Targeting of δ-catenin to postsynaptic sites through interaction with the Shank3 N-terminus

Standard

Targeting of δ-catenin to postsynaptic sites through interaction with the Shank3 N-terminus. / Hassani Nia, Fatemeh; Woike, Daniel; Martens, Victoria; Klüssendorf, Malte; Hönck, Hans-Hinrich; Harder, Sönke; Kreienkamp, Hans-Jürgen.

In: MOL AUTISM, Vol. 11, No. 1, 28.10.2020, p. 85.

Research output: SCORING: Contribution to journalSCORING: Journal articleResearchpeer-review

Harvard

Hassani Nia, F, Woike, D, Martens, V, Klüssendorf, M, Hönck, H-H, Harder, S & Kreienkamp, H-J 2020, 'Targeting of δ-catenin to postsynaptic sites through interaction with the Shank3 N-terminus', MOL AUTISM, vol. 11, no. 1, pp. 85. https://doi.org/10.1186/s13229-020-00385-8

APA

Vancouver

Hassani Nia F, Woike D, Martens V, Klüssendorf M, Hönck H-H, Harder S et al. Targeting of δ-catenin to postsynaptic sites through interaction with the Shank3 N-terminus. MOL AUTISM. 2020 Oct 28;11(1):85. https://doi.org/10.1186/s13229-020-00385-8

Bibtex

@article{2fb9172753974a379d12bece24d3885b,
title = "Targeting of δ-catenin to postsynaptic sites through interaction with the Shank3 N-terminus",
abstract = "BACKGROUND: Neurodevelopmental disorders such as autism spectrum disorder (ASD) may be caused by alterations in genes encoding proteins that are involved in synapse formation and function. This includes scaffold proteins such as Shank3, and synaptic adhesion proteins such as Neurexins or Neuroligins. An important question is whether the products of individual risk genes cooperate functionally (exemplified in the interaction of Neurexin with Neuroligin isoforms). This might suggest a common pathway in pathogenesis. For the SHANK3 gene, heterozygous loss of function, as well as missense mutations have been observed in ASD cases. Several missense mutations affect the N-terminal part of Shank3 which contains the highly conserved Shank/ProSAP N-terminal (SPN) and Ankyrin repeat (Ank) domains. The role of these domains and the relevance of these mutations for synaptic function of Shank3 are widely unknown.METHODS: We used purification from a synaptic protein fraction, as well as a variety of biochemical and cell biological approaches to identify proteins which associate with the Shank3 N-terminus at postsynaptic sites.RESULTS: We report here that δ-catenin, which is encoded by CTNND2, an autism candidate gene, directly interacts with the Ank domain of Shank3 at postsynaptic sites through its Armadillo-repeat domain. The interaction is not affected by well-known posttranslational modifications of δ-catenin, i.e. by phosphorylation or palmitoylation. However, an ASD-associated mutation in the SPN domain of Shank3, L68P, significantly increases the interaction of Shank3 with δ-catenin. By analysis of postsynaptic fractions from mice, we show that the lack of SPN-Ank containing, large isoforms of Shank3 results in the loss of postsynaptic δ-catenin. Further, expression of Shank3 variants containing the N-terminal domains in primary cultured neurons significantly increased the presence of coexpressed δ-catenin at postsynaptic sites.LIMITATIONS: Work in model organisms such as mice, and in primary cultured neurons may not reproduce faithfully the situation in human brain neurons. Work in primary cultured neurons was also hampered by lack of a specific antibody for endogenous δ-catenin.CONCLUSIONS: Our data show that the interaction between Shank3 N-terminus and δ-catenin is required for the postsynaptic targeting of δ-catenin. Failure of proper targeting of δ-catenin to postsynaptic sites may contribute to the pathogenesis of autism spectrum disorder.",
author = "{Hassani Nia}, Fatemeh and Daniel Woike and Victoria Martens and Malte Kl{\"u}ssendorf and Hans-Hinrich H{\"o}nck and S{\"o}nke Harder and Hans-J{\"u}rgen Kreienkamp",
year = "2020",
month = oct,
day = "28",
doi = "10.1186/s13229-020-00385-8",
language = "English",
volume = "11",
pages = "85",
journal = "MOL AUTISM",
issn = "2040-2392",
publisher = "BioMed Central Ltd.",
number = "1",

}

RIS

TY - JOUR

T1 - Targeting of δ-catenin to postsynaptic sites through interaction with the Shank3 N-terminus

AU - Hassani Nia, Fatemeh

AU - Woike, Daniel

AU - Martens, Victoria

AU - Klüssendorf, Malte

AU - Hönck, Hans-Hinrich

AU - Harder, Sönke

AU - Kreienkamp, Hans-Jürgen

PY - 2020/10/28

Y1 - 2020/10/28

N2 - BACKGROUND: Neurodevelopmental disorders such as autism spectrum disorder (ASD) may be caused by alterations in genes encoding proteins that are involved in synapse formation and function. This includes scaffold proteins such as Shank3, and synaptic adhesion proteins such as Neurexins or Neuroligins. An important question is whether the products of individual risk genes cooperate functionally (exemplified in the interaction of Neurexin with Neuroligin isoforms). This might suggest a common pathway in pathogenesis. For the SHANK3 gene, heterozygous loss of function, as well as missense mutations have been observed in ASD cases. Several missense mutations affect the N-terminal part of Shank3 which contains the highly conserved Shank/ProSAP N-terminal (SPN) and Ankyrin repeat (Ank) domains. The role of these domains and the relevance of these mutations for synaptic function of Shank3 are widely unknown.METHODS: We used purification from a synaptic protein fraction, as well as a variety of biochemical and cell biological approaches to identify proteins which associate with the Shank3 N-terminus at postsynaptic sites.RESULTS: We report here that δ-catenin, which is encoded by CTNND2, an autism candidate gene, directly interacts with the Ank domain of Shank3 at postsynaptic sites through its Armadillo-repeat domain. The interaction is not affected by well-known posttranslational modifications of δ-catenin, i.e. by phosphorylation or palmitoylation. However, an ASD-associated mutation in the SPN domain of Shank3, L68P, significantly increases the interaction of Shank3 with δ-catenin. By analysis of postsynaptic fractions from mice, we show that the lack of SPN-Ank containing, large isoforms of Shank3 results in the loss of postsynaptic δ-catenin. Further, expression of Shank3 variants containing the N-terminal domains in primary cultured neurons significantly increased the presence of coexpressed δ-catenin at postsynaptic sites.LIMITATIONS: Work in model organisms such as mice, and in primary cultured neurons may not reproduce faithfully the situation in human brain neurons. Work in primary cultured neurons was also hampered by lack of a specific antibody for endogenous δ-catenin.CONCLUSIONS: Our data show that the interaction between Shank3 N-terminus and δ-catenin is required for the postsynaptic targeting of δ-catenin. Failure of proper targeting of δ-catenin to postsynaptic sites may contribute to the pathogenesis of autism spectrum disorder.

AB - BACKGROUND: Neurodevelopmental disorders such as autism spectrum disorder (ASD) may be caused by alterations in genes encoding proteins that are involved in synapse formation and function. This includes scaffold proteins such as Shank3, and synaptic adhesion proteins such as Neurexins or Neuroligins. An important question is whether the products of individual risk genes cooperate functionally (exemplified in the interaction of Neurexin with Neuroligin isoforms). This might suggest a common pathway in pathogenesis. For the SHANK3 gene, heterozygous loss of function, as well as missense mutations have been observed in ASD cases. Several missense mutations affect the N-terminal part of Shank3 which contains the highly conserved Shank/ProSAP N-terminal (SPN) and Ankyrin repeat (Ank) domains. The role of these domains and the relevance of these mutations for synaptic function of Shank3 are widely unknown.METHODS: We used purification from a synaptic protein fraction, as well as a variety of biochemical and cell biological approaches to identify proteins which associate with the Shank3 N-terminus at postsynaptic sites.RESULTS: We report here that δ-catenin, which is encoded by CTNND2, an autism candidate gene, directly interacts with the Ank domain of Shank3 at postsynaptic sites through its Armadillo-repeat domain. The interaction is not affected by well-known posttranslational modifications of δ-catenin, i.e. by phosphorylation or palmitoylation. However, an ASD-associated mutation in the SPN domain of Shank3, L68P, significantly increases the interaction of Shank3 with δ-catenin. By analysis of postsynaptic fractions from mice, we show that the lack of SPN-Ank containing, large isoforms of Shank3 results in the loss of postsynaptic δ-catenin. Further, expression of Shank3 variants containing the N-terminal domains in primary cultured neurons significantly increased the presence of coexpressed δ-catenin at postsynaptic sites.LIMITATIONS: Work in model organisms such as mice, and in primary cultured neurons may not reproduce faithfully the situation in human brain neurons. Work in primary cultured neurons was also hampered by lack of a specific antibody for endogenous δ-catenin.CONCLUSIONS: Our data show that the interaction between Shank3 N-terminus and δ-catenin is required for the postsynaptic targeting of δ-catenin. Failure of proper targeting of δ-catenin to postsynaptic sites may contribute to the pathogenesis of autism spectrum disorder.

U2 - 10.1186/s13229-020-00385-8

DO - 10.1186/s13229-020-00385-8

M3 - SCORING: Journal article

C2 - 33115499

VL - 11

SP - 85

JO - MOL AUTISM

JF - MOL AUTISM

SN - 2040-2392

IS - 1

ER -