Please wait a minute...
Frontiers of Chemical Science and Engineering

ISSN 2095-0179

ISSN 2095-0187(Online)

CN 11-5981/TQ

Postal Subscription Code 80-969

2018 Impact Factor: 2.809

Front. Chem. Sci. Eng.    2018, Vol. 12 Issue (2) : 283-295    https://doi.org/10.1007/s11705-017-1687-2
RESEARCH ARTICLE
Head-to-tail cyclization of a heptapeptide eliminates its cytotoxicity and significantly increases its inhibition effect on amyloid β-protein fibrillation and cytotoxicity
Shuai Ma1, Huan Zhang1, Xiaoyan Dong1(), Linling Yu1, Jie Zheng2, Yan Sun1
1. Department of Biochemical Engineering and Key Laboratory of Systems Bioengineering of the Ministry of Education, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China
2. Department of Chemical and Biomolecular Engineering, The University of Akron, Akron, OH 44325, USA
 Download: PDF(534 KB)   HTML
 Export: BibTeX | EndNote | Reference Manager | ProCite | RefWorks
Abstract

Amyloid-β (Aβ) protein aggregation is the main hallmark of Alzheimer’s disease (AD). Inhibition of Aβ fibrillation is thus a promising therapeutic approach to the prevention and treatment of AD. Recently, we designed a heptapeptide inhibitor, LVFFARK (LK7). LK7 shows a promising inhibitory capability on Aβ fibrillation, but is prone to self-assembling and displays high cytotoxicity, which would hinder its practical application. Herein, we modified LK7 by a head-to-tail cyclization and obtained a cyclic LK7 (cLK7). cLK7 exhibits a different self-assembly behavior from LK7, and has higher stability against proteolysis than LK7 and little cytotoxicity to SH-SY5Y cells. Thermodynamic analysis revealed that both LK7 and cLK7 could bind to Aβ40 by electrostatic interactions, hydrogen bonding and hydrophobic interactions, but the binding affinity of cLK7 for Aβ40 (KD = 4.96 µmol/L) is six times higher than that of LK7 (KD = 32.2 µmol/L). The strong binding enables cLK7 to stabilize the secondary structure of Aβ40 and potently inhibit its nucleation, fibrillation and cytotoxicity at extensive concentration range, whereas LK7 could only moderately inhibit Aβ40 fibrillation and cytotoxicity at low concentrations. The findings indicate that the peptide cyclization is a promising approach to enhance the performance of peptide-based amyloid inhibitors.

Keywords Alzheimer’s disease      amyloid β-protein      cyclic peptide      inhibition      protein aggregation     
Corresponding Author(s): Xiaoyan Dong   
Just Accepted Date: 25 September 2017   Online First Date: 11 January 2018    Issue Date: 09 May 2018
 Cite this article:   
Shuai Ma,Huan Zhang,Xiaoyan Dong, et al. Head-to-tail cyclization of a heptapeptide eliminates its cytotoxicity and significantly increases its inhibition effect on amyloid β-protein fibrillation and cytotoxicity[J]. Front. Chem. Sci. Eng., 2018, 12(2): 283-295.
 URL:  
https://academic.hep.com.cn/fcse/EN/10.1007/s11705-017-1687-2
https://academic.hep.com.cn/fcse/EN/Y2018/V12/I2/283
Fig.1  Scheme 1 Chemical structures of (a) LK7 and (b) cLK7
Fig.2  Characterization of LK7 and cLK7. TEM images of (a) LK7 and (b) cLK7 (200 µmol/L for both) after incubation at 37 °C for 72 h. The scale bar of 200 nm is for the two images. (c) The time course of intact peptides by incubating with α-chymotrypsin. Error bars represent the standard deviation of the measurement (n = 3). (d) Cytotoxicity induced by LK7 and cLK7 aggregates detected using SH-SY5Y cells. In the MTT assay, the cell viability treated with PBS buffer alone was set to 100%
Fig.3  Aggregation kinetics of Aβ40 incubated without and with different concentrations of (a) LK7, and (b) cLK7. Aβ40 concentration was 25 µmol/L
Concentration /(µmol?L1) Tlag /h Ttrans /h
LK7
0 14.5±1.1 8.3±0.8
25 14.9±0.9 8.9±1.0
50 14.5±0.8 11.5±1.5
100 15.0±0.6 15.2±1.6
200 12.1±1.4 16.2±2.7
cLK7
0 14.5±1.1 8.3±0.8
25 15.4±2.2 9.3±1.8
50 16.2±1.2 11.0±1.6
100 17.6±0.9 11.9±2.7
200 20.1±1.0 17.1±3.3
Tab.1  Lag time (Tlag) and transition time (Ttrans) for aggregation kinetics of 25 µmol/L Aβ40 in the presence of different concentrations of LK7 or cLK7
Fig.4  TEM images of Aβ40 (25 µmol/L) in the absence and presence of LK7 and cLK7. (a) Aβ40 alone; (b to e) Aβ40 with LK7 at concentrations of (b) 25 µmol/L, (c) 50 µmol/L, (d) 100 µmol/L and (e) 200 µmol/L; (f to i) Aβ40 with cLK7 at concentrations of (f) 25 µmol/L, (g) 50 µmol/L, (h) 100 µmol/L and (i) 200 µmol/L. The scale bar of 200 nm is for all the images. All samples were taken after incubation at 37 °C for 72 h
Fig.5  Far-UV circular dichroism spectra of Aβ40 (25 µmol/L) incubated in the absence and presence of different concentrations of (a) LK7 and (b) cLK7. The co-incubation of Aβ40 was conducted at 37 °C
Fig.6  Inhibitory effects of LK7 and cLK7 on Aβ40-induced cytotoxicity for SH-SY5Y cell lines: ***p <0.001, compared to control groups; #p<0.05, ##p<0.01, ###p<0.001, compared to the Aβ40 treated group (Aβ40 only)
LK7 cLK7
KD /(µmol?L1) 32.2±1.5 4.96±0.25
DH /(kcal?mol1) −4.25±0.18 −4.92±0.27
TDS/(kcal?mol1) 2.12±0.06 2.61±0.14
DG/(kcal?mol1) −6.37±0.24 −7.53±0.41
Tab.2  Thermodynamic parameters for the interactions between the inhibitors and Aβ40 at 37 °C
Fig.7  Schematic representations of Aβ40 aggregation and cytotoxicity: (a) on-pathway aggregation of Aβ40 monomers, (b) Aβ40 aggregation in the presence of LK7, and (c) Aβ40 aggregation in the presence of cLK7
1 Goedert M, Spillantini M G. A century of Alzheimer’s disease. Science, 2006, 314(5800): 777–781
https://doi.org/10.1126/science.1132814
2 Hardy J, Selkoe D J. The amyloid hypothesis of Alzheimer’s disease: Progress and problems on the road to therapeutics. Science, 2002, 297(5580): 353–356
https://doi.org/10.1126/science.1072994
3 Knowles T P, Vendruscolo M, Dobson C M. The amyloid state and its association with protein misfolding diseases. Nature Reviews. Molecular Cell Biology, 2014, 15(6): 384–396
https://doi.org/10.1038/nrm3810
4 Selkoe D J. Soluble oligomers of the amyloid β-protein impair synaptic plasticity and behavior. Behavioural Brain Research, 2008, 192(1): 106–113
https://doi.org/10.1016/j.bbr.2008.02.016
5 Chimon S, Shaibat M A, Jones C R, Calero D C, Aizezi B, Ishii Y. Evidence of fibril-like β-sheet structures in a neurotoxic amyloid intermediate of Alzheimer’s β-amyloid. Nature Structural & Molecular Biology, 2008, 14(12): 1157–1164
https://doi.org/10.1038/nsmb1345
6 Härd T, Lendel C. Inhibition of amyloid formation. Journal of Molecular Biology, 2012, 421(4): 441–465
https://doi.org/10.1016/j.jmb.2011.12.062
7 Wang Q M, Yu X, Li L Y, Zheng J. Inhibition of amyloid-β aggregation in Alzheimer’s disease. Current Pharmaceutical Design, 2014, 20(8): 1223–1243
https://doi.org/10.2174/13816128113199990068
8 Craik D J, Fairlie D P, Liras S, Price D. The future of peptide-based drugs. Chemical Biology & Drug Design, 2013, 81(1): 136–147
https://doi.org/10.1111/cbdd.12055
9 Tjernberg L O, Näslund J, Lindqvist F, Johansson J, Karlström A R, Thyberg J, Terenius L, Nordstedt C. Arrest of β-amyloid fibril formation by a pentapeptide ligand. Journal of Biological Chemistry, 1996, 271(15): 8545–8548
https://doi.org/10.1074/jbc.271.15.8545
10 Liu F F, Du W J, Sun Y, Zheng J, Dong X Y. Atomistic characterization of binding modes and affinity of peptide inhibitors to amyloid-β protein. Frontiers of Chemical Science and Engineering, 2014, 8(4): 433–444
https://doi.org/10.1007/s11705-014-1454-6
11 Soto C, Kindy M S, Baumann M, Frangione B. Inhibition of Alzheimer’s amyloidosis by peptides that prevent β-sheet conformation. Biochemical and Biophysical Research Communications, 1996, 226(3): 672–680
https://doi.org/10.1006/bbrc.1996.1413
12 Bansal S, Maurya I K, Yadav N, Thota C K, Kumar V, Tikoo K, Chauhan V S, Jain R. C-terminal fragment, Aβ32-37, analogues protect against Aβ aggregation-induced toxicity. ACS Chemical Neuroscience, 2016, 7(5): 615–623
https://doi.org/10.1021/acschemneuro.6b00006
13 Fradinger E A, Monien B H, Urbanc B, Lomakin A, Tan M, Li H, Spring S M, Condron M M, Cruz L, Xie C W, Benedek G B, Bitan G. C-terminal peptides coassemble into Aβ42 oligomers and protect neurons against Aβ42-induced neurotoxicity. Proceedings of the National Academy of Sciences of the United States of America, 2008, 105(37): 14175–14180
https://doi.org/10.1073/pnas.0807163105
14 Takahashi T, Mihara H. Peptide and protein mimetics inhibiting amyloid β-peptide aggregation. Accounts of Chemical Research, 2008, 41(10): 1309–1318
https://doi.org/10.1021/ar8000475
15 Turner J P, Lutzrechtin T, Moore K A, Rogers L, Bhave O, Moss M A, Servoss S L. Rationally designed peptoids modulate aggregation of amyloid-β 40. ACS Chemical Neuroscience, 2014, 5(7): 552–558
https://doi.org/10.1021/cn400221u
16 Arai T, Sasaki D, Araya T, Sato T, Sohma Y, Kanai M. A cyclic KLVFF-derived peptide aggregation inhibitor induces the formation of less-toxic off-pathway amyloid-β oligomers. ChemBioChem, 2014, 15(17): 2577–2583
https://doi.org/10.1002/cbic.201402430
17 Xiong N, Dong X Y, Zheng J, Liu F F, Sun Y. Design of LVFFARK and LVFFARK-functionalized nanoparticles for inhibiting amyloid β-protein fibrillation and cytotoxicity. ACS Applied Materials & Interfaces, 2015, 7(10): 5650–5662
https://doi.org/10.1021/acsami.5b00915
18 Arai T, Araya T, Sasaki D, Taniguchi A, Sato T, Sohma Y, Kanai M. Rational design and identification of a non-peptidic aggregation inhibitor of amyloid-β based on a pharmacophore motif obtained from cyclo [-Lys-Leu-Val-Phe-Phe-]. Angewandte Chemie International Edition, 2014, 53(31): 8236–8239
https://doi.org/10.1002/anie.201405109
19 Luo J H, Otero J M, Yu C H, Wärmländer S K, Gräslund A, Overhand M, Abrahams J P. Inhibiting and reversing amyloid-β peptide (1–40) fibril formation with gramicidin S and engineered analogues. Chemistry (Weinheim an der Bergstrasse, Germany), 2013, 19(51): 17338–17348
https://doi.org/10.1002/chem.201301535
20 Cho P Y, Joshi G, Boersma M D, Johnson J A, Murphy R M. A cyclic peptide mimic of the β-amyloid binding domain on transthyretin. ACS Chemical Neuroscience, 2015, 6(5): 778–789
https://doi.org/10.1021/cn500272a
21 Zheng J, Baghkhanian A M, Nowick J S. A hydrophobic surface is essential to inhibit the aggregation of a Tau-protein-derived hexapeptide. Journal of the American Chemical Society, 2013, 135(18): 6846–6852
https://doi.org/10.1021/ja310817d
22 Richman M, Wilk S, Chemerovski M, Wärmländer S K T S, Wahlström A, Gräslund A, Rahimipour S. In vitro and mechanistic studies of an antiamyloidogenic self-assembled cyclic D,L-α-peptide architecture. Journal of the American Chemical Society, 2013, 135(9): 3474–3484
https://doi.org/10.1021/ja310064v
23 Choi S J, Jeong W J, Kang S K, Lee M, Kim E, Ryu D Y, Lim Y B. Differential self-assembly behaviors of cyclic and linear peptides. Biomacromolecules, 2012, 13(7): 1991–1995
https://doi.org/10.1021/bm3005947
24 Ziehm T, Brener O, Groen T, Kadish I, Frenzel D, Tusche M, Kutzsche J, Reiss K, Gremer L, Nagel-Steger L, et al. Increase of positive net charge and conformational rigidity enhances the efficacy of D-enantiomeric peptides designed to eliminate cytotoxic Aβ species. ACS Chemical Neuroscience, 2016, 7(8): 1088–1096
https://doi.org/10.1021/acschemneuro.6b00047
25 March D R, Abbenante G, Bergman D A, Brinkworth R I, Wickramasinghe W, Begun J, Martin J L, Fairlie D P. Substrate-based cyclic peptidomimetics of Phe-Ile-Val that Inhibit HIV-1 protease using a novel enzyme-binding mode. Journal of the American Chemical Society, 1996, 118(14): 3375–3379
https://doi.org/10.1021/ja953790z
26 Rezai T, Yu B, Millhauser G L, Jacobson M P, Lokey R S. Testing the conformational hypothesis of passive membrane permeability using synthetic cyclic peptide diastereomers. Journal of the American Chemical Society, 2006, 128(8): 2510–2511
https://doi.org/10.1021/ja0563455
27 Wang Q M, Shah N, Zhao J, Wang C, Zhao C, Liu L, Li L Y, Zhou F, Zheng J. Structural, morphological, and kinetic studies of β-amyloid peptide aggregation on self-assembled monolayers. Physical Chemistry Chemical Physics, 2011, 13(33): 15200–15210
https://doi.org/10.1039/c1cp21156k
28 Gordon D J, Sciarretta K L, Meredith S C. Inhibition of β-amyloid (40) fibrillogenesis and disassembly of β-amyloid (40) fibrils by short β-amyloid congeners containing N-methyl amino acids at alternate residues. Biochemistry, 2001, 40(28): 8237–8245
https://doi.org/10.1021/bi002416v
29 Ferrie J J, Gruskos J J, Goldwaser A L, Decker M E, Guarracino D A. A comparative protease stability study of synthetic macrocyclic peptides that mimic two endocrine hormones. Bioorganic & Medicinal Chemistry Letters, 2013, 23(4): 989–995
https://doi.org/10.1016/j.bmcl.2012.12.041
30 Yu R, Seymour V A L, Berecki G, Jia X, Akcan M, Adams D J, Kaas Q, Craik D J. Less is more: Design of a highly stable disulfide-deleted mutant of analgesic cyclic α-conotoxin Vc1.1. Scientific Reports, 2015, 5(1): 13264
https://doi.org/10.1038/srep13264
31 Cheng P N, Liu C, Zhao M, Eisenberg D, Nowick J S. Amyloid β-sheet mimics that antagonize protein aggregation and reduce amyloid toxicity. Nature Chemistry, 2012, 4(11): 927–933
https://doi.org/10.1038/nchem.1433
32 Cabaleiro-Lago C, Szczepankiewicz O, Linse S. The effect of nanoparticles on amyloid aggregation depends on the protein stability and intrinsic aggregation rate. Langmuir, 2012, 28(3): 1852–1857
https://doi.org/10.1021/la203078w
33 Luo J H, Yu C H, Yu H X, Borstnar R, Kamerlin S C, Gräslund A, Abrahams J P, Wärmländer S K. Cellular polyamines promote amyloid-beta (Aβ) peptide fibrillation and modulate the aggregation pathways. ACS Chemical Neuroscience, 2013, 4(3): 454–462
https://doi.org/10.1021/cn300170x
34 Micsonai A, Wien F, Kernya L, Lee Y H, Goto Y, Réfrégiers M, Kardos J. Accurate secondary structure prediction and fold recognition for circular dichroism spectroscopy. Proceedings of the National Academy of Sciences of the United States of America, 2015, 112(24): 3095–3103
https://doi.org/10.1073/pnas.1500851112
35 Choi H J, Huber A H, Weis W I. Thermodynamics of β-catenin-ligand interactions: The roles of the N- and C-terminal tails in modulating binding affinity. Journal of Biological Chemistry, 2006, 281(2): 1027–1038
https://doi.org/10.1074/jbc.M511338200
36 Wiseman T, Williston S, Brandts J F, Lin L N. Rapid measurement of binding constants and heats of binding using a new titration calorimeter. Analytical Biochemistry, 1989, 179(1): 131–137
https://doi.org/10.1016/0003-2697(89)90213-3
37 Freyer M W, Lewis E A. Isothermal titration calorimetry: Experimental design, data analysis, and probing macromolecule/ligand binding and kinetic interactions. Methods in Cell Biology, 2008, 84: 79–113
https://doi.org/10.1016/S0091-679X(07)84004-0
38 Fotakis G, Timbrell J A. In vitro cytotoxicity assays: Comparison of LDH, neutral red, MTT and protein assay in hepatoma cell lines following exposure to cadmium chloride. Toxicology Letters, 2006, 160(2): 171–177
https://doi.org/10.1016/j.toxlet.2005.07.001
39 Gupta M, Bagaria A, Mishra A, Mathur P, Basu A, Ramakumar S, Chauhan V S. Self-assembly of a dipeptide-containing conformationally restricted dehydrophenylalanine residue to form ordered nanotubes. Advanced Materials, 2007, 19(6): 858–861
https://doi.org/10.1002/adma.200601774
40 Huang R L, Su R X, Qi W, Zhao J, He Z M. Hierarchical, interface-induced self-assembly of diphenylalanine: Formation of peptide nanofibers and microvesicles. Nanotechnology, 2011, 22(24): 245609
https://doi.org/10.1088/0957-4484/22/24/245609
41 Biancalana M, Koide S. Molecular mechanism of thioflavin-T binding to amyloid fibrils. Biochimica et Biophysica Acta (BBA)-Proteins Proteomics, 2010, 1804(7): 1405–1412
https://doi.org/10.1016/j.bbapap.2010.04.001
42 Cohen S I, Linse S, Luheshi L M, Hellstrand E, White D A, Rajah L, Otzen D E, Vendruscolo M, Dobson C M, Knowles T P. Proliferation of amyloid-β42 aggregates occurs through a secondary nucleation mechanism. Proceedings of the National Academy of Sciences of the United States of America, 2013, 110(24): 9758–9763
https://doi.org/10.1073/pnas.1218402110
43 Arosio P, Knowles T P, Linse S. On the lag phase in amyloid fibril formation. Physical Chemistry Chemical Physics, 2015, 17(12): 7606–7618
https://doi.org/10.1039/C4CP05563B
44 Zhao Z J, Zhu L, Li H Y, Cheng P, Peng J X, Yin Y D, Yang Y, Wang C, Hu Z Y, Yang Y L. Antiamyloidogenic activity of Aβ42-binding peptoid in modulating amyloid oligomerization. Small, 2017, 13(1): 1602857
https://doi.org/10.1002/smll.201602857
45 Sugiura Y, Ikeda K, Nakano M. High membrane curvature enhances binding, conformational changes, and fibrillation of amyloid-β on lipid bilayer surfaces. Langmuir, 2015, 31(42): 11549–11557
https://doi.org/10.1021/acs.langmuir.5b03332
46 Nagarathinam A, Höflinger P, Bühler A, Schäfer C, Mcgovern G, Jeffrey M, Staufenbiel M, Jucker M, Baumann F. Membrane-anchored Aβ accelerates amyloid formation and exacerbates amyloid-associated toxicity in mice. Journal of Neuroscience, 2013, 33(49): 19284–19294
https://doi.org/10.1523/JNEUROSCI.2542-13.2013
47 Bartolini M, Bertucci C, Bolognesi M L, Cavalli A, Melchiorre C, Andrisano V. Insight into the kinetic of amyloid-β (1–42) peptide self-aggregation: Elucidation of inhibitors’ mechanism of action. ChemBioChem, 2007, 8(17): 2152–2161
https://doi.org/10.1002/cbic.200700427
48 Ehrnhoefer D E, Bieschke J, Boeddrich A, Herbst M, Masino L, Lurz R, Engemann S, Pastore A, Wanker E E. EGCG redirects amyloidogenic polypeptides into unstructured, off-pathway oligomers. Nature Structural & Molecular Biology, 2008, 15(6): 558–566
https://doi.org/10.1038/nsmb.1437
49 Du W J, Guo J J, Gao M T, Hu S Q, Dong X Y, Han Y F, Liu F F, Jiang S, Sun Y. Brazilin inhibits amyloid β-protein fibrillogenesis, remodels amyloid fibrils and reduces amyloid cytotoxicity. Scientific Reports, 2015, 5(1): 7992
https://doi.org/10.1038/srep07992
50 Kumar S, Udgaonkar J B. Mechanisms of amyloid fibril formation by proteins. Current Science, 2010, 98(5): 639–656
51 Tu Y L, Ma S, Liu F F, Sun Y, Dong X Y. Hematoxylin inhibits amyloid β-protein fibrillation and alleviates amyloid-induced cytotoxicity. Journal of Physical Chemistry B, 2016, 120(44): 11360–11368
https://doi.org/10.1021/acs.jpcb.6b06878
52 Qiang W, Yau W M, Luo Y, Mattson M P, Tycko R. Antiparallel β-sheet architecture in iowa-mutant β-amyloid fibrils. Proceedings of the National Academy of Sciences of the United States of America, 2012, 109(12): 4443–4448
https://doi.org/10.1073/pnas.1111305109
53 Acerra N, Kad N M, Griffith D A, Ott S, Crowther D C, Mason J M. Retro-inversal of intracellular selected β-amyloid-interacting peptides: Implications for a novel Alzheimer’s disease treatment. Biochemistry, 2014, 53(13): 2101–2111
https://doi.org/10.1021/bi5001257
54 Liu J, Wang W, Zhang Q, Zhang S H, Yuan Z. Study on the efficiency and interaction mechanism of a decapeptide inhibitor of β-amyloid aggregation. Biomacromolecules, 2014, 15(3): 931–939
https://doi.org/10.1021/bm401795e
55 Ross P D, Subramanian S. Thermodynamics of protein association reactions: Forces contributing to stability. Biochemistry, 1981, 20(11): 3096–3102
https://doi.org/10.1021/bi00514a017
56 Cairo C W, Strzelec A, Murphy R M, Kiessling L L. Affinity-based inhibition of β-amyloid toxicity. Biochemistry, 2002, 41(27): 8620–8629
https://doi.org/10.1021/bi0156254
57 Liao Y H, Chang Y J, Yoshiike Y, Chang Y C, Chen Y R. Negatively charged gold nanoparticles inhibit Alzheimer’s amyloid-β fibrillization, induce fibril dissociation, and mitigate neurotoxicity. Small, 2012, 8(23): 3631–3639
https://doi.org/10.1002/smll.201201068
[1] FCE-17032-OF-MS_suppl_1 Download
[1] LIU Zhenfa, WANG Yanji, GAO Yuhua, ZHANG Lihui. Synergistic scale inhibition of polyaspartic acid composite with magnetic field[J]. Front. Chem. Sci. Eng., 2007, 1(3): 261-265.
Viewed
Full text


Abstract

Cited

  Shared   
  Discussed