., 2013). We also present evidence indicating that malin–laforin complex and p62 form multimeric structures that are recognized by LC3 which would target them to the autophagosome (Fig. 8). In summary, we describe here two proteins that interact with the malin–laforin complex and modulate its ubiquitination activity. These new partners again highlight a link between proteins involved in LD and autophagy. Presumably, as we propose in Fig. 9, UBE2N interacts with the malin–laforin complex in order to promote the incorporation of K63-linked ubiquitin chains in the corresponding substrates. These chains would be recognized by p62, which would direct them to degradation via autophagy. This hypothesis would be in agreement with the previous observation that there is autophagy dysfunction in laforin- and malin-deficient mice (Aguado et al., 2010, Criado et al., 2012) and further corroborates the relevance of a functional malin–laforin complex in the maintenance of cellular protein homeostasis.