In our project, the ultimate goal is to selectively degrade progerin, the mutant and toxic form of lamin A responsible for Hutchinson–Gilford Progeria Syndrome, while preserving the normal lamin A protein.
To achieve this high level of specificity, we used bioinformatics tools to design bait proteins capable of binding exclusively to progerin. By leveraging structure-based modeling, interface prediction, and sequence comparison, we generated and evaluated candidate interactors that specifically recognize the unique features of progerin’s altered C-terminal region.
Our computational approach integrates the AI-based models AlphaFold3, RFdiffusion, and ProteinMPNN, in combination with the molecular docking platforms HADDOCK and ClusPro and affinity estimation with PRODIGY. The pipeline begins with structural modeling and refinement of the target, proceeds through binder backbone and sequence design, and concludes with docking simulations and estimation of binding affinity.

How to test protein-protein interactions with an easy tool?
This is one of the questions we asked ourselves when we started working on ProgERASE. And the answer was the simplest existing eukaryote: Saccharomyces cerevisiae.
S. cerevisiae offers an amazing platform for the study of protein interactions called Yeast Two Hybrid (Y2H), a commonly known test that makes use of two plasmids co-transformed into the same cells (in our case pGAD and pGBK). If interaction occurs, a specific molecular pathway is activated, allowing us to observe the interaction.

We implemented the Y2H test by testing it firstly on a known progerin interactor, BUBR1. This protein provides a critical contribution to the Progeria phenotype which arises from progerin’s interaction with BUBR1, a core component of the spindle assembly checkpoint (SAC):
- The C-terminus of progerin binds strongly to the N-terminal region of BUBR1 (BUBR1-N);
- This tethering mislocalizes BUBR1 to the nuclear membrane in interphase cells;
- Consequently, BUBR1 cannot localize to the kinetochore or interact with other SAC proteins
The result is impaired checkpoint activity, accelerated cellular senescence, and severe mitotic defects, including chromosome mis-segregation and aneuploidy.
After implementing the Y2H test on BUBR1, we further validated our findings by testing the peptides predicted by our bioinformatics analysis
We also implemented an S. cerevisiae model of progeria; to do so, we made use of pYES2 plasmid to express progerin in a galactose-dependent manner. Our results show that progerin limit yeast cell growth and provides the basis for further studies regarding phenotype rescue, for example through the usage of a chemical library.
Our degradation system evolves from the RING-Bait system [1] , an advanced version of earlier models like TRIM-Away [2] and PROTACs. These systems use the RING domains of E3 ligases to mediate protein degradation via the Ubiquitin-Proteasome System (UPS), which degrades 90% of cellular proteins. PROTACs (Proteolysis-Targeting Chimeras) are engineered molecules that hijack the UPS to selectively degrade disease-causing proteins. Inspired by this concept, we focused on a PROTAC-like system based on the RING domain of TRIM proteins, particularly TRIM21 [3].
The TRIM Protein Family
TRIM (Tripartite Motif) proteins share a conserved N-terminal tripartite structure:
- RING domain: E3 domain that interacts with E2 enzymes, catalyzing polyubiquitination (mainly K48- and K63-linked).
- B-Boxes: modulate RING activity, sometimes with independent E3 ligase roles.
- Coiled-coil domain: mediates dimerization and structural stability.
Spotlight on TRIM21

TRIM21 is unique because it functions as both:
- An E3 ubiquitin ligase;
- A cytosolic antibody receptor, via its PRY/SPRY domain binding Fc antibodies;
The RING-Bait System for Progeria: Concept and Mechanism
The RING-Bait system fuses the TRIM21 RING domain with a protein-specific “Bait.” Once the Bait binds its target, clustering of RING domains occurs, triggering ubiquitination and proteasomal degradation. This system was first developed for degrading Tau aggregates in neurodegenerative diseases.
Our four-step strategy
-
Confirming RING’s nuclear activity
We tested the ability of RING to dimerize and induce degradation by co-transfecting RING-SpyCatcher and SpyTag-mEmEGFP-SpyTag constructs into MRC-5 cell lines, exploiting the interaction between SpyCatcher and SpyTag, two known self-assembling peptides. In this setup, the binding of RING-SpyCatcher to both SpyTags brings two RING domains into proximity, triggering proteasomal degradation of mEGFP. Transfection experiments in 24-well plates, followed by FACS analysis, will allow us to assess mEGFP degradation. Meanwhile, cell viability has to be monitored with AlamarBlue assays to ensure that fluorescence decrease is due to degradation rather than toxicity.
-
Testing progerin degradation
To evaluate progerin degradation, we focused on key pathological features of HGPS, such as increased ROS, reduced viability, and nuclear abnormalities. After confirming progerin expression through Western blot, we compared proliferation and viability between progerin-expressing cells and controls. Co-transfections with RING-SpyCatcher were then performed to determine whether degradation of progerin could reverse disease-associated phenotypes.
-
Validating peptide–progerin interactions
To test synthetic peptide binding to progerin, we used the NanoBiT® complementation assay. By fusing progerin and the candidate peptides to NanoLuc fragments, protein–peptide interactions could be monitored in living fibroblasts through luminescence. This strategy provided direct experimental evidence supporting our in silico predictions.
-
Assessing degradation in HGPS patient-derived cells
Finally, the most promising interactors were expressed in primary fibroblasts derived from a Progeria patient. This step provided a biologically relevant validation, allowing us to assess the potential of our RING-based system in a disease-specific cellular context.

-
Nuclear Localization:
- Progerin accumulates in the nucleus;
- While TRIM21 can shuttle into the nucleus, the PRY/SPRY domain (important for nuclear translocation) is absent from RING-Bait constructs
-
Specificity vs. Off-target Effects:
- Progerin is nearly identical to lamin A;
- Risk: RING-Bait may also degrade wild-type lamin A;
- However, HGPS mouse models lacking both progerin and lamin A showed a better phenotype than those expressing both, suggesting that lamin A degradation might be tolerable if progerin is effectively removed
-
Clustering Requirements:
- Tau forms large homogeneous aggregates that enable efficient clustering;
- Progerin aggregates incorporate lamin A and other proteins, possibly limiting RING activation
-
Delivery and Immune Response:
- Adeno-Associated Viruses (AAV vectors) can deliver RING-Bait systemically and even cross the blood-brain barrier. However, AAV can trigger immune responses in adults;
- In pediatric HGPS patients, reduced immune priming might make therapy more feasible
- [1] L.V.C. Miller et al. (2024). Co-opting templated aggregation to degrade pathogenic tau assemblies and improve motor function. Cell, https://doi.org/10.1016/j.cell.2024.08.024
- [2] D. Clift et al. (2017). A Method for the Acute and Rapid Degradation of Endogenous Proteins. Cell, 171. https://doi.org/10.1016/j.cell.2017.10.033
- [3] Jones EL, Laidlaw SM, Dustin LB (2021). TRIM21/Ro52 - Roles in Innate Immunity and Autoimmune Disease. Front Immunol, 12. https://doi.org/10.3389/fimmu.2021.738473