Movatterモバイル変換


[0]ホーム

URL:


Skip to main page content
U.S. flag

An official website of the United States government

Dot gov

The .gov means it’s official.
Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

Https

The site is secure.
Thehttps:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

NIH NLM Logo
Log inShow account info
Access keysNCBI HomepageMyNCBI HomepageMain ContentMain Navigation
pubmed logo
Advanced Clipboard
User Guide

Full text links

eLife Sciences Publications, Ltd full text link eLife Sciences Publications, Ltd Free PMC article
Full text links

Actions

Share

doi: 10.7554/eLife.10222.

Ternatin and improved synthetic variants kill cancer cells by targeting the elongation factor-1A ternary complex

Affiliations

Ternatin and improved synthetic variants kill cancer cells by targeting the elongation factor-1A ternary complex

Jordan D Carelli et al. Elife..

Abstract

Cyclic peptide natural products have evolved to exploit diverse protein targets, many of which control essential cellular processes. Inspired by a series of cyclic peptides with partially elucidated structures, we designed synthetic variants of ternatin, a cytotoxic and anti-adipogenic natural product whose molecular mode of action was unknown. The new ternatin variants are cytotoxic toward cancer cells, with up to 500-fold greater potency than ternatin itself. Using a ternatin photo-affinity probe, we identify the translation elongation factor-1A ternary complex (eEF1A·GTP·aminoacyl-tRNA) as a specific target and demonstrate competitive binding by the unrelated natural products, didemnin and cytotrienin. Mutations in domain III of eEF1A prevent ternatin binding and confer resistance to its cytotoxic effects, implicating the adjacent hydrophobic surface as a functional hot spot for eEF1A modulation. We conclude that the eukaryotic elongation factor-1A and its ternary complex with GTP and aminoacyl-tRNA are common targets for the evolution of cytotoxic natural products.

Keywords: biochemistry; cancer; cell biology; cyclic peptide; elongation factor-1A; human; natural product; protein synthesis; target identification.

PubMed Disclaimer

Conflict of interest statement

The authors declare that no competing interests exist.

Figures

Figure 1.
Figure 1.. New ternatin variants inspired byAspergillus-derived cyclic peptides.
(a) Design of ternatin variants3 and4, based on the partially elucidated structure of A3. (b) Effect of cyclic peptides 1–4(three-fold dilutions) on HCT116 cell proliferation over 72 hr. (c) Compounds1 and4 were tested against a panel of 21 cancer cell lines. Shown is a scatter plot comparing IC50 values of1 and4 for each cell line (Spearman correlation, 0.84, p < 0.0001).DOI:http://dx.doi.org/10.7554/eLife.10222.003
Figure 1—figure supplement 1.
Figure 1—figure supplement 1.. Epimers4 and4b have similar antiproliferative activity.
(a) Chemical structure of4b, containing (2S,4S)-dehydro-homoleucine. (b) Antiproliferative activity of4 and epimer4b toward HCT116 colorectal cancer cells (4b IC50 7.4 ± 1.3 nM).DOI:http://dx.doi.org/10.7554/eLife.10222.005
Figure 2.
Figure 2.. Ternatins inhibit global protein synthesis.
(a) HCT116 cells were treated with compound4 (five-fold dilutions) for 5 hr before labeling with35S-Met for 1 hr. Cell lysates were separated by gel electrophoresis. Newly synthesized and total proteins were visualized by autoradiography and Coomassie staining. (b) Cells were treated as in (a), and35S-labeled proteins were quantified by liquid scintillation counting after TCA precipitation (mean ± SEM, n = 3). (c) Left panel: HeLa cells were treated with4 (5 μM) or DMSO for 20 min. Right panel: HeLa cells were treated with4 (5 μM), cycloheximide (CHX, 100 μg/mL), or DMSO for 15 min, followed by harringtonine (HT, 2 μg/mL) for 20 min. After compound treatment, lysates were fractionated on 10–50% sucrose density gradients with absorbance detection at 254 nm.DOI:http://dx.doi.org/10.7554/eLife.10222.006
Figure 3.
Figure 3.. Photo-affinity labeling reveals eEF1A as a direct ternatin target.
(a) Clickable photo-affinity probe5. (b) HEK293T cell lysates were treated with5 (two-fold dilutions) for 20 min at room temp followed by UV irradiation (355 nm, 1000 W, 90 s). A control sample with 20 μM5 was not irradiated. Samples were subjected to click chemistry with TAMRA-azide, separated by gel electrophoresis, and scanned for in-gel fluorescence. (c) Cell lysates were treated with increasing concentrations of4 for 10 min before adding5 (2 μM) for 20 min, UV irradiation, and processing as in (b). (d) HEK293T cells were transfected with Flag-eEF1A. Lysates were treated with probe5 (2 μM) ±4 and photolyzed as in (b), then immunoprecipitated with magnetic anti-Flag beads. Samples were eluted with SDS, subjected to click chemistry with TAMRA-azide, and analyzed by in-gel fluorescence scanning and Western blotting. Coomassie-stained gels corresponding to (b) and (c) are shown in Figure 3—figure supplement 1.DOI:http://dx.doi.org/10.7554/eLife.10222.007
Figure 3—figure supplement 1.
Figure 3—figure supplement 1.. Ternatin photo-affinity probe specifically labels a 50-kDa protein.
(a) 72-hr cell proliferation assay with serial dilutions of5. (b) 72-hr HEK293T cell proliferation assay. (c) Coomassie-stained gel corresponding to Figure 3b. (d) Dependence of photo-crosslinking intensity on the concentration of photo-ternatin5 based on in-gel TAMRA fluorescence (Figure 3b). For each of the indicated bands (p40, p50, and p55), the background-corrected fluorescence intensity at a given concentration of5 was normalized to its intensity in the sample containing 20 μM5. Labeling of p50, but not p40 or p55, achieves saturation at ~10 μM5. (e) Coomassie-stained gel corresponding to Figure 3c.DOI:http://dx.doi.org/10.7554/eLife.10222.008
Figure 4.
Figure 4.. Photo-ternatin5 binds specifically to the eEF1A ternary complex.
(a) HEK293T cell lysates were treated with RNase A for 20 min before (lanes 3–4) or after (lanes 5–6) incubation with5 and UV irradiation. (b) Purified eEF1A was incubated with GTP ± Phe-tRNA or GDP for 30 min at room temp. Reactions were treated with DMSO or4 for 10 min, then5 for 20 min, photolyzed and processed as in Figure 3b. (c) Translation elongation inhibitors didemnin B (DB) and ansatrienin B (AB). (d) A solution of eEF1A, GTP, and Phe-tRNA was incubated with the indicated compound (DB and AB: 0.1, 1.0, 10 μM) for 10 min before 20-min treatment with5 (1 μM), followed by UV irradiation and processing as in Figure 3b. Coomassie-stained gels corresponding to (a) and (d) are shown in Figure 4—figure supplement 1.DOI:http://dx.doi.org/10.7554/eLife.10222.009
Figure 4—figure supplement 1.
Figure 4—figure supplement 1.. Coomassie-stained gels corresponding to (a) Figure 4a, and (b) Figure 4d.
In (b), the major 50-kDa band is eEF1A and the minor ~45-kDa band is an unknown contaminant.DOI:http://dx.doi.org/10.7554/eLife.10222.010
Figure 4—figure supplement 2.
Figure 4—figure supplement 2.. Photo-affinity labeling of HEK293T cell lysates by photo-ternatin5 in the presence of increasing concentrations of (a) didemnin B (DB), or (b) ansatrienin B (AB).
DOI:http://dx.doi.org/10.7554/eLife.10222.011
Figure 5.
Figure 5.. Ala399 mutation inEEF1A1 confers resistance to4.
(a) Effect of4 (two-fold dilutions) on proliferation of WT andEEF1A1-mutant HCT116 cells over 72 hr. (b) HEK293T cells were transfected with WT or A399V Flag-eEF1A. Lysates were treated with probe5 and processed as described in Figure 3d. (c) WT HCT116 cells (left) or cells homozygous for A399VEEF1A1 (right) were transduced with a bicistronic lentiviral vector encoding eEF1A (WT or A399V) and mCherry. Cells were labeled with carboxyfluorescein succinimidyl ester (CFSE), treated with4 for 72 hr (four-fold dilutions), and analyzed by FACS. Proliferation was assessed by CFSE dilution in high mCherry-expressing cells (mean fluorescence intensity, MFI = 6.7–7.4 × 104). CFSE histograms are shown in Figure 5—figure supplement 1. (d) Homozygous A399VEEF1A1 HCT116 cells were transduced with WT eEF1A/mCherry as described in (c). Antiproliferative effects of4 (left) were analyzed in cells gated according to the indicated mCherry mean fluorescence intensity (right).DOI:http://dx.doi.org/10.7554/eLife.10222.012
Figure 5—figure supplement 1.
Figure 5—figure supplement 1.. Ternatin sensitivity is genetically dominant.
WT HCT116 cells (top row) or cells homozygous for A399VEEF1A1 (bottom row) were transduced with a bicistronic lentiviral vector encoding WT eEF1A (left column) or A399V eEF1A (right column) linked to mCherry by the ribosome-skipping P2A peptide. Cells were labeled with carboxyfluorescein succinimidyl ester (CFSE), treated with increasing concentrations of 4 for 72 hr, and analyzed by FACS. (a) CFSE histograms for high mCherry cells (mCherry MFI = 6.7–7.4 × 104) were used to generate dose-response curves in Figure5c. (b) Scatter plots show mCherry and CFSE fluorescence of single cells treated with 10 μM 4. Note the correlation between mCherry and CFSE fluorescence among cells homozygous for A399VEEF1A1 and transduced with WT eEF1A (bottom left plot, red box). In this group, only those cells ectopically expressing high levels of WT eEF1A (indicated by high mCherry fluorescence) fail to proliferate in the presence of 10 μM 4 (indicated by high CFSE fluorescence).DOI:http://dx.doi.org/10.7554/eLife.10222.013
Figure 6.
Figure 6.. Ternatin may inhibit eEF1A by a mechanism related to kirromycin inhibition of EF-Tu.
Left: crystal structure of archaeal EF1A (PDB code: 3WXM, 51% identity with human eEF1A), showing the location of A399 (human numbering) on the surface of domain III. Right: crystal structure of EF-Tu (PDB code: 1OB2, 25% sequence identity with human eEF1A), showing the kirromycin binding site at the interface of domain I and III.DOI:http://dx.doi.org/10.7554/eLife.10222.014
Figure 7.
Figure 7.. Synthesis of (2S, 4R)- and (2S, 4S)-methyl 2-amino-4-methylhex-5-enoate (dehydro-homoleucine).
Reagents and conditions: (a)nBuLi, THF, –78°C; (b) TBAF, THF, 0°C; (c) Dess-Martin periodinane, CH2Cl2; (d) MePPh3Br,nBuLi, THF, 0°C; (e) TFA, H2O/MeCN.DOI:http://dx.doi.org/10.7554/eLife.10222.015
Figure 8.
Figure 8.. Synthesis of ternatin variants.
Reagents and conditions: (a) HATU, DIPEA, DCM/DMF; (b) 2 M HCl, MeOH, 30°C; (c) 1 N LiOH, H2O/THF; (d) HATU, DIPEA, DMF.DOI:http://dx.doi.org/10.7554/eLife.10222.016
Figure 9.
Figure 9.. Compound 12.
DOI:http://dx.doi.org/10.7554/eLife.10222.017
Figure 10.
Figure 10.. Compound 13.
DOI:http://dx.doi.org/10.7554/eLife.10222.018
Figure 11.
Figure 11.. Compound 14.
DOI:http://dx.doi.org/10.7554/eLife.10222.019
Figure 12.
Figure 12.. Compound 15.
DOI:http://dx.doi.org/10.7554/eLife.10222.020
Figure 13.
Figure 13.. Compound 18.
DOI:http://dx.doi.org/10.7554/eLife.10222.021
Figure 14.
Figure 14.. Compound 3.
DOI:http://dx.doi.org/10.7554/eLife.10222.022
Figure 15.
Figure 15.. Compound 4.
DOI:http://dx.doi.org/10.7554/eLife.10222.023
Figure 16.
Figure 16.. Compound 4b.
DOI:http://dx.doi.org/10.7554/eLife.10222.024
Figure 17.
Figure 17.. Compound 5.
DOI:http://dx.doi.org/10.7554/eLife.10222.025
See this image and copyright information in PMC

Similar articles

See all similar articles

Cited by

See all "Cited by" articles

References

    1. Ahuja D, Vera MD, SirDeshpande BV, Morimoto H, Williams PG, Joullié MM, Toogood PL. Inhibition of protein synthesis by Didemnin B: how EF-1alpha mediates inhibition of translocation. Biochemistry. 2000;39:4339–4346. doi: 10.1021/bi992202h. - DOI - PubMed
    1. Andersen GR, Nissen P, Nyborg J. Elongation factors in protein biosynthesis. Trends in Biochemical Sciences. 2003;28:434–441. doi: 10.1016/S0968-0004(03)00162-2. - DOI - PubMed
    1. Barretina J, Caponigro G, Stransky N, Venkatesan K, Margolin AA, Kim S, Wilson CJ, Lehár J, Kryukov GV, Sonkin D, Reddy A, Liu M, Murray L, Berger MF, Monahan JE, Morais P, Meltzer J, Korejwa A, Jané-Valbuena J, Mapa FA, Thibault J, Bric-Furlong E, Raman P, Shipway A, Engels IH, Cheng J, Yu GK, Yu J, Aspesi P, de Silva M, Jagtap K, Jones MD, Wang L, Hatton C, Palescandolo E, Gupta S, Mahan S, Sougnez C, Onofrio RC, Liefeld T, MacConaill L, Winckler W, Reich M, Li N, Mesirov JP, Gabriel SB, Getz G, Ardlie K, Chan V, Myer VE, Weber BL, Porter J, Warmuth M, Finan P, Harris JL, Meyerson M, Golub TR, Morrissey MP, Sellers WR, Schlegel R, Garraway LA. The cancer cell line encyclopedia enables predictive modelling of anticancer drug sensitivity. Nature. 2012;483:603–307. doi: 10.1038/nature11003. - DOI - PMC - PubMed
    1. Besemer J, Harant H, Wang S, Oberhauser B, Marquardt K, Foster CA, Schreiner EP, de Vries JE, Dascher-Nadel C, Lindley IJ. Selective inhibition of cotranslational translocation of vascular cell adhesion molecule 1. Nature. 2005;436:290–293. doi: 10.1038/nature03670. - DOI - PubMed
    1. Blunt J, Cole T, Munro M, Sun L, Weber J-F, Ramasamy K, Abu Bakar H, Abdul Majeed ABB. International Patent WO 2010/062159 A1 2010

Publication types

MeSH terms

Substances

Related information

Grants and funding

LinkOut - more resources

Full text links
eLife Sciences Publications, Ltd full text link eLife Sciences Publications, Ltd Free PMC article
Cite
Send To

NCBI Literature Resources

MeSHPMCBookshelfDisclaimer

The PubMed wordmark and PubMed logo are registered trademarks of the U.S. Department of Health and Human Services (HHS). Unauthorized use of these marks is strictly prohibited.


[8]ページ先頭

©2009-2025 Movatter.jp