Abstract
Proteolysis targeting chimeras (PROTACs) are bivalent molecules that bring a cellular protein to a ubiquitin ligase E3 for ubiquitination and subsequent degradation. Although PROTAC has emerged as a promising therapeutic means for cancers as it rewires the ubiquitin pathway to destroy key cancer regulators, the degradation signals/pathways for PROTACs remain underdeveloped. Here we append single amino acids, the simplest degradation signal, to a ligand specific for estrogen-related receptor α (ERRα) and demonstrate their utility in ERRα knockdown via the N-end rule pathway and also their efficiency in the growth inhibition of breast cancer cells. The modular design described offers unique advantages including smaller molecular size with shortest degradation sequences and degradation speed modulation with different amino acids. Our study expands the repertoire of limited ubiquitin pathways currently available for PROTACs and could be easily adapted for broad use in targeted protein degradation.
Keywords: protein degradation, protein turnover, proteolysis, proteasome, ubiquitin, drug design, drug development, protein targeting, drug therapy, N-end rule, PROTAC
Introduction
Proteolysis targeting chimeras (PROTAC)2 has emerged as an effective means for cancer therapy by shuttling cancer regulators to the proteasome for degradation (1–4). The principle of PROTAC-mediated protein knockdown strategy is based on ubiquitin-mediated proteolysis; a ubiquitin ligase (E3) recognizes its target and covalently attaches ubiquitin, a conserved 76-residue protein, onto the substrate, leading to subsequent degradation by the proteasome (1, 5, 6). PROTACs contain two moieties, which separately bind a relevant drug target and a ubiquitin ligase, with a flexible linker in the middle (1, 4). Thereby PROTACs rewire the ubiquitin-proteasome pathway to rapidly eliminate, rather than inhibit, specific target proteins on contact, which could minimize the risk that acquired defects develop over time and also phenotypes are reversed, common shortcomings of targeted therapies. Furthermore, as PROTACs require only transient drug-target binding without the need of direct inhibition of substrate activity, this approach could be easily used to manipulate previously undruggable cancer targets via selective degradation.
Of the two main components of PROTACs, whereas the ligands for various targets have been developed and refined over the years, the degradation motifs adapted for PROTACs remain underdeveloped (4, 7). Thus far, the degradation moieties specific for a few ubiquitin ligases (e.g. VHL, cereblon, MDM2, and XIAP) have been validated for PROTAC-mediated destruction of a variety of key cancer proteins (7, 8). However, one major caveat with these degradation signals employed is the lack of degradation speed control, which can be a problem as too fast or slow substrate turnover may be harmful in some cases. It is also important to reduce the sizes of PROTACs that currently are much larger than typical drugs (often less than 500 Da), which could improve cellular permeability and potency. Moreover, the ubiquitin ligases Cereblon and VHL, which are most frequently employed in PROTACs currently, may not be easily adopted for cancers in kidney, lung, and brain, due to their low expression/activity in these cells (9–11). Therefore, how to expand the repertoire of ubiquitin ligases and make this strategy more generally applicable remains a critical, pressing issue.
Results and discussion
We reason that a short binding motif to a potent ubiquitin ligase may greatly improve the utility of the PROTAC approach. The shortest degradation motif is a single amino acid, a destabilizing residue (e.g. Arg, His, Lys, Leu, and Ile) at the N terminus of a protein that targets the substrate to a ubiquitin ligase termed Ubr1 for rapid degradation via the N-end rule pathway, the first ubiquitin-dependent degradation pathway identified (12–14). We synthesized adaptor molecules starting with Arg or His and fused with a flexible linker (15) to a ligand for estrogen-related receptor α (ERRα) (3) (Fig. 1A), a nuclear receptor that is a major regulator of several critical metabolic pathways and also is a prognostic marker of breast cancer (16). ERRα inhibition has been shown to reduce the proliferation of breast tumor cells in vitro and in vivo. We found that both compounds Arg-TERRa and His-TERRa reduced the endogenous ERRα level in the MCF-7 metastatic breast cancer cell line (Fig. 1B). To assess whether the ERRα decrease was due to protein turnover, we treated the cells with cycloheximide to block protein synthesis and collected the samples at various time points for Western blot analysis. Both Arg-TERRa and His-TERRa triggered ERRα degradation (Fig. 1C and Fig. S1).
Figure 1.
Single amino acid-based PROTACs induce ERRα turnover. A, structure of PROTACs Arg-TERRa and His-TERRa. Arg and His were linked to the ligand previously demonstrated specifically for ERRα (3). B, Arg-TERRa and His-TERRa trigger ERRα reduction in a dose-dependent manner. Varying amounts of Arg-TERRa or His-TERRa were added to MCF-7 cells for 48 h. ERRα levels were determined by Western blot analysis. Equal amounts of protein extracts were used and ascertained by blotting with GAPDH antibody in the experiments. All experiments were done at least 3 times. C, ERRα stability was examined in the absence or presence of PROTACs by a protein expression shut-off assay. MCF-7 cells with or without PROTACs (10 μm Arg-TERRa and 5 μm His-TERRa) were treated with 200 μg/ml of cycloheximide to shut off protein synthesis. Samples were then collected at the indicated time points. Extracts were processed for immunoblotting with ERRα antibody. GAPDH serves as a loading control (lower panels). The experiments were done more than three times and quantified as shown in Fig. S1. A representative image is shown here.
We then evaluated whether PROTAC-induced ERRα degradation is mediated by the proteasome. We found that Arg-TERRa– or His-TERRa–triggered ERRα turnover was compromised upon the treatment of the proteasome inhibitor MG132 (Fig. 2, A and B, and Fig. S2), suggesting the involvement of the proteasome in ERRα turnover. To ascertain that ERRα is degraded by the N-end rule pathway, we took advantage that dipeptides starting with a destabilizing residue (e.g. Arg, His) can block the degradation of N-end rule substrates through their competition for the ubiquitin ligase Ubr1 (13, 14). We found that the PROTAC-exposed ERRα level was increased upon the addition of the dipeptide Arg-Ala or His-Ala, but not the control peptide Ala-Ala bearing a stabilizing residue (Fig. 2, C and D, and Fig. S3).
Figure 2.
ERRα turnover is blocked by the inhibitors of the proteasome and the N-end rule pathway. A and B, ERRα degradation involves the proteasome. MCF-7 cells were incubated with 10 μm Arg-TERRa or 5 μm His-TERRa for 32 h and then mixed with DMSO control or the proteasome inhibitor MG132 (25 μm). ERRα stability was determined after the addition of cycloheximide as described in the legend to Fig. 1C. C and D, Arg-TERRa- or His-TERRa-induced ERRα reduction was reversed by dipeptides Arg-Ala and His-Ala, respectively. MCF-7 cells incubated with 10 μm Arg-TERRa (C) or 5 μm His-TERRa (D) were treated with 100 μm dipeptides Arg-Ala (RA), His-Ala (HA), or control Ala-Ala (AA). Cell extracts were subjected to Western blot analysis to determine the levels of ERRα and control GAPDH.
We next examined the biological effects of the PROTAC molecules in MCF-7 cells. Both compounds led to decreased proliferation of MCF-7 cells (Fig. 3A). As cell migration is key to tumorigenesis, we then evaluated whether the compounds affect cell motility by the scratch wound healing method and the ThinCert cell migration assay. We found both Arg-TERRa and His-TERRa reduced MCF-7 cancer cell migration and wound repair (Fig. 3, B and C, and Fig. S4). Furthermore, we monitored the expression of several markers of epithelial-mesenchymal transition (EMT) that is crucial for cancer cell migration and invasion. PROTACs led to increased expression of E-cadherin and reduced expression of N-cadherin, SNAIL, and fibronectin (Fig. 3D), indicating EMT is repressed upon ERRα depletion. Combined, these results suggest that Arg-TERRa and His-TERRa could effectively modulate ERRα and be explored for anticancer purpose.
Figure 3.
Biological effects of Arg-TERRa and His-TERRa in MCF-7 cells. A, PROTACs Arg-TERRa and His-TERRa inhibit breast cancer cell proliferation. MCF-7 cells were treated with DMSO, 10 μm Arg-TERRa, or 5 μm His-TERRa. Cell proliferation was determined by the MTT assay after drug exposure on the indicated days. B, Arg-TERRa and His-TERRa impede wound healing. Confluent monolayers of MCF-7 cells were scraped by a pipette tip to generate wounds and then treated with DMSO, 10 μm Arg-TERRa, or 5 μm His-TERRa. Wound repair was photographed at the indicated time points. Scale bar, 100 μm. C, Arg-TERRa and His-TERRa reduce breast cancer cell migration. MCF-7 cells were allowed to invade Transwell chambers for 48 h in the presence or absence of PROTACs. Invaded cells were then fixed, stained, and photographed. Scale bar, 20 μm. D, effects of Arg-TERRa and His-TERRa on EMT markers. After the exposure to 10 μm Arg-TERRa or 5 μm His-TERRa, cell extracts were analyzed by Western blotting to examine the expression of various EMT markers including fibronectin, N-cadherin, SNAIL, and E-cadherin.
Our study demonstrates that the N-end rule-based PROTACs efficiently trigger target destruction and inhibit the proliferation of breast cancer cells. The advantage of our approach lies in the use of the simplest degradation signal-single amino acid, which can be any one of the 13 destabilizing residues in the N-end rule pathway (13, 14) that in turn may lead to different degradation speeds. The PROTAC method presented here overcomes limitations of existing approaches with significantly smaller molecular size of E3 targeting moiety, likely leading to higher permeability and better efficacy, and a modulatable potent degradation activity, allowing better control and broader application. Furthermore, the N-end rule degradation pathway is universally present and constitutively active, making it ideally suited for the PROTAC approach. This modular design expands the repertoire of limited ubiquitin pathways currently available for PROTACs and could be easily adapted for broad use in targeted protein degradation.
Experimental procedures
Cell cultures
The MCF-7 cell line (ATCC, Manassas, VA) was maintained at 37 °C with 5% CO2 in Dulbecco's modified Eagle's medium with 2 mm l-glutamine that was modified to contain 1 mm sodium pyruvate, 0.1 mm nonessential amino acids, and 1.5 g/liter of sodium bicarbonate. It was also supplemented with 10% fetal bovine serum, 10 μg/ml of bovine insulin, and 100 units/μl of penicillin/streptomycin. All drug treatment studies were supplemented with 2% FBS. Functional assays, including MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide), migration assays were done in 10% FBS in Dulbecco's modified Eagle's medium.
Peptides and compounds
Details of Arg-TERRa and His-TERRa syntheses are described under supporting information. Dipeptides Arg-Ala and His-Ala were purchased from Bachem Americas (Torrance, CA). MG132 is a proteasome inhibitor obtained from Calbiochem (Gibbstown, NJ). Cycloheximide and dipeptide Ala-Ala were purchased from Sigma.
Cell proliferation assay
The MTT method was used to measure living cells through mitochondrial dehydrogenase activity (Sigma). Cells were plated in a 96-well plate, 5000 cells/100 μl/well. After 24 h, the cells were treated with PROTACs in fresh medium. At the indicated time point, the medium was removed and DMSO was added as MTT solubilization solution, followed by 100 μl of stop solution. Absorbance was measured at 550 nm.
Western blot analysis
Cells were lysed in RIPA buffer (Sigma) with the addition of protease inhibitors tablet and phosphatase inhibitors mixture (Sigma). Lysates were resolved by SDS-polyacrylamide gels, transferred onto nitrocellulose membranes, and then probed with antibodies as indicated. Antibodies against ERRα, E-Cadherin, N-Cadherin, Snail, and GAPDH were obtained from Cell Signaling (Danvers, MA). Fibronectin antibody was obtained from Sigma.
Wound-healing assay
Cells were allowed to grow to near confluence in 6-well dishes. A uniform scratch was then made down the center of the plate using a 200-μl micropipette tip, followed by washing twice with PBS. The same marked field of the scratch wound was photographed using an Olympus light microscope (×4 objective) at the indicated time points. The width of the scratch wound was measured at three different areas with ImageJ software.
Migration assay
MCF-7 cells (1 × 104) were seeded on an 8 μm pore size Thin-cert for 24-well plates (Greiner Bio-One) in serum-free media. PROTAC compounds were added to the bottom chamber as chemoattractant. After 48 h, cells on the top of the membrane were removed with a cotton swab. The migrated cells at the bottom side were washed with PBS, fixed with 70% ethanol, and stained using 0.1% crystal violet to visualize the migrated cells. Migrated cells attached to the lower side of the membrane were enumerated using a light microscope at ×10 magnification.
Author contributions
K. S., P. S., H. C., B. C., S. F. M., and T. L. data curation; K. S. and H. R. formal analysis; K. S. validation; K. S., P. S., H. C., B. C., S. F. M., and T. L. investigation; K. S., P. S., H. C., B. C., S. F. M., T. L., and H. R. methodology; K. S., P. S., H. C., and T. L. writing-original draft; P. S., H. C., B. C., S. F. M., T. L., and H. R. writing-review and editing; S. F. M., T. L., and H. R. supervision; S. F. M. and H. R. project administration; T. L. and H. R. funding acquisition; H. R. conceptualization.
Supplementary Material
Acknowledgments
We are grateful to Drs. Y. Rao and J. Zhou for discussion.
This work was supported by Cancer Prevention Institute of Texas Grants RP170686 and RP180769, the Mays Cancer Center, the William & Ella Owens Medical Research Foundation, National Center for Advancing Translational Science Grant UL1TR001120 (to H. R.), the Peking-Tsinghua Center for Life Sciences, Beijing National Laboratory for Molecular Sciences, National Science Foundation of China Grants 31521004, 21672011, and 21822101, and Ministry of Science and Technology Grant 2017YFA0104000 (to T. L.), and Cancer Prevention Institute of Texas Grant RP160844 (to S. F. M.). The authors declare that they have no conflicts of interest with the contents of this article.
This article contains Figs. S1–S4 and supporting information.
- PROTAC
- proteolysis targeting chimeras
- ERRα
- estrogen-related receptor α
- EMT
- epithelial-mesenchymal transition
- MTT
- 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide
- GAPDH
- glyceraldehyde-3-phosphate dehydrogenase.
References
- 1. Sakamoto K. M., Kim K. B., Kumagai A., Mercurio F., Crews C. M., and Deshaies R. J. (2001) Protacs: chimeric molecules that target proteins to the Skp1-Cullin-F box complex for ubiquitination and degradation. Proc. Natl. Acad. Sci. U.S.A. 98, 8554–8559 10.1073/pnas.141230798 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2. Winter G. E., Buckley D. L., Paulk J., Roberts J. M., Souza A., Dhe-Paganon S., and Bradner J. E. (2015) Drug development: phthalimide conjugation as a strategy for in vivo target protein degradation. Science 348, 1376–1381 10.1126/science.aab1433 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. Bondeson D. P., Mares A., Smith I. E., Ko E., Campos S., Miah A. H., Mulholland K. E., Routly N., Buckley D. L., Gustafson J. L., Zinn N., Grandi P., Shimamura S., Bergamini G., Faelth-Savitski M., et al. (2015) Catalytic in vivo protein knockdown by small-molecule PROTACs. Nat. Chem. Biol. 11, 611–617 10.1038/nchembio.1858 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Paiva S. L., and Crews C. M. (2019) Targeted protein degradation: elements of PROTAC design. Curr. Opin Chem. Biol. 50, 111–119 10.1016/j.cbpa.2019.02.022 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. Zhou P., and Howley P. M. (1998) Ubiquitination and degradation of the substrate recognition subunits of SCF ubiquitin-protein ligases. Mol. Cell 2, 571–580 10.1016/S1097-2765(00)80156-2 [DOI] [PubMed] [Google Scholar]
- 6. Hu R., and Hochstrasser M. (2016) Recent progress in ubiquitin and ubiquitin-like protein (Ubl) signaling. Cell Res. 26, 389–390 10.1038/cr.2016.43 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Farnaby W., Koegl M., Roy M. J., Whitworth C., Diers E., Trainor N., Zollman D., Steurer S., Karolyi-Oezguer J., Riedmueller C., Gmaschitz T., Wachter J., Dank C., Galant M., Sharps B., et al. (2019) BAF complex vulnerabilities in cancer demonstrated via structure-based PROTAC design. Nat. Chem. Biol. 15, 672–680 10.1038/s41589-019-0294-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Ward C. C., Kleinman J. I., Brittain S. M., Lee P. S., Chung C. Y. S., Kim K., Petri Y., Thomas J. R., Tallarico J. A., McKenna J. M., Schirle M., and Nomura D. K. (2019) Covalent ligand screening uncovers a RNF4 E3 ligase recruiter for targeted protein degradation applications. ACS Chem. Biol. 10.1021/acschembio.8b01083 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Sekido Y., Bader S., Latif F., Gnarra J. R., Gazdar A. F., Linehan W. M., Zbar B., Lerman M. I., and Minna J. D. (1994) Molecular analysis of the von Hippel-Lindau disease tumor suppressor gene in human lung cancer cell lines. Oncogene 9, 1599–1604 [PubMed] [Google Scholar]
- 10. Zhu Y. X., Shi C. X., Bruins L. A., Wang X., Riggs D. L., Porter B., Ahmann J. M., de Campos C. B., Braggio E., Bergsagel P. L., and Stewart A. K. (2019) Identification of lenalidomide resistance pathways in myeloma and targeted resensitization using cereblon replacement, inhibition of STAT3 or targeting of IRF4. Blood Cancer J. 9, 19 10.1038/s41408-019-0173-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Senft D., Qi J., and Ronai Z. A. (2018) Ubiquitin ligases in oncogenic transformation and cancer therapy. Nat. Rev. Cancer 18, 69–88 10.1038/nrc.2017.105 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Bachmair A., Finley D., and Varshavsky A. (1986) In vivo half-life of a protein is a function of its amino-terminal residue. Science 234, 179–186 10.1126/science.3018930 [DOI] [PubMed] [Google Scholar]
- 13. Varshavsky A. (2017) The ubiquitin system, autophagy, and regulated protein degradation. Annu. Rev. Biochem. 86, 123–128 10.1146/annurev-biochem-061516-044859 [DOI] [PubMed] [Google Scholar]
- 14. Tasaki T., Sriram S. M., Park K. S., and Kwon Y. T. (2012) The N-end rule pathway. Annu. Rev. Biochem. 81, 261–289 10.1146/annurev-biochem-051710-093308 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Wurz R. P., Dellamaggiore K., Dou H., Javier N., Lo M. C., McCarter J. D., Mohl D., Sastri C., Lipford J. R., and Cee V. J. (2018) A “click chemistry platform” for the rapid synthesis of bispecific molecules for inducing protein degradation. J. Med. Chem. 61, 453–461 10.1021/acs.jmedchem.6b01781 [DOI] [PubMed] [Google Scholar]
- 16. Xia H., Dufour C. R., and Giguere V. (2019) ERRα as a bridge between transcription and function: role in liver metabolism and disease. Front. Endocrinol (Lausanne) 10, 206 [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.



