1. Academic Validation
  2. Preso enhances mGluR1-mediated excitotoxicity by modulating the phosphorylation of mGluR1-Homer1 complex and facilitating an ER stress after traumatic brain injury

Preso enhances mGluR1-mediated excitotoxicity by modulating the phosphorylation of mGluR1-Homer1 complex and facilitating an ER stress after traumatic brain injury

  • Cell Death Discov. 2024 Mar 26;10(1):153. doi: 10.1038/s41420-024-01916-5.
Zhuoyuan Zhang # 1 2 Xiangyu Gao # 1 Zhicheng Tian # 1 Erwan Yang # 1 Yutao Huang 1 Dan Liu 1 2 Shuhui Dai 1 Haofuzi Zhang 1 Mingdong Bao 1 Xiaofan Jiang 3 Xin Li 4 Peng Luo 5
Affiliations

Affiliations

  • 1 Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China.
  • 2 School of Life Science, Northwest University, Xi'an, China.
  • 3 Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China. [email protected].
  • 4 Department of Anesthesiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China. [email protected].
  • 5 Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China. [email protected].
  • # Contributed equally.
Abstract

Glutamate receptor (GluR)-mediated excitotoxicity is an important mechanism causing delayed neuronal injury after traumatic brain injury (TBI). Preso, as a core scaffolding protein of postsynaptic density (PSD), is considered an important regulator during excitotoxicity and TBI and combines with glutamate receptors to form functional units for excitatory glutamatergic neurotransmission, and elucidating the mechanisms of these functional units will provide new targets for the treatment of TBI. As a multidomain scaffolding protein, Preso directly interacts with metabotropic GluR (mGluR) and another scaffold protein, Homer. Because the mGluR-Homer complex plays a crucial role in TBI, modulation of this complex by Preso may be an important mechanism affecting the excitotoxic damage to neurons after TBI. Here, we demonstrate that Preso facilitates the interaction between metabotropic mGluR1 and Homer1 to activate mGluR1 signaling and cause excitotoxic neuronal injury and endoplasmic reticulum (ER) stress after TBI. The regulatory effect of Preso on the mGluR1-Homer1 complex is dependent on the direct association between Preso and this complex and also involves the phosphorylation of the interactive binding sites of mGluR1 and Homer1 by Preso. Further studies confirmed that Preso, as an adaptor of cyclin-dependent kinase 5 (CDK5), promotes the phosphorylation of the Homer1-binding site on mGluR1 by CDK5 and thereby enhances the interaction between mGluR1 and Homer1. Preso can also promote the formation of the mGluR1-Homer1 complex by inhibiting the phosphorylation of the Homer1 hinge region by Ca2+/calmodulin-dependent protein kinase IIα (CaMKIIα). Based on these molecular mechanisms, we designed several blocking Peptides targeting the interaction between Preso and the mGluR1-Homer1 complex and found that directly disrupting the association between mGluR1 and scaffolding proteins significantly promotes the recovery of motor function after TBI.

Figures
Products