The intermediate filament synemin regulates non-homologous end joining DNA repair upon genotoxic damage in an ATM dependent manner


The intermediate filament synemin regulates non-homologous end joining DNA repair upon genotoxic damage in an ATM dependent manner

Deville, S. S.; Vehlow, A.; Förster, S.; Dickreuter, E.; Borgmann, K.; Cordes, N.

Background: Therapy resistance is a great challenge during cancer treatment. A well-known determinant of radiochemoresistance is cell adhesion to extracellular matrix. Targeting focal adhesion proteins (FAPs) has been shown to enhance cancer radiochemosensitivity in various tumor entities. Previous studies demonstrated a functional crosstalk between specific FAPs and DNA repair processes; however, the molecular mechanism remains unsolved. This study aimed to identify alternative FAPs associated with DNA damage repair mechanisms and radioresistance in head and neck squamous cell carcinomas (HNSCC).
Materials and Methods: A novel 3D High Throughput RNAi Screen (3DHT-RNAi-S) using laminin-rich extracellular matrix was established to determine radiation-induced residual DNA double strand breaks (DSBs) and clonogenic radiation survival using UTSCC15 cells expressing pEGFP-53BP1. Validations were performed in 10 3D grown HNSCC cell lines. DNA repair mechanisms, protein expression and kinetics post irradiation were investigated using immuno-fluorescence/-blotting, reporter assays for DSB repair activity and kinase activity profiling (PamGene) upon protein knockdown with/-out X-ray exposure. Protein-protein interactions were determined using immunoprecipitation (IP) and proximity ligation assay.
Results: In the 3DHT-RNAi-S, Synemin emerged as resulted one of the most promising candidates to determine HNSCC cell radiosensitivitysurvival and DNA damage repair. Synemin silencing radiosensitized HNSCC cells, while its exogenous overexpression induced radioprotection. Synemin depletion elicited a 40% reduction in non-homologous end joining activity without affecting other DNA DSB repair mechanisms. In line, ATM, DNA-PKcs and c-Abl phosphorylation as well as Ku70 expression strongly declined in synemin depleted and irradiated cells relative to controls. In kinome analysis, tyrosine kinases showed significantly reduced activity after synemin silencing relative to controls. Furthermore, IP revealed a protein complex formed between synemin, DNA-PKcs and c-Abl. This protein complex dispersed when ATM was pharmacologically inhibited. Using different protein constructs of synemin (ΔLink-Tail, ΔHead-Link, Synemin_301-961, Synemin_962-1565, S1114A and S1159A), the phosphorylation site at the serine 1114 located on the distal portion of synemin´s tail was identified as essential protein-protein interaction site involved in synemin´s function in DNA repair. Using different protein constructs with domain deletions of synemin, the distal portion of synemin´s tail was identified as essential protein site regulating synemin´s function in DNA repair processes.
Conclusions: The 3DHT-RNAi-S provides a robust screening platform for identifying novel targets involved in therapy resistance. Based on this screen and detailed mechanistic analyses, the intermediate filament synemin was discovered as a novel important determinant of DNA repair, tyrosine kinase activity and radioresistance of HNSCC cells. These results fundamentally support the concept of cytoarchitectural elements as co-regulators of nuclear events.further support the concept that DNA repair is regulated by cooperative interactions between nuclear and cytoplasmic proteins.

Keywords: Radioresistance; HNSCC; Synemin

  • Lecture (Conference)
    DeGBS, 23.09.2019, Mannheim, Deutschland

Permalink: https://www.hzdr.de/publications/Publ-29435
Publ.-Id: 29435