当前位置:高等教育资讯网  >  中国高校课件下载中心  >  大学文库  >  浏览文档

复旦大学:《药物设计学》课程教学资源(教学研究)基于靶点结构的药物设计_Molecular Insights on the Cyclic Peptide Nanotube-Mediated Transportation of Antitumor Drug

资源类别:文库,文档格式:PDF,文档页数:10,文件大小:4.07MB,团购合买
点击下载完整版文档(PDF)

pharmaceutics articles Molecular Insights on the cyclic Peptide Nanotube-Mediated Transportation of Antitumor Drug 5-Fluorouracil Huifang liu, Jian Chen, Qing Shen, Wei Fu, and Wei Wu School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai, 201203, P. R. China Received August 19, 2010: Revised Manuscript Received October 11, 2010: Accepted October 21. 2010 Abstract: Self-assembled cyclic peptide nanotubes(CPNs)show a potential use in drug delivery In this study, the CPN composed of (Trp-D-Leu)4-GIn-D-Leu was synthesized and tested for the transport of the antitumor drug 5-fluorouracil(5-FU). CPN-mediated release of 5-FU from liposomes experimentally tested the transportation function of the synthetic CPNs. To explore the transportation mechanism of CPNs, computational studies have been performed on the CPN models stacked by 8 subuints, including conventional molecular dynamics simulations, and steered molecular dynamics(SMD) simulations in the environment of hy dimyristoylphosphatidylcholine(DMPC)lipid bilayer. Our CMD simulations demonstrat the ortho-CPN is the most stable nanotube, in which the GIn residue is in the ortho-position relative to other residues the calculated diffusion coefficient value for inner water molecules as 1.068 x 10-5 cm2.s-1, almost half that of the bulky water and 24 times faster than that of the typical gramicidin A channel. The CPN conserved its hollow structure along the 10 ns CMD simulations, with a tile angle of 50 relative to the normal of DMPC membrane. Results from SMD simulations showed that the 5-FU molecule was transported by hopping through different potential energy minima distributed along subunits, and finally exited the nanotube by escaping from the kink region at the last two subunits. The hopping of 5-FU was driven by switching from interactions of 5-FU with the backbone carbonyl group and amide group of ortho-CPN.The calculated binding free energy profile of 5-FU interacting with the CPn indicated that there was an energy well near the outer end of the nanotube Keywords: 5-fluorouracil; Cyclic peptide nanotube; steered molecular dynamics; drug trans porter; antitumor Introduction tubes are emerging as promising and selective drug trans- Malignant tumor is the most severe disease threatening porters. Our preliminary in vitro and in vivo studies have the health of human beings. More and more antitumor drugs demonstrated that the synthetic decedapeptide tube could have been discovered. However, the biggest problem is that efficiently enhance the antitumor potency of 5-fluorouracil these drugs could not efficiently exhibit their activity in (5-FU)(unpublished data) killing tumors due to the difficulty in their ability to penetrate Cyclic peptide nanotubes(CPNs) are a class of artificial through the cell membrane. Synthetic cyclic peptide nano- channels formed by closed peptide rings which consist of sCorrespondingauthors.E-mail:wfu@fudan.edu.cn(WF) wuwei@fudan.edu.cn(W.W.).Mailingaddress:FudanUni-(1)Adson,A.Burton,PS:Raub,TJ:Barsuhn,CL:Audus versity, School of Pharmacy, 826 Zhangheng Road, Shanghai K. L: Ho, N. F. Passive diffusion of weak organic electrolytes 201203,P.R. China.Fax:+86-21-51980010. Phone:+86- across Caco-2 cell monolayers: uncoupling the contributions of 21-51980010. hydrodynamic, transcellular, and paracellular barriers. J. Pharm 10.1021/mp100274f 2010 American Chemical Society VOL 7, NO. 6, 1985-1994 MOLECULAR PHARMACEUTICS 1985 Published on Web 10/21/2010

Molecular Insights on the Cyclic Peptide Nanotube-Mediated Transportation of Antitumor Drug 5-Fluorouracil Huifang Liu,† Jian Chen,† Qing Shen, Wei Fu,* and Wei Wu* School of Pharmacy, Fudan UniVersity, 826 Zhangheng Road, Shanghai, 201203, P. R. China Received August 19, 2010; Revised Manuscript Received October 11, 2010; Accepted October 21, 2010 Abstract: Self-assembled cyclic peptide nanotubes (CPNs) show a potential use in drug delivery. In this study, the CPN composed of (Trp-D-Leu)4-Gln-D-Leu was synthesized and tested for the transport of the antitumor drug 5-fluorouracil (5-FU). CPN-mediated release of 5-FU from liposomes experimentally tested the transportation function of the synthetic CPNs. To explore the transportation mechanism of CPNs, computational studies have been performed on the CPN models stacked by 8 subuints, including conventional molecular dynamics (CMD) simulations, and steered molecular dynamics (SMD) simulations in the environment of hydrated dimyristoylphosphatidylcholine (DMPC) lipid bilayer. Our CMD simulations demonstrated that the ortho-CPN is the most stable nanotube, in which the Gln residue is in the ortho-position relative to other residues. The calculated diffusion coefficient value for inner water molecules was 1.068 × 10-5 cm2 · s-1, almost half that of the bulky water and 24 times faster than that of the typical gramicidin A channel. The CPN conserved its hollow structure along the 10 ns CMD simulations, with a tile angle of 50° relative to the normal of DMPC membrane. Results from SMD simulations showed that the 5-FU molecule was transported by hopping through different potential energy minima distributed along subunits, and finally exited the nanotube by escaping from the kink region at the last two subunits. The hopping of 5-FU was driven by switching from hydrophobic interactions between 5-FU and the interior wall of the nanotube to hydrogen bonding interactions of 5-FU with the backbone carbonyl group and amide group of ortho-CPN. The calculated binding free energy profile of 5-FU interacting with the CPN indicated that there was an energy well near the outer end of the nanotube. Keywords: 5-fluorouracil; Cyclic peptide nanotube; steered molecular dynamics; drug trans￾porter; antitumor Introduction Malignant tumor is the most severe disease threatening the health of human beings. More and more antitumor drugs have been discovered. However, the biggest problem is that these drugs could not efficiently exhibit their activity in killing tumors due to the difficulty in their ability to penetrate through the cell membrane.1 Synthetic cyclic peptide nano￾tubes are emerging as promising and selective drug trans￾porters. Our preliminary in Vitro and in ViVo studies have demonstrated that the synthetic decedapeptide tube could efficiently enhance the antitumor potency of 5-fluorouracil (5-FU) (unpublished data). Cyclic peptide nanotubes (CPNs) are a class of artificial channels formed by closed peptide rings which consist of an even number of alternating D- and L-amino acid residues.2 * Corresponding authors. E-mail: wfu@fudan.edu.cn (W.F.), wuwei@fudan.edu.cn (W.W.). Mailing address: Fudan Uni￾versity, School of Pharmacy, 826 Zhangheng Road, Shanghai, 201203, P. R. China. Fax: +86-21-51980010. Phone: +86- 21-51980010. † These authors contributed equally to this work. (1) Adson, A.; Burton, P. S.; Raub, T. J.; Barsuhn, C. L.; Audus, K. L.; Ho, N. F. Passive diffusion of weak organic electrolytes across Caco-2 cell monolayers: uncoupling the contributions of hydrodynamic, transcellular, and paracellular barriers. J. Pharm. Sci. 1995, 84 (10), 1197–1204. articles 10.1021/mp100274f  2010 American Chemical Society VOL. 7, NO. 6, 1985–1994 MOLECULAR PHARMACEUTICS 1985 Published on Web 10/21/2010

articles By simply adjusting the number and kinds of amino acid Asp-D-Phe )2-] 2 In 1998, studies by polarized attenuated residues, both the internal diameter and external surface total reflectance infrared (ATR-IR)spectroscopy showed that properties can be tailored. Generally, the interior surface of CPNs oriented themselves in a transport-competent mem- such a tube is hydrophilic as indicated by the presence of brane orientation. The cyclic peptide nanotubes could act water molecules, while the exterior surface is hydrophobic, as highly selective and efficient transmembrane channels for permitting facile dissolution in nonpolar solvents. A network ions and small molecules. In 1994, Ghadiri confirmed that of hydrogen bonds between oxygen atoms of the participating the synthetic decapeptide cyclo[-(Trp-D-Leu)4-GIn-D-Leu-I carbonyl groups and amino groups at the backbone of has the function of glucose transportation. Since then adjacent cyclic peptide subunits drives them into a quasi-B- atomic-level theoretical and computational work has helped strand self-assembled tubular structure. The internal diam- to better understand the characteristics of structure, dynamics eter of the nanotube could be adjusted by simply varying and transport activity of the cyclic peptide nanotubes. Engels the size of the peptide ring. Electron diffraction analysis et al. found that cyclic D, L-octapeptide could self-assemble indicated that the tightly hydrogen-bonded rings stacked with as nanotubes. The diffusion of water molecules inside that an average intersubunit distance of 4.7 A. The cyclic D, L- peptide nanotube was much faster than that inside the peptides with appropriate hydrophobic side chains can self- gramicidin A channel. Asthagiri et al. calculated the semble and insert into lipid bilayers, and finally transport solvation free energies of Li, Nat, rb and Cl inside a ons or guest molecules across the membrane self-assembled (D, L)-octapeptide nanotubes using MD-based Studies on CPNs began from the year of 1974 and became perturbation free energy calculations; Tarek et al. found a hot research topic in the area of ion or small molecule that peptide nanotubes could tilt along the normal of lipid transportation. Both experimental and theoretical studies on bilayer after the nanotubes were equilibrated, but the hollow CPNs have been devoted to an understanding of their tubular structure was conserved. Hwang et al. calculated structural and dynamical characteristics, and transportation the free energy barrier for Na and k ions diffusing through properties. In 1993, Ghadiri et al. reported the first well- the cyclic peptide nanotube to be x2. 4 kcal/mol, and found characterized structure of self-assembled cyclic octapeptide that the carbonyl groups of the cyclic peptide were structu subunits cyclo[-(D-Ala-Glu-D-Ala-Gln)2-) in crystalline ally rigid because of the network of hydrogen bonding nanotubular arrays, convincingly establishing that the ring- All these studies confirmed that cyclic peptide readily self- aped subunits can stack through antiparallel B-sheet assembles to be a nanotube with a potential transportation hydrogen bonds to form hollow tubes. Later, Lambert and function. The questions are how the size and kind of peptide co-workers demonstrated their result by the Ir spectrum and determine the properties of a nanotube, and how the morphology of the crystals formed by cyclo[-(Asn-D-Phe- assembled nanotube transports guest molecules like drugs Answers to these questions have great significance and (2)Ghadir, M.R. Self-assembled nanoscale tubular ensembles. Adu. potential applications in the area of drug delivery Mater.1995,7(7),675-677 (3) Buriak, J M; Ghadiri, M. R. Self-assembly of peptide based (12) Polaskova, M. E; Ede, N. J. Lambert. J. N. Synthesis of nanotubes. Mater. Sci. Eng: C 1997, 4(4), 207-212. nanotubule-forming cyclic octapeptides via an Fmoc strategy. Aust (4)Karlstrom, A: Unden, A. Association of cyclic peptides in aqueous J.Chem.1998,51(7),535-540 solution measured by fluorescence quenching Biopolymers 1997 (3) Kim, H. S: Hartgerink, J. D. Ghadiri, M. R. Oriented self- 4(1),1 ssembly of cyclic peptide nanotubes in lipid membranes. J.Am (5)Hartgerink, J. D: Granja, J. R; Milligan, R. A: Ghadiri, M.R. Self-assembling peptide nanotubes. J. A. Chem. Soc. 1996. I (14) Sanchez-Quesada, J: Ghadiri, M.R.: Bayley, H: Braha, O. Cyclic ptides as molecular adapters for a pore-forming protein. J.A. (6) Ghadir, M.R.; Granja, J.R. Buchler, L. K. Artificial Trans- Chem. Soc. 2000, 122(48), 11757-11766. Ashkenasy, G; Ghadir embrane lon Channels from Self-Assembling Peptide Nanotubes. M. R. Boolean logic functions of a synthetic peptide network Nature1994,369(6478),301-304 Am.Chem.Soc.2004,126(36,ll140-11141 (7)Desantis, P ; Morosett, S: Rizzo, R. Conformational-Analysis of (15)Granja, J. R: Ghadiri, M. R. Channel-Mediated Transport of Regular Enantiomeric Sequences. Macromolecules 1974, 7(1) Glucose across Lipid Bilayers. J. Am. Chem. Soc. 1994, 116(23) 52-58. 10785-10786. (8)Liu, Z. W: Xu, Y Tang, P. Steered molecular dynamics (16) Engels, M: Bashford, D. Ghadir, M.R. Structure and Dynamics Na of Self-Assembling Peptide Nanotubes and the Channel-Mediated J.Phys.Chem.B2006.ll0(25),12789-12795 Water Organization and Self-Diffusion-A Molecular-Dynamics (9)Chen, G J. Su, S.J.; Liu, R. Z. Theoretical studies of monomer Study.J.Am.Chem.soc.1995,17(36),9151-9158 d dimer of cyclo[(-L-Phe'-D-Ala'-)ml and cyclo((-L-Phe' (7)Asthagiri, D; Pratt, L. R: Ashbaugh, H. S. Absolute hydration EN-Ala2-)a](n=3-6).Phys. Chem. B2002,106(7),15 free energies of ions, ion-water clusters, and quasichemical theory chen.Phys.2003,I19(5),2702-2708 (10)Lewis, J. P; Pawley, N. H: Sankey, O. F. Theoretical investigation (18) Tarek, M : Maigret, B: Hipot, C. Molecular dynamics investiga- of the cyclic peptide system cyclo((D-Ala-Glu-D-Ala-GIn)meI-4l tion of an oriented cyclic peptide nanotube in DMPC bilayer J.Phys.Chem.B1997,lO1(49),10576-1058 Biophys.J.2003.85(4),2287-2298 (l) Ghadir, M.R. Granja, J.R. Milligan, R. A: McRee, D. E:(19) Hwang, H: Schatz, G. C: Ratner, M. A. Steered molecular Khazanovich, N. Self-Assembling Organic Nanotubes Based on dynamics studies of the potential of mean force of a Na* or K+ Cyclic Peptide Architecture. Nature 1993, 366 ion in a cyclic peptide nanotube. J. Phys. Che. B 2006. 110 (51),2644826460. 1986 MOLECULAR PHARMACEUTICS VOL 7. NO 6

By simply adjusting the number and kinds of amino acid residues, both the internal diameter and external surface properties can be tailored. Generally, the interior surface of such a tube is hydrophilic as indicated by the presence of water molecules, while the exterior surface is hydrophobic, permitting facile dissolution in nonpolar solvents.3 A network of hydrogen bonds between oxygen atoms of the participating carbonyl groups and amino groups at the backbone of adjacent cyclic peptide subunits drives them into a quasi-- strand self-assembled tubular structure.4 The internal diam￾eter of the nanotube could be adjusted by simply varying the size of the peptide ring. Electron diffraction analysis indicated that the tightly hydrogen-bonded rings stacked with an average intersubunit distance of ∼4.7 Å.5 The cyclic D,L￾peptides with appropriate hydrophobic side chains can self￾assemble and insert into lipid bilayers, and finally transport ions or guest molecules across the membrane.6 Studies on CPNs began from the year of 1974 and became a hot research topic in the area of ion or small molecule transportation.7 Both experimental and theoretical studies on CPNs have been devoted to an understanding of their structural and dynamical characteristics, and transportation properties.8-10 In 1993, Ghadiri et al. reported the first well￾characterized structure of self-assembled cyclic octapeptide subunits cyclo[-(D-Ala-Glu-D-Ala-Gln)2-)] in crystalline nanotubular arrays, convincingly establishing that the ring￾shaped subunits can stack through antiparallel -sheet hydrogen bonds to form hollow tubes.11 Later, Lambert and co-workers demonstrated their result by the IR spectrum and morphology of the crystals formed by cyclo[-(Asn-D-Phe￾Asp-D-Phe)2-].12 In 1998, studies by polarized attenuated total reflectance infrared (ATR-IR) spectroscopy showed that CPNs oriented themselves in a transport-competent mem￾brane orientation.13 The cyclic peptide nanotubes could act as highly selective and efficient transmembrane channels for ions and small molecules.14 In 1994, Ghadiri confirmed that the synthetic decapeptide cyclo[-(Trp-D-Leu)4-Gln-D-Leu-] has the function of glucose transportation.15 Since then, atomic-level theoretical and computational work has helped to better understand the characteristics of structure, dynamics and transport activity of the cyclic peptide nanotubes. Engels et al. found that cyclic D,L-octapeptide could self-assemble as nanotubes. The diffusion of water molecules inside that peptide nanotube was much faster than that inside the gramicidin A channel.16 Asthagiri et al. calculated the solvation free energies of Li+, Na+, Rb+ and Cl- inside a self-assembled (D,L)-octapeptide nanotubes using MD-based perturbation free energy calculations;17 Tarek et al. found that peptide nanotubes could tilt along the normal of lipid bilayer after the nanotubes were equilibrated, but the hollow tubular structure was conserved.;18 Hwang et al. calculated the free energy barrier for Na+ and K+ ions diffusing through the cyclic peptide nanotube to be ∼2.4 kcal/mol, and found that the carbonyl groups of the cyclic peptide were structur￾ally rigid because of the network of hydrogen bonding.19 All these studies confirmed that cyclic peptide readily self￾assembles to be a nanotube with a potential transportation function. The questions are how the size and kind of peptide determine the properties of a nanotube, and how the assembled nanotube transports guest molecules like drugs. Answers to these questions have great significance and (2) Ghadiri, M. R. Self-assembled nanoscale tubular ensembles. AdV. potential applications in the area of drug delivery. Mater. 1995, 7 (7), 675–677. (3) Buriak, J. M.; Ghadiri, M. R. Self-assembly of peptide based nanotubes. Mater. Sci. Eng.: C 1997, 4 (4), 207–212. (4) Karlstrom, A.; Unden, A. Association of cyclic peptides in aqueous solution measured by fluorescence quenching. Biopolymers 1997, 41 (1), 1–4. (5) Hartgerink, J. D.; Granja, J. R.; Milligan, R. A.; Ghadiri, M. R. Self-assembling peptide nanotubes. J. Am. Chem. Soc. 1996, 118 (1), 43–50. (6) Ghadiri, M. R.; Granja, J. R.; Buehler, L. K. Artificial Trans￾membrane Ion Channels from Self-Assembling Peptide Nanotubes. Nature 1994, 369 (6478), 301–304. (7) Desantis, P.; Morosett, S.; Rizzo, R. Conformational-Analysis of Regular Enantiomeric Sequences. Macromolecules 1974, 7 (1), 52–58. (8) Liu, Z. W.; Xu, Y.; Tang, P. Steered molecular dynamics simulations of Na+ permeation across the gramicidin a channel. J. Phys. Chem. B 2006, 110 (25), 12789–12795. (9) Chen, G. J.; Su, S. J.; Liu, R. Z. Theoretical studies of monomer and dimer of cyclo[(-L-Phe1 -D-Ala2 -)n] and cyclo[(-L-Phe1 -D- MeN-Ala2 -)n] (n ) 3-6). J. Phys. Chem. B 2002, 106 (7), 1570– 1575. (10) Lewis, J. P.; Pawley, N. H.; Sankey, O. F. Theoretical investigation of the cyclic peptide system cyclo[(D-Ala-Glu-D-Ala-Gln)m)1-4]. J. Phys. Chem. B 1997, 101 (49), 10576–10583. (11) Ghadiri, M. R.; Granja, J. R.; Milligan, R. A.; McRee, D. E.; Khazanovich, N. Self-Assembling Organic Nanotubes Based on a Cyclic Peptide Architecture. Nature 1993, 366 (6453), 324– 327. (12) Polaskova, M. E.; Ede, N. J.; Lambert, J. N. Synthesis of nanotubule-forming cyclic octapeptides via an Fmoc strategy. Aust. J. Chem. 1998, 51 (7), 535–540. (13) Kim, H. S.; Hartgerink, J. D.; Ghadiri, M. R. Oriented self￾assembly of cyclic peptide nanotubes in lipid membranes. J. Am. Chem. Soc. 1998, 120 (18), 4417–4424. (14) Sanchez-Quesada, J.; Ghadiri, M. R.; Bayley, H.; Braha, O. Cyclic peptides as molecular adapters for a pore-forming protein. J. Am. Chem. Soc. 2000, 122 (48), 11757–11766. Ashkenasy, G.; Ghadiri, M. R. Boolean logic functions of a synthetic peptide network. J. Am. Chem. Soc. 2004, 126 (36), 11140–11141. (15) Granja, J. R.; Ghadiri, M. R. Channel-Mediated Transport of Glucose across Lipid Bilayers. J. Am. Chem. Soc. 1994, 116 (23), 10785–10786. (16) Engels, M.; Bashford, D.; Ghadiri, M. R. Structure and Dynamics of Self-Assembling Peptide Nanotubes and the Channel-Mediated Water Organization and Self-DiffusionsA Molecular-Dynamics Study. J. Am. Chem. Soc. 1995, 117 (36), 9151–9158. (17) Asthagiri, D.; Pratt, L. R.; Ashbaugh, H. S. Absolute hydration free energies of ions, ion-water clusters, and quasichemical theory. J. Chem. Phys. 2003, 119 (5), 2702–2708. (18) Tarek, M.; Maigret, B.; Chipot, C. Molecular dynamics investiga￾tion of an oriented cyclic peptide nanotube in DMPC bilayers. Biophys. J. 2003, 85 (4), 2287–2298. (19) Hwang, H.; Schatz, G. C.; Ratner, M. A. Steered molecular dynamics studies of the potential of mean force of a Na+ or K+ ion in a cyclic peptide nanotube. J. Phys. Chem. B 2006, 110 (51), 26448–26460. articles Liu et al. 1986 MOLECULAR PHARMACEUTICS VOL. 7, NO. 6

Molecular Insights on Antitumor Drug 5-Fluorouracil articles Scheme 1. Two-Dimensional Structure of the cyclo[-(Trp-D-Leu)4-GIn-D-Leu-] Peptide L H L NH In this work, we synthesized the cyclo[-(Trp-D-Leu)4-GIn- peptide was analyzed by reverse phase high performance D-Leu-] subunit, performed dialysis studies of 5-FU-loaded liquid chromatography(RP-HPLC) in order to determine the liposomes in the environment of cyclic peptide solution, and end point of the cyclization reaction, and the crude product investigated the antitumor activity by means of in vitro was purified by semipreparative RP-HPLC. Finally, the studies Based on our preliminary experimental findings we cyclized peptide was structurally analyzed by ESI-MS and examined the structural and dynamical characteristics of H NMR. CPN-mediated transportation of 5-FU from lipo- CPNS in a fully hydrated dimyristoylphosphatidylcholine somes was studied by an in vitro dialysis method. First, (MPC)lipid bilayer by conventional molecular dynamics 5-FU-loaded liposomes and cyclic peptides in dilute DMF (CMD) simulations. The transportation mechanism of 5-FU solution were sealed in a dialysis bag and dialyzed against by the CPNs is explored by means of steered molecular phosphate buffer saline(pH 7. 3)as the receptor phase(30 dynamics(SMD)simulations. Our results show that synthetic mL, 37C, 100 rpm) within a period of 90 min. The release CPN is able to efficiently transport the drug 5-FU by hopping of 5-FU was tested at 2 mg/mL concentrations of the cyclic the 5-FU molecule through different energy minima along peptides. The amount of the released 5-FU was quantified the nanotube. Our findings are helpful for the design of more at each time interval by HPLC. The mean values of six efficient drug delivery vehicles replicates were reported. Description of the Model Systems. The structural model Experimental Methods of cyclo[-(Trp-D-Leu)4-Gln-D-Leu-] nanotube was built Synthesis of cyclo[-(Trp-D-Leu)-GIn-D-Leu-1 Pep. according to Ghadiri,s model and modified according to the ide and Transportation Test. cyclo[-(Trp-D-Leu)4-Gln X-ray crystallographic structures of related peptide ensem D-Leu-] peptide(as shown in Scheme 1)was synthesized bles by using Sybyl 6.9 software (Tripos Inc, St and cyclized by a traditional solid-phase synthesis strategy. 20 Louis, MO). Although our experiments did not determine Briefly, the linear decapeptide was assembled by standard the exact number of subunits in one CPn nanotube. we Boc chemistry in the solid phase and subsequently cyclized selected the CPN assembly of 8 peptide subunits in order to in solution with high efficiency and reproducibility. L roughly match the thickness of the DMPC bilayer. All the Glutamine was adopted in the amino acid sequence for the side chains of the 8 peptides point outward attributed to their convenience of the peptide synthesis. The synthetic cyclic alternated D and L chirality. As the L-Gln could have three (20) Chen, J. Zhang, B; Xie, C; Lu, Y; Wu, w. Synthesis of a highly (21)Zhu, J. C; Cheng, J; Liao, Z.X.; Lai, Z.H. Liu, B Investigation hydrophobic cyclic decapeptide by solid-phase synthesis of linear of structures and properties of cyclic peptide nanotubes by peptide and cyclization in solution. Chin. Che. Lett. 2010 experiment and molecular dynamics. J Comput.-Aided Mol Des. 4),391-394. 2008,22(1),773-781. VOL 7. NO 6 MOLECULAR PHARMACEUTICS 1987

In this work, we synthesized the cyclo[-(Trp-D-Leu)4-Gln￾D-Leu-] subunit, performed dialysis studies of 5-FU-loaded liposomes in the environment of cyclic peptide solution, and investigated the antitumor activity by means of in Vitro studies. Based on our preliminary experimental findings we examined the structural and dynamical characteristics of CPNs in a fully hydrated dimyristoylphosphatidylcholine (DMPC) lipid bilayer by conventional molecular dynamics (CMD) simulations. The transportation mechanism of 5-FU by the CPNs is explored by means of steered molecular dynamics (SMD) simulations. Our results show that synthetic CPN is able to efficiently transport the drug 5-FU by hopping the 5-FU molecule through different energy minima along the nanotube. Our findings are helpful for the design of more efficient drug delivery vehicles. Experimental Methods Synthesis of cyclo[-(Trp-D-Leu)4-Gln-D-Leu-] Pep￾tide and Transportation Test. cyclo[-(Trp-D-Leu)4-Gln￾D-Leu-] peptide (as shown in Scheme 1) was synthesized and cyclized by a traditional solid-phase synthesis strategy.20 Briefly, the linear decapeptide was assembled by standard Boc chemistry in the solid phase and subsequently cyclized in solution with high efficiency and reproducibility. L￾Glutamine was adopted in the amino acid sequence for the convenience of the peptide synthesis. The synthetic cyclic peptide was analyzed by reverse phase high performance liquid chromatography (RP-HPLC) in order to determine the end point of the cyclization reaction, and the crude product was purified by semipreparative RP-HPLC. Finally, the cyclized peptide was structurally analyzed by ESI-MS and 1 H NMR. CPN-mediated transportation of 5-FU from lipo￾somes was studied by an in Vitro dialysis method. First, 5-FU-loaded liposomes and cyclic peptides in dilute DMF solution were sealed in a dialysis bag and dialyzed against phosphate buffer saline (pH 7.3) as the receptor phase (30 mL, 37 °C, 100 rpm) within a period of 90 min. The release of 5-FU was tested at 2 mg/mL concentrations of the cyclic peptides. The amount of the released 5-FU was quantified at each time interval by HPLC. The mean values of six replicates were reported. Description of the Model Systems. The structural model of cyclo[-(Trp-D-Leu)4-Gln-D-Leu-] nanotube was built according to Ghadiri’s model and modified according to the X-ray crystallographic structures of related peptide ensem￾bles11,18,21 by using Sybyl 6.9 software (Tripos Inc., St. Louis, MO). Although our experiments did not determine the exact number of subunits in one CPN nanotube, we selected the CPN assembly of 8 peptide subunits in order to roughly match the thickness of the DMPC bilayer. All the side chains of the 8 peptides point outward attributed to their alternated D and L chirality. As the L-Gln could have three (20) Chen, J.; Zhang, B.; Xie, C.; Lu, Y.; Wu, W. Synthesis of a highly hydrophobic cyclic decapeptide by solid-phase synthesis of linear peptide and cyclization in solution. Chin. Chem. Lett. 2010, 21 (4), 391–394. (21) Zhu, J. C.; Cheng, J.; Liao, Z. X.; Lai, Z. H.; Liu, B. Investigation of structures and properties of cyclic peptide nanotubes by experiment and molecular dynamics. J. Comput.-Aided Mol. Des. 2008, 22 (11), 773–781. Scheme 1. Two-Dimensional Structure of the cyclo[-(Trp-D-Leu)4-Gln-D-Leu-] Peptide Molecular Insights on Antitumor Drug 5-Fluorouracil articles VOL. 7, NO. 6 MOLECULAR PHARMACEUTICS 1987

articles different positions relative to other amino acids in the peptide set to 4.5 x 10, 4.5x 10, 4.5x 10-, 0, 0, 0 bar for chain, namely the ortho-position, meta-position and para- xx, yy, z, xy/yx, xzx and ya/zy components for water and position, the 8 cyclic peptides are stacked in three possible DMPC simulations. All bond lengths, including those to low-energy modes(see Figure SI in the Supporting Informa hydrogen atoms, were constrained by the LINCs algorithm. tion). Therefore, three low-energy stacking models of CPn Electrostatic interactions between charged groups within 9 were constructed For each CPN, the interior tube diameter A were calculated explicitly, while long-range electrostatic is 10 A, the side chains of L-Trp and the D-Leu are distributed interactions were calculated using the Particle-Mesh Ewald uniformly, and the center-to-center distance between neigh- method26 with a grid width of 1.2 A and a fourth-order spline boring subunits is 4.75 A. For convenience, the nanotube interpolation. A cutoff distance of 14 A was applied for the subunits are numerically denoted. As displayed in Figure S1 Lennard-Jones interactions. Numerical integration of the equa in the Supporting Information, APR(a-plane region) rep- tions of motion used a time step of 2 fs with atomic coordinates resents the plane of Ca atoms along one peptide subunit, saved every I ps for later analysis. Finally, three 10 ns MD while MPR (midplane region) represents the region between simulations were performed on these systems under the periodic two APRs. The whole length of one CPN is 38.7 A according boundary conditions in the NPT canonical ensemble. to the distance from apri to aprs Constant Velocity SMD Simulations. The SMD has Conventional Molecular Dynamics Simulation. After proved as an effective computational approach to simulate each of the three CPNs (i.e, ortho-CPN, meta-CPN and the transportation process of a small molecule permeating para-CPN) was built, it was inserted into the fully hydrated through a protein channel. In SMD simulations, a guest DMPC bilayer by aligning the axis of Cpn to the normal of molecule or an ion of interest is steered by an imaginary the lipid bilayer. The process of aligning CPN with the atomic force microscopy(AFM) tip, and the time-dependent normal of the DMPC membrane was similar to those used external force is added on the guest molecule to facilitate in our previous membrane protein simulations. When its transportation through the channel. In the present study, solvating the CPN/DMPC system, 42 Na and 42 CI ions we performed SMD simulations in order to explore how the were added in order to simulate the 150 mM physiological 5-FU molecule is transported by the CPn channel. In detail, ion strength. The size of the whole solvated system was 77 5-FU was pulled through the tube of the CPn by employing A X 83 A X 110 A, including one CPN, 189 DMPC an artificial harmonic force on the center of mass(COM)of molecules and 15087 water molecules 5-FU along the longitudinal axis of the CPN (Figure 1). 5-FU Energy minimizations were performed for each of the three molecule was first placed on the top of CPN, 1.27 nm from CPN/DMPC/water systems, first for all water molecules, ther the center of subunit 1, and then the whole system was for the whole system until the maximum force became equilibrated for 1 ns. The molecular topology file for 5-FU smallerthan10.00kcal/mol.a.Theenergy-minimizedCpn/wasgeneratedbytheProDrgserver(http://davapcl DMPC/water system was then subjected to MD simulation. bioch. dundee. ac uk/prodrg). The partial atomic charges of The MD simulations were performed by using the GROMACS 5-FU were determined with the DFT/B3LYP/6-31IG**basis package version 3.3.3 with the GROMOS96 force field. 23 set by using the CHelpG method implemented in GAMESS The solvent(water and DMPC)molecules of each initial program. To avoid large fluctuation in the position, a stiff the solute(CPNs)at 300 K for 20 ps. Then the Cpn was 5-FU. It should be pointed out that the pulling velocity (Pull) equilibrated for 5 ps while the solvent molecules were Is an important parameter in our SMD simulations. Higher constrained at 10, 50, 100, 200, and 298 K. Afterward, each pulling velocity may lead to remarkable nonequilibrium effects, system was equilibrated for 500 ps without any constraints. resulting in obvious errors of the simulation results. Very low To maintain the systems at a constant temperature of 300 velocity will make the SMD simulations extremely time- K, the Berendsen thermostat was applied using a coupling consuming, thus computationally not doable. To find an time of 0. I ps for the bulk water and DMPC. The pressure appropriate pulling velocity, five SMD simulations were was maintained by coupling to a reference pressure of 1 bar. performed using different pulling velocities(0. 1 A ps-,5 x The values of the anisotropic isothermal compressibility were (24)Berendsen, H. J. C. Postma, J. P. M: Vangunsteren, W.F.: Dinola. A: Haak, J. R. Molecular d (22) Fu. W: Shen, J. H: Luo, X. M.: Zhu, w. L: Cheng. J. G: Yu. external bath. J. Chem. Phys. 1984. 81(8). 3684-3690. K. Q; Briggs, J. M. Jin, G. Z; Chen, K. X: Jiang, H. L.(25)Hess, B: Bekker, H: Berendsen, H J C. Fraaije, J. LINCS: A Dopamine DI receptor agonist and D2 receptor antagonist effects linear constraint solver for molecular simulations. J. Comput of the natural product(-)-stepholidine: Molecular Modeling and Chem.1997,l8(12),1463-1472. dynamics Simulations. Biophys J. 2007, 93(5),1431-1441 (26) Darden, T: York, D; Pedersen, L. Particle Mesh Ewald-an (23)Vandrunen, R. Vanderpoel, D. Berendsen, H J C. GROMACS-a N Log(N) Method for Ewald Sums in Large Systems. J. Chem. software package and parallel computer for molecular dynamics Phys.1993,98(12,10089-10092. In Abstracts of Papers, 209th National Meeting of the American (27)Schuettelkopf, A. W. van Aalten, D M. F. PRODRG-a tool for Chemical Society: American Chemical Society: Washington, DC, high-throughput crystallography of protein-ligand complexes. Acta 1995:P49-COMP. Daura, X: Mark, A E; van Gunsteren, w.F. Crystallogr. 2004, D60, 1355-1363 Par arametrization of aliphatic CHn united atoms of GROMOS96 (28)Bode, B. M: Gordon, M. S. MacMolPlt: A graphical user interface force field. J. Comput. Chem. 1998,19(5),535-547 for GAMESS. J. Mol Graphics Modell. 1998. 16(3). 133 1988 MOLECULAR PHARMACEUTICS VOL 7. NO 6

different positions relative to other amino acids in the peptide chain, namely the ortho-position, meta-position and para￾position, the 8 cyclic peptides are stacked in three possible low-energy modes (see Figure S1 in the Supporting Informa￾tion). Therefore, three low-energy stacking models of CPN were constructed. For each CPN, the interior tube diameter is 10 Å, the side chains of L-Trp and the D-Leu are distributed uniformly, and the center-to-center distance between neigh￾boring subunits is 4.75 Å. For convenience, the nanotube subunits are numerically denoted. As displayed in Figure S1 in the Supporting Information, APR (R-plane region) rep￾resents the plane of CR atoms along one peptide subunit, while MPR (midplane region) represents the region between two APRs. The whole length of one CPN is 38.7 Å according to the distance from APR1 to APR8. Conventional Molecular Dynamics Simulation. After each of the three CPNs (i.e., ortho-CPN, meta-CPN and para-CPN) was built, it was inserted into the fully hydrated DMPC bilayer by aligning the axis of CPN to the normal of the lipid bilayer. The process of aligning CPN with the normal of the DMPC membrane was similar to those used in our previous membrane protein simulations.22 When solvating the CPN/DMPC system, 42 Na+ and 42 Cl- ions were added in order to simulate the 150 mM physiological ion strength. The size of the whole solvated system was 77 Å × 83 Å × 110 Å, including one CPN, 189 DMPC molecules and 15087 water molecules. Energy minimizations were performed for each of the three CPN/DMPC/water systems, first for all water molecules, then for the whole system until the maximum force became smaller than 10.00 kcal/mol ·Å. The energy-minimized CPN/ DMPC/water system was then subjected to MD simulation. The MD simulations were performed by using the GROMACS package version 3.3.3 with the GROMOS96 force field.23 The solvent (water and DMPC) molecules of each initial system were equilibrated with CPN structures by constraining the solute (CPNs) at 300 K for 20 ps. Then the CPN was equilibrated for 5 ps while the solvent molecules were constrained at 10, 50, 100, 200, and 298 K. Afterward, each system was equilibrated for 500 ps without any constraints. To maintain the systems at a constant temperature of 300 K, the Berendsen thermostat24 was applied using a coupling time of 0.1 ps for the bulk water and DMPC. The pressure was maintained by coupling to a reference pressure of 1 bar. The values of the anisotropic isothermal compressibility were set to 4.5 × 10-5 , 4.5 × 10-5 , 4.5 × 10-5 , 0, 0, 0 bar-1 for xx, yy, zz, xy/yx, xz/zx and yz/zy components for water and DMPC simulations. All bond lengths, including those to hydrogen atoms, were constrained by the LINCS algorithm.25 Electrostatic interactions between charged groups within 9 Å were calculated explicitly, while long-range electrostatic interactions were calculated using the Particle-Mesh Ewald method26 with a grid width of 1.2 Å and a fourth-order spline interpolation. A cutoff distance of 14 Å was applied for the Lennard-Jones interactions. Numerical integration of the equa￾tions of motion used a time step of 2 fs with atomic coordinates saved every 1 ps for later analysis. Finally, three 10 ns MD simulations were performed on these systems under the periodic boundary conditions in the NPT canonical ensemble. Constant Velocity SMD Simulations. The SMD has proved as an effective computational approach to simulate the transportation process of a small molecule permeating through a protein channel.8 In SMD simulations, a guest molecule or an ion of interest is steered by an imaginary atomic force microscopy (AFM) tip, and the time-dependent external force is added on the guest molecule to facilitate its transportation through the channel. In the present study, we performed SMD simulations in order to explore how the 5-FU molecule is transported by the CPN channel. In detail, 5-FU was pulled through the tube of the CPN by employing an artificial harmonic force on the center of mass (COM) of 5-FU along the longitudinal axis of the CPN (Figure 1). 5-FU molecule was first placed on the top of CPN, 1.27 nm from the center of subunit 1, and then the whole system was equilibrated for 1 ns. The molecular topology file for 5-FU was generated by the PRODRG server (http://davapc1. bioch.dundee.ac.uk/prodrg/).27 The partial atomic charges of 5-FU were determined with the DFT/B3LYP/6-311G** basis set by using the CHelpG method implemented in GAMESS program.28 To avoid large fluctuation in the position, a stiff spring (280 pN·Å) rather than a soft spring was assigned to 5-FU.19 It should be pointed out that the pulling velocity (Vpull) is an important parameter in our SMD simulations. Higher pulling velocity may lead to remarkable nonequilibrium effects, resulting in obvious errors of the simulation results. Very low velocity will make the SMD simulations extremely time￾consuming, thus computationally not doable. To find an appropriate pulling velocity, five SMD simulations were performed using different pulling velocities (0.1 Å· ps-1 , 5 × (22) Fu, W.; Shen, J. H.; Luo, X. M.; Zhu, W. L.; Cheng, J. G.; Yu, K. Q.; Briggs, J. M.; Jin, G. Z.; Chen, K. X.; Jiang, H. L. Dopamine D1 receptor agonist and D2 receptor antagonist effects of the natural product (-)-stepholidine: Molecular Modeling and dynamics Simulations. Biophys. J. 2007, 93 (5), 1431–1441. (23) Vandrunen, R.; Vanderspoel, D.; Berendsen, H. J. C. GROMACSsa software package and parallel computer for molecular dynamics. In Abstracts of Papers, 209th National Meeting of the American Chemical Society; American Chemical Society: Washington, DC; 1995; p 49-COMP. Daura, X.; Mark, A. E.; van Gunsteren, W. F. Parametrization of aliphatic CHn united atoms of GROMOS96 force field. J. Comput. Chem. 1998, 19 (5), 535–547. (24) Berendsen, H. J. C.; Postma, J. P. M.; Vangunsteren, W. F.; Dinola, A.; Haak, J. R. Molecular dynamics with coupling to an external bath. J. Chem. Phys. 1984, 81 (8), 3684–3690. (25) Hess, B.; Bekker, H.; Berendsen, H. J. C.; Fraaije, J. LINCS: A linear constraint solver for molecular simulations. J. Comput. Chem. 1997, 18 (12), 1463–1472. (26) Darden, T.; York, D.; Pedersen, L. Particle Mesh Ewaldsan N.Log(N) Method for Ewald Sums in Large Systems. J. Chem. Phys. 1993, 98 (12), 10089–10092. (27) Schuettelkopf, A. W.; van Aalten, D. M. F. PRODRGsa tool for high-throughput crystallography of protein-ligand complexes. Acta Crystallogr. 2004, D60, 1355–1363. (28) Bode, B. M.; Gordon, M. S. MacMolPlt: A graphical user interface for GAMESS. J. Mol. Graphics Modell. 1998, 16 (3), 133. articles Liu et al. 1988 MOLECULAR PHARMACEUTICS VOL. 7, NO. 6

Molecular Insights on Antitumor Drug 5-Fluorouracil articles Time(min) Figure 2. Transportation profiles of 5-FU mediated by cyclic peptides. The transportation of 5-FU is represented Figure 1. Representative of initial CPN/DMPC/water in terms of the accumulated transport percentage of drugs system for SMD simulation. The front half of the bilayer as a function of time. The concentration of the cyclic and subunits are sliced away to display a view of the inner peptides was 35 uM(; the molar ratio of cyclic peptides wall of the tube, and the tube molecular surface of each to total phosphatidylcholine and cholesterol was 1: 600) subunit is shown in a different color 5-FU is shown as The control group was without cyclic peptides. The stick, which was pulled into the tube along the tube axis results were presented as raw data through a harmonic potential (spring and arrow).The nitrogen and oxygen of the choline and phosphorate are Table 1. The First- order Rate Constant k(min ) and shown as blue and orange balls; the other atoms of the lipid as well as water molecules are represented as sticks. concn of CP (uM) k(min) v(nmol/min) 0.0004 1209±2.06 10-2Aps-1,1×10-2Aps-,5×10-3Aps-1,2.5×10-3 0.0114 0.56±1945 A.-). Our testing results showed that a SMD simulation with the pulling velocity in the range of 2.5 x 10-3A. The concentration of cyclic peptide in its solvent DMF, whose ps to 0. 1 final molar ratio to total phosphatidylcholine and A.- produced a similar force profile. Herein, the trajectories released system is 1: 600 at minimum velocity 2.5 X 10-3A ps- was adopted to illus- (Table 1). The presence of a small amount of DMF has no trate the CPN-mediated transportation mechanism of 5-FU significant effect on 5-FU transportation rate. The observed Results and discussion linear relation between transport rate and 5-FU concentration strongly supports a simple transmembrane channel-mediated Experimentally Observed 5-FU Transportation by transportation process. Moreover, studies on three kinds of CPN Nanotube cyclo[-(Trp-D-Leu) -GIn-D-Leu-]subunits carcinoma cell lines in vitro and the mice inoculated with were synthesized by a two-step solid-phase/solution synthesis S180 solid tumor in vivo demonstrated that the administration strategy with a purity of over 98% and were structurally of cyclic peptide nanotubes efficiently enhanced the antitu characterized by H NMR and mass spectrometry. The mor activity of 5-FU (unpublished data). All of these absorption, fluorescence spectrophotometry and gel perme- experiments demonstrate that the synthetic cyclic peptides ation studies have shown that the synthetic cyclic peptides stack and self-assemble into tubes, followed by their insertion tend to self-assemble and diffuse into lipid bilayers due to into the liposome membrane and transportation of antitumor the hydrophobic interactions of their side chains with the drug 5-FU across the liposome membrane hydrophobic chains of lipid when it is distributed into The ortho.cpn Is the most Stable self-Assembled aqueous suspension of liposomes. Our dialysis experiments anotube. The conventional CMd simulations for 3 model test such an assembling and insertion process. The release systems were conducted to investigate the structural and percentage of 5-FU from drug-loaded liposomes after adding dynamical properties of synthesized CPN. The 10 ns CMD cyclic peptides is shown in Figure 2. It can be seen that only simulations showed that ortho-CPN, meta-CPN and para 5% of 5-FU diffused from liposomes without cyclic peptides. CPN behaved differently in explicitly hydrolyzed DMPC In contrast, nearly 70%o of 5-FU was released into the solution bilayer. As shown in Figure S2 in the Supporting Informa when the concentration of cyclic peptide increased to 2 mg/ tion, meta-CPN sustained the hollow structure at the begin mL. A similar profile was found for the diffusion of glucose ning, but it began to bend at 2.6 ns and kept the curving across the CPNs. Similarly, the first-order transport rate posture in the rest of simulation For para-CPN, the tube constant k and average transport rate v in 90 min were much started to collapse during the first 1 ns simulation and higher than that of the control group(without cyclic peptides) completely collapsed at the end, demonstrating para-CPN VOL 7. NO 6 MOLECULAR PHARMACEUTICS 1989

10-2 Å· ps-1 , 1 × 10-2 Å· ps-1 , 5 × 10-3 Å· ps-1 , 2.5 × 10-3 Å·ps-1 ). Our testing results showed that a SMD simulation with the pulling velocity in the range of 2.5 × 10-3 Å· ps to 0.1 Å· ps-1 produced a similar force profile. Herein, the trajectories at minimum velocity 2.5 × 10-3 Å · ps-1 was adopted to illus￾trate the CPN-mediated transportation mechanism of 5-FU. Results and Discussion Experimentally Observed 5-FU Transportation by CPN Nanotube. cyclo[-(Trp-D-Leu)4-Gln-D-Leu-] subunits were synthesized by a two-step solid-phase/solution synthesis strategy with a purity of over 98% and were structurally characterized by 1 H NMR and mass spectrometry.20 The absorption, fluorescence spectrophotometry and gel perme￾ation studies6 have shown that the synthetic cyclic peptides tend to self-assemble and diffuse into lipid bilayers due to the hydrophobic interactions of their side chains with the hydrophobic chains of lipid when it is distributed into aqueous suspension of liposomes. Our dialysis experiments test such an assembling and insertion process. The release percentage of 5-FU from drug-loaded liposomes after adding cyclic peptides is shown in Figure 2. It can be seen that only 5% of 5-FU diffused from liposomes without cyclic peptides. In contrast, nearly 70% of 5-FU was released into the solution when the concentration of cyclic peptide increased to 2 mg/ mL. A similar profile was found for the diffusion of glucose across the CPNs.15 Similarly, the first-order transport rate constant k and average transport rate V in 90 min were much higher than that of the control group (without cyclic peptides) (Table 1). The presence of a small amount of DMF has no significant effect on 5-FU transportation rate. The observed linear relation between transport rate and 5-FU concentration strongly supports a simple transmembrane channel-mediated transportation process. Moreover, studies on three kinds of carcinoma cell lines in Vitro and the mice inoculated with S180 solid tumor in ViVo demonstrated that the administration of cyclic peptide nanotubes efficiently enhanced the antitu￾mor activity of 5-FU (unpublished data). All of these experiments demonstrate that the synthetic cyclic peptides stack and self-assemble into tubes, followed by their insertion into the liposome membrane and transportation of antitumor drug 5-FU across the liposome membrane. The ortho-CPN Is the Most Stable Self-Assembled Nanotube. The conventional CMD simulations for 3 model systems were conducted to investigate the structural and dynamical properties of synthesized CPN. The 10 ns CMD simulations showed that ortho-CPN, meta-CPN and para￾CPN behaved differently in explicitly hydrolyzed DMPC bilayer. As shown in Figure S2 in the Supporting Informa￾tion, meta-CPN sustained the hollow structure at the begin￾ning, but it began to bend at ∼2.6 ns and kept the curving posture in the rest of simulation. For para-CPN, the tube started to collapse during the first 1 ns simulation and completely collapsed at the end, demonstrating para-CPN Figure 1. Representative of initial CPN/DMPC/water system for SMD simulation. The front half of the bilayer and subunits are sliced away to display a view of the inner wall of the tube, and the tube molecular surface of each subunit is shown in a different color. 5-FU is shown as stick, which was pulled into the tube along the tube axis through a harmonic potential (spring and arrow). The nitrogen and oxygen of the choline and phosphorate are shown as blue and orange balls; the other atoms of the lipid as well as water molecules are represented as sticks. Figure 2. Transportation profiles of 5-FU mediated by cyclic peptides. The transportation of 5-FU is represented in terms of the accumulated transport percentage of drugs as a function of time. The concentration of the cyclic peptides was 35 µM ([; the molar ratio of cyclic peptides to total phosphatidylcholine and cholesterol was 1:600). The control group (9) was without cyclic peptides. The results were presented as raw data. Table 1. The First-Order Rate Constant k (min-1) and Average Transport Rate v (nmol/min) of 5-FU in 90 min concn of CP (µM) k (min-1) v (nmol/min) 0 0.0004 12.09 ( 2.06 35a 0.0114 140.56 ( 19.45 a The concentration of cyclic peptide in its solvent DMF, whose final molar ratio to total phosphatidylcholine and cholesterol in the released system is 1:600. Molecular Insights on Antitumor Drug 5-Fluorouracil articles VOL. 7, NO. 6 MOLECULAR PHARMACEUTICS 1989

articles 目盟8品 Time(ps) Figure 3. (A)The time-evolutionary radius of gyration(Rg, a measure of the compactness of the protein) and tilt- with respect to the normal of bilayer for the self-assembled ortho-CPN.(B) Conformational transition of the ortho within the course of the simulatio was quite unstable When checking the structures from CMD tion. Interestingly, the side chains of the eight intersubunit simulations, we found that the internal hydrogen bonding peptides rearranged their orientations and have led to the network(see Figure S3 in the Supporting Information) of formation of a new hydrogen bond network in consecutive para-CPN collapsed finally. The ortho-CPN retains a hollow rings, particularly between L-Trp and L-Gln or between L-Gln tubular structure with only slight distortion at the outer end and L-Gln. This suggests that Gln residue helps to stabilize and has tilted ca 50 compared with its starting orientation. the tubular arrays, which is in accord with the findings in a Such an extent of tilt is close to the reported tilt angle of previously reported study. 30 39 for cyclo[(L-Trp-D-Leu)3-L-GIn-D-Leu] by ATR-IR spec Water Molecules inside the ortho- CPn Nanotube troscopy measurements. Slight structural distortion was also During the CMD simulations, water molecules increasingly reported at the outer end of the octapeptide tube. Based diffused into the hollow cylindrical tube and flowed smoothly on our CMD simulations, the ortho-CPn was selected as through the whole tube. The average number of water the starting structure for our SMD simulations, and further molecules inside the ortho-CPn was 38 +5. As shown in structural analysis was performed only for this nanotube Figure 4, there are about 4-5 water molecules in each MPR Along the course of CMD simulation, the average inter- layer and 1-2 water molecules at each APR. As reported, 6 18 unit distance for ortho-CPN was within the range of 4.73 octapeptide nanotube could hold only 20 water molecules A to 4.79 A. Such an average distance is in good agreement with a typical"1-2 "water structure (i.e,APR-1; MPR-2) with the reported distance(4.73 A)from Fourier-transform inside the tube. Such a difference in the number of water infrared spectroscopy, electron diffraction and X-ray crystal molecules mainly comes from the different size of the lographic analyses on cyclo[-(GIn-D-Ala-Glu-D-Ala)4) nanotube. The diameter of our ortho-CPN is 10 A,much CPN. The backbone Rg profile(Figure 3A)also supports larger than that of the octapeptide tube. The MPR water the structural stability of ortho-CPN. A further look at the number is bigger than that of APR, because the water napshots of CMd on ortho-CPN(Figure 3B)showed that molecules at MPR formed hydrogen bonds with the it began to tilt at 1 ns, causing a slight kink in R7 at 1. 4 ns. hydrophilic C=O and N-H backbone of the tube wall. The tilt angle reached 45 at 4.2 ns. The extent of such a The calculated diffusion coefficient value for water tilt kept within the range of 45-50 at the rest of the CMd molecules inside the ortho-CPN is 1. x 10cm"s simulations(Figure 3). As observed, the subunits R7 and almost half of the diffusion coefficient value of bulky r8 tilted more than the other subunits 1 to 6 Previous study demonstrated that the backbone hydrogen (29) Jingchuan, Z: Jie, C: Zhouxiong, L. Zhonghong, L: Bo, L. bond network formed by C-O and N-H groups between Investigation of structures and properties of cyclic peptide nanotubes by experiment and molecular dynamics. J. Compi different subunits are essential for the structural stability of Aided Mo.Des.2008,22,773-781 the tube. Such a hydrogen bond network in our ortho-CPN (30) Jillian, M: Buriak, M: Reza, G Self-assembly of peptide based was relatively persistent during the course of CMD simula nanotubes. Mater. Sci. Eng: C 1997, 4, 207-212. 1990 MOLECULAR PHARMACEUTICS VOL 7. NO 6

was quite unstable. When checking the structures from CMD simulations, we found that the internal hydrogen bonding network (see Figure S3 in the Supporting Information) of para-CPN collapsed finally. The ortho-CPN retains a hollow tubular structure with only slight distortion at the outer end and has tilted ca. 50° compared with its starting orientation. Such an extent of tilt is close to the reported tilt angle of 39° for cyclo[(L-Trp-D-Leu)3-L-Gln-D-Leu] by ATR-IR spec￾troscopy measurements.18 Slight structural distortion was also reported at the outer end of the octapeptide tube.18 Based on our CMD simulations, the ortho-CPN was selected as the starting structure for our SMD simulations, and further structural analysis was performed only for this nanotube. Along the course of CMD simulation, the average inter￾subunit distance for ortho-CPN was within the range of 4.73 Å to 4.79 Å. Such an average distance is in good agreement with the reported distance (4.73 Å) from Fourier-transform infrared spectroscopy, electron diffraction and X-ray crystal￾lographic analyses on cyclo[-(Gln-D-Ala-Glu-D-Ala)4] CPN.5 The backbone Rg profile (Figure 3A) also supports the structural stability of ortho-CPN. A further look at the snapshots of CMD on ortho-CPN (Figure 3B) showed that it began to tilt at 1 ns, causing a slight kink in R7 at 1.4 ns. The tilt angle reached 45° at ∼4.2 ns. The extent of such a tilt kept within the range of 45-50° at the rest of the CMD simulations (Figure 3). As observed, the subunits R7 and R8 tilted more than the other subunits 1 to 6. Previous study demonstrated that the backbone hydrogen bond network formed by CdO and N-H groups between different subunits are essential for the structural stability of the tube.29 Such a hydrogen bond network in our ortho-CPN was relatively persistent during the course of CMD simula￾tion. Interestingly, the side chains of the eight intersubunit peptides rearranged their orientations and have led to the formation of a new hydrogen bond network in consecutive rings, particularly between L-Trp and L-Gln or between L-Gln and L-Gln. This suggests that Gln residue helps to stabilize the tubular arrays, which is in accord with the findings in a previously reported study.30 Water Molecules inside the ortho-CPN Nanotube. During the CMD simulations, water molecules increasingly diffused into the hollow cylindrical tube and flowed smoothly through the whole tube. The average number of water molecules inside the ortho-CPN was 38 ( 5. As shown in Figure 4, there are about 4-5 water molecules in each MPR layer and 1-2 water molecules at each APR. As reported,16,18 octapeptide nanotube could hold only ∼20 water molecules with a typical “1-2” water structure (i.e., APR-1; MPR-2) inside the tube. Such a difference in the number of water molecules mainly comes from the different size of the nanotube. The diameter of our ortho-CPN is 10 Å, much larger than that of the octapeptide tube. The MPR water number is bigger than that of APR, because the water molecules at MPR formed hydrogen bonds with the hydrophilic CdO and N-H backbone of the tube wall. The calculated diffusion coefficient value for water molecules inside the ortho-CPN is 1.068 × 10-5 cm2 ·s-1 , almost half of the diffusion coefficient value of bulky (29) Jingchuan, Z.; Jie, C.; Zhouxiong, L.; Zhonghong, L.; Bo, L. Investigation of structures and properties of cyclic peptide nanotubes by experiment and molecular dynamics. J. Comput.- Aided Mol. Des. 2008, 22, 773–781. (30) Jillian, M.; Buriak, M.; Reza, G. Self-assembly of peptide based nanotubes. Mater. Sci. Eng.: C 1997, 4, 207–212. Figure 3. (A) The time-evolutionary radius of gyration (Rg, a measure of the compactness of the protein) and tilt-angle with respect to the normal of bilayer for the self-assembled ortho-CPN. (B) Conformational transition of the ortho-CPN within the course of the simulation. articles Liu et al. 1990 MOLECULAR PHARMACEUTICS VOL. 7, NO. 6

Molecular Insights on Antitumor Drug 5-Fluorouracil articles atoms of the phosphate groups and the indole Nh group of L-Trp or with the amide group of L-Gln; and those formed by the carbonyl group of the lipid with the L-Trp and L-Gln residues. The anchoring interactions of the lipid bilayer molecules with L-Gln and L-Trp residues may contribute to the tilt of the tube. The end subunit of the ortho-cPn is anchored via a hydrogen bond between the oxygen of phosphate or carbonyl group and the Trp or GIn. The number of hydrogen bonds between DMPC and R& is bigger than that between DMPC and Rl(Figure 5). This could be the structural determinant for the observed slight distortion of subunits R7 and R8. These interactions result in the slight distortion in the subunits r7 and r8. overall. the mode of hydrogen bonding interactions between the ortho-CPN subunits and the surrounding lipid bilayer molecules are similar to that reported for the gramicidin channel and octapeptide tube embedded in a lipid membrane The Transportation Mechanism of 5-FU across ortho-CPN. As depicted in Figure 6A, the pulling force was first averaged around zero at the beginning of SMD simula- tions. This force increased and formed different local peak when 5-FU passed through each subunit of ortho-CPN. The force reached the maximum when 5-FU was passing througl Figure 4. The structure of the internal diffusing water the subunit R7 and R& near the outer end Correspondingly. molecules in the ortho- cPn nanotubes the distance between the center of mass(COM) of 5-FU and the geometry center of the tube featured an eight ladder water molecules(calculated as 2.181 x 10-5 cm2.s-I and decrease landscape when 5-FU passed through each subuni experimentally determined as 2.3x 105cm2.s- at T= Finally, 5-FU passed through the entire tube, and the pulling 298 K). As a water channel, the calculated water force decreased sharply to near zero. The"sawtoothed" diffusion coefficient value was reported as 1.068 10 pulling force profiles and"ladder"style walking of 5-FU cm2.s, and the calculated water diffusion coefficient ( Figure 6A)revealed that 5-FU spanned each APR in a way value for cyclo[(Trp-D-Leu)3-Gln-D-Leu] channel was only of"hopping"through the whole tube. The pulling force 0.44 x 10-cm.s. The large water diffusion coefficient increased when the aromatic plane of 5-FU became perpen- value suggests that water molecules diffuse very easily dicular to the hydrophobic APR as there was a molecular through our ortho-CPN channel friction among 5-FU, water and the hydrophobic APR. The ortho-CPN Interactions with Surrounding Lipid pulling force decreased when 5-FU went through the neighbor Bilayer. The ortho-CPN interacts with the surrounding lipid ing MPR as hydrogen bonds were formed among 5-FU, the bilayer through two kinds of contacts. At the middle region inner wall of the nanotube and the diffusing water molecules. f the lipid bilayer, they interact with each other by Such rise-and-fall of the pulling force appeared repeatedly as hydrophobic contacts. At the two ends of the lipid bilayer, the 5-FU went through different APRs and MPRs, making the hydrogen bonding interactions were found. Analysis of the 5-FU hopping along the axis of the ortho-CPN 10 ns MD trajectory revealed two types of long-lived Figure 6B shows several critical kinds of interactions ydrogen bonds, viz., those formed between the oxygen between 5-FU and the ortho-CPN. The direct hydrogen bond O RI DMP o R8 DMP 2000 8000 800010000 Time(ps) Time(ps) Figure 5. Time-evolutionary hydrogen bonding between ortho-CPN and the lipid bilayer VOL 7. NO 6 MOLECULAR PHARMACEUTICS 199

water molecules (calculated as 2.181 × 10-5 cm2 ·s-1 and experimentally determined as 2.3 × 10-5 cm2 ·s-1 at T ) 298 K).31 As a water channel, the calculated water diffusion coefficient value was reported as 1.068 × 10-5 cm2 ·s-1 , and the calculated water diffusion coefficient value for cyclo[(Trp-D-Leu)3-Gln-D-Leu] channel was only 0.44 × 10-5 cm2 ·s-1 . The large water diffusion coefficient value suggests that water molecules diffuse very easily through our ortho-CPN channel. ortho-CPN Interactions with Surrounding Lipid Bilayer. The ortho-CPN interacts with the surrounding lipid bilayer through two kinds of contacts. At the middle region of the lipid bilayer, they interact with each other by hydrophobic contacts. At the two ends of the lipid bilayer, hydrogen bonding interactions were found. Analysis of the 10 ns MD trajectory revealed two types of long-lived hydrogen bonds, viz., those formed between the oxygen atoms of the phosphate groups and the indole NH group of L-Trp or with the amide group of L-Gln; and those formed by the carbonyl group of the lipid with the L-Trp and L-Gln residues. The anchoring interactions of the lipid bilayer molecules with L-Gln and L-Trp residues may contribute to the tilt of the tube. The end subunit of the ortho-CPN is anchored via a hydrogen bond between the oxygen of phosphate or carbonyl group and the Trp or Gln. The number of hydrogen bonds between DMPC and R8 is bigger than that between DMPC and R1 (Figure 5). This could be the structural determinant for the observed slight distortion of subunits R7 and R8. These interactions result in the slight distortion in the subunits R7 and R8. Overall, the mode of hydrogen bonding interactions between the ortho-CPN subunits and the surrounding lipid bilayer molecules are similar to that reported for the gramicidin channel and octapeptide tube embedded in a lipid membrane.18,32 The Transportation Mechanism of 5-FU across ortho-CPN. As depicted in Figure 6A, the pulling force was first averaged around zero at the beginning of SMD simula￾tions. This force increased and formed different local peaks when 5-FU passed through each subunit of ortho-CPN. The force reached the maximum when 5-FU was passing through the subunit R7 and R8 near the outer end. Correspondingly, the distance between the center of mass (COM) of 5-FU and the geometry center of the tube featured an eight ladder decrease landscape when 5-FU passed through each subunit. Finally, 5-FU passed through the entire tube, and the pulling force decreased sharply to near zero. The “sawtoothed” pulling force profiles and “ladder” style walking of 5-FU (Figure 6A) revealed that 5-FU spanned each APR in a way of “hopping” through the whole tube. The pulling force increased when the aromatic plane of 5-FU became perpen￾dicular to the hydrophobic APR as there was a molecular friction among 5-FU, water and the hydrophobic APR. The pulling force decreased when 5-FU went through the neighbor￾ing MPR as hydrogen bonds were formed among 5-FU, the inner wall of the nanotube and the diffusing water molecules. Such rise-and-fall of the pulling force appeared repeatedly as the 5-FU went through different APRs and MPRs, making the 5-FU hopping along the axis of the ortho-CPN. Figure 6B shows several critical kinds of interactions between 5-FU and the ortho-CPN. The direct hydrogen bond Figure 4. The structure of the internal diffusing water molecules in the ortho-CPN nanotubes. Figure 5. Time-evolutionary hydrogen bonding between ortho-CPN and the lipid bilayer. Molecular Insights on Antitumor Drug 5-Fluorouracil articles VOL. 7, NO. 6 MOLECULAR PHARMACEUTICS 1991

articles R4 RS R6 RT R in APR, showing an intrinsic driving force for the hopping of 5-FU. The number of DHB is about 2-3 on average when 5-FU walked from subunit 2 to subunit 6. The number of DHB increased to 3-4 when 5-FU went through the mPr between subunit 6 and subunit 7. The 5-fu dwelled 2 ns at this kink region of ortho-cpn before it went to the final steps across subunit 7 and subunit 8. The number of DhB dramatically increased to 6 when 5-FU was passing through the kink structure around subunits 7 and 8. Meanwhile the number of WB strikingly changed from 2 to 6 when 5-FU dwelled in the region of subunits 7 and 8. There is no obvious difference between the kink region and rest of the tube. The above analyses fully illuminate the importance of the interaction between 5-FU, diffusing inner water and the inner wall of tube in the transport process of 5-FU Figure 7A represents typical snapshots from SMD trajectory for 5-FU hopping through the ortho-CPN, and Figure S4 in R4 R the Supporting Information shows the tracked changes of angle between the 5-FU plane and the axis of the nanotube. These results confirm the perpendicular jump of 5-FU in each APR M as well as the reorientation of 5-Fu in each MPR. Such a LM hopping mode of 5-FU was also confirmed by the potential energy profile for 5-FU moving through the ortho-CPN(Figur 7B). The characteristics of the hopping mode of 5-FU as shown in Figure 7 are consistent with the mode of changes for the alling force and for the critical interactions among 5-Fu, diffusing waters and the interior wall of the nanotube(Figure As displayed in Figure 6A, the maximum force pulling 5-FU through the tube ranged from 250 to 550 pN before 5-FU entered the region of the last two subunits(subunit 7 and subunit 8). The pulling force dramatically increased to 1250 pN and formed two peaks when 5-FU arrived at subunit 7 and subunit 8. As discussed above. the nanotube formed a Figure 6.(A) The pulling force profile and distance minor kink at subunit 7 and subunit 8, but the artificial feature between the center of mass of 5-FU and the pulling force did not change its direction at the kink region hydrogen bond(DHB), hydrophobic interactions(HI) artificial force was needed to help the 5-FU molecule escaper and water bridges(WB)between 5-FU and ortho-CPN from the trap. Several questions could be raised as to how along the whole SMD simulations. Note: A hydrogen the 5-FU escaped from the kink structure of the nanotube bond between 5-FU and CPN(D-H-.)is defined if For example, how did the pulling force affectthe 5-FU the intermolecular distances are Ro-As3 3 A, RH-As moving trajectory Did the kink structure in the end subunit 90°, and the h…A- AA angle is≥90°, where AA is the affect the translocation rate of 5-FU? Was the energy well tom attached to the acceptor; the van der Waals located at the kink region? To answer these questions, the contact of nonpolar atoms(Rc-C s 4.0 A) was used to binding free energy profiles of 5-FU with ortho-CPN along define the hydrophobic interaction; the water bridge with SMD simulations were calculated using the Au- exists if several water molecules could form a hydrogen toDock 4.0 scoring function. The calculated results are shown bond network with 5-FU and CPN (31) Stout, D. G; Steponkus, P. L: Bustard, L. D; Cotts, R. M.Water (DHB), hydrophobic interactions(HI) and water bridges permeability of Chlorella cell membranes by nuclear magnetic WB)between 5-FU and ortho-CPn were tracked along the resonance measured diffusion coefficients and relaxation times whole SMD simulations. When moving across each subunit Plant Physiol.1978,62(1).146-15 of ortho-cPN. the interaction between 5-FU and ortho-CPN (32)Woolf, T.B. Roux, B. Molecular dynamics simulation of the changed from one kind to another, including alternative old Gramicidin channel in a phospholipid bilayer. Proc. Natl. Sci. U.S.A. 1994, 91(24), 11631-11635 Roux. B Computa hydrogen bond breaking and new hydrogen bond formation. studies of the gramicidin channel. Acc. Chem. Res. 2002 The number of DHB in MPR is generally bigger than that 366-375 992 MOLECULAR PHARMACEUTICS VOL 7. NO. 6

(DHB), hydrophobic interactions (HI) and water bridges (WB) between 5-FU and ortho-CPN were tracked along the whole SMD simulations. When moving across each subunit of ortho-CPN, the interaction between 5-FU and ortho-CPN changed from one kind to another, including alternative old hydrogen bond breaking and new hydrogen bond formation. The number of DHB in MPR is generally bigger than that in APR, showing an intrinsic driving force for the hopping of 5-FU. The number of DHB is about 2-3 on average when 5-FU walked from subunit 2 to subunit 6. The number of DHB increased to 3-4 when 5-FU went through the MPR between subunit 6 and subunit 7. The 5-FU dwelled 2 ns at this kink region of ortho-CPN before it went to the final steps across subunit 7 and subunit 8. The number of DHB dramatically increased to 6 when 5-FU was passing through the kink structure around subunits 7 and 8. Meanwhile, the number of WB strikingly changed from 2 to 6 when 5-FU dwelled in the region of subunits 7 and 8. There is no obvious difference between the kink region and rest of the tube. The above analyses fully illuminate the importance of the interaction between 5-FU, diffusing inner water and the inner wall of tube in the transport process of 5-FU. Figure 7A represents typical snapshorts from SMD trajectory for 5-FU hopping through the ortho-CPN, and Figure S4 in the Supporting Information shows the tracked changes of angle between the 5-FU plane and the axis of the nanotube. These results confirm the perpendicular jump of 5-FU in each APR as well as the reorientation of 5-FU in each MPR. Such a hopping mode of 5-FU was also confirmed by the potential energy profile for 5-FU moving through the ortho-CPN (Figure 7B). The characteristics of the hopping mode of 5-FU as shown in Figure 7 are consistent with the mode of changes for the pulling force and for the critical interactions among 5-FU, diffusing waters and the interior wall of the nanotube (Figure 6). As displayed in Figure 6A, the maximum force pulling 5-FU through the tube ranged from 250 to 550 pN before 5-FU entered the region of the last two subunits (subunit 7 and subunit 8). The pulling force dramatically increased to 1250 pN and formed two peaks when 5-FU arrived at subunit 7 and subunit 8. As discussed above, the nanotube formed a minor kink at subunit 7 and subunit 8, but the artificial pulling force did not change its direction at the kink region. As a result, 5-FU was trapped in the kink and a bigger artificial force was needed to help the 5-FU molecule escape from the trap. Several questions could be raised as to how the 5-FU escaped from the kink structure of the nanotube. For example, how did the pulling force affectthe 5-FU moving trajectory? Did the kink structure in the end subunit affect the translocation rate of 5-FU? Was the energy well located at the kink region? To answer these questions, the binding free energy profiles of 5-FU with ortho-CPN along with SMD simulations were calculated using the Au￾toDock4.0 scoring function. The calculated results are shown (31) Stout, D. G.; Steponkus, P. L.; Bustard, L. D.; Cotts, R. M. Water permeability of Chlorella cell membranes by nuclear magnetic resonance: Measured diffusion coefficients and relaxation times. Plant Physiol. 1978, 62 (1), 146–151. (32) Woolf, T. B.; Roux, B. Molecular dynamics simulation of the Gramicidin channel in a phospholipid bilayer. Proc. Natl. Acad. Sci. U.S.A. 1994, 91 (24), 11631–11635. Roux, B. Computational studies of the gramicidin channel. Acc. Chem. Res. 2002, 35 (6), 366–375. Figure 6. (A) The pulling force profile and distance feature between the center of mass of 5-FU and the geometry center of the nanotube. (B) The direct hydrogen bond (DHB), hydrophobic interactions (HI) and water bridges (WB) between 5-FU and ortho-CPN along the whole SMD simulations. Note: A hydrogen bond between 5-FU and CPN (D-H ···A) is defined if the intermolecular distances are RD-A e 3.3 Å, RH-A e 2.6 Å, and the donor-acceptor angle is D-H ···A g 90°, and the H ···A-AA angle is g90°, where AA is the atom attached to the acceptor; the van der Waals contact of nonpolar atoms (RC-C e 4.0 Å) was used to define the hydrophobic interaction; the water bridge exists if several water molecules could form a hydrogen bond network with 5-FU and CPN. articles Liu et al. 1992 MOLECULAR PHARMACEUTICS VOL. 7, NO. 6

Molecular Insights on Antitumor Drug 5-Fluorouracil articles APr-3 MPR-1 APR Energy well 1》(刺肌游)骑 Figure 7(A) The representative snapshots of 5-Fu going through the nanotube along the whole SMD simulations. ( B) The potential energy profile for 5-FU transported through the nanotube by SMD simulation in Figure 8. Corresponding to the peak of pulling force at apparently"single-file"inner water structure. However, the kink region, the calculated binding free energy based on Ghadiri et al. indicated that the alternating 1-2 water the reaction coordinate became as low as-4.9 kcal/mol. structure (i.e, APR-1; MPR-2)inside the octapeptide channel Obviously, there must be an energy well at this kink region may enhance the water diffusion Based on our experimental of the ortho-CPn (indicated in Figure 7A). The existence observations and the results of SMD simulations as described f such an energy well at the kink region certainly slowed down the transporting of 5-FU across the nanotube. Other (33)Roux, B. Prodhom, B: Karplus, M Ion transport studies also suggested that structural kinking significantl hannel-molecular dynamics study le oc- delays water diffusion in the gramicidin channel which has cupancy. Biophys J. 1995, 68(3),876-892 VOL 7. NO 6 MOLECULAR PHARMACEUTICS 199

in Figure 8. Corresponding to the peak of pulling force at the kink region, the calculated binding free energy based on the reaction coordinate became as low as -4.9 kcal/mol. Obviously, there must be an energy well at this kink region of the ortho-CPN (indicated in Figure 7A). The existence of such an energy well at the kink region certainly slowed down the transporting of 5-FU across the nanotube. Other studies also suggested that structural kinking significantly delays water diffusion in the gramicidin channel which has apparently “single-file” inner water structure.33 However, Ghadiri et al.16 indicated that the alternating 1-2 water structure (i.e., APR-1; MPR-2) inside the octapeptide channel may enhance the water diffusion. Based on our experimental observations and the results of SMD simulations as described (33) Roux, B.; Prodhom, B.; Karplus, M. Ion transport in the gramicidin channelsmolecular dynamics study of single and double oc￾cupancy. Biophys. J. 1995, 68 (3), 876–892. Figure 7. (A) The representative snapshots of 5-FU going through the nanotube along the whole SMD simulations. (B) The potential energy profile for 5-FU transported through the nanotube by SMD simulations. Molecular Insights on Antitumor Drug 5-Fluorouracil articles VOL. 7, NO. 6 MOLECULAR PHARMACEUTICS 1993

articles 3R4 1500 and molecular mechanism of 5-FU transportation by the nanotube by CMD simulations and SMD simulations. Detailed analyses of the whole CMD trajectory revealed that the B-sheetlike cylindrical tube conserved its hollow tubular structure by means of intersubunit hydrogen bond network between the carbonyl and amide groups on the backbone of the tube. The CMD simulation demonstrated that cPn tilted about 50 with respect to the normal of the bilayer. The value of this tilting angle is close to the reported tilt angle of 39o for octapeptide nanotube by ATR-IR spectroscopy measurements. The robust hydrogen bond network formed by the carbonyl and amide group of the backbone in CPN, the inner diffusing water and the oxygen of the phosphate on surrounding lipid contributed to the stability and tilt of the tube. On average Reaction Coordinate(nm) 38 diffusing water molecules were found in the tube and Figure 8. The pulling force and calculated binding free most of them preferred to reside in the middle zone (MPr) energy profile for 5-FU interacting with ortho-CPN of neighboring cyclic peptide subunits. Analysis of the motion above, the ortho-CPN is able to transport water molecules and of diffusing water molecules in the tube gives a diffusion con- small drug molecules, and the transporting process should be stant of 1. x 10 cm2.s- This value of diffusion coefficient is approximately half of the value of self-diffusion much faster than that of the gramicidin channel or octapeptide coefficient of bulk water(2.3x 10-5.s-), 2.4 times the channel. The slower walking process of 5-FU at the kink region as well as the re\-ssvery arge"wiggle"space of the tube(10A value for octapeptide tube, and 42 times the value of water in the diameter)at this local region enabled more water molecules diffusing inside the gramicidin A channel. Such a molecules surrounding the polar 5-FU(Figure 4). Because yrge value of diffusion coefficient indicates a bulk-like behavior the dynamic nature of water molecules(Figure 4), the entropic of the inner diffusing water effects may help the 5-FU molecule escape from the kink region SMD simulation was conducted to uncover the transport and move further along the nanotube process of 5-FU across the lipid bilayer. Based on our Ghadiri et al. tested the glucose transport of the cyclo- simulation results and analyses, we proposed a molecular octapeptide tube by glucose-entrapped unilamellar lipid mechanism for the transport of 5-FU across CPN. During vesicle studies, but the molecular mechanism of glucose the transporting process, 5-FU hopped over each APR and ansporting has not been well addressed. The report resided in each MPR by switching from hydrophobic transporting mechanism of Na and K along the octapeptide interactions with the interior wall of the nanotube to hydrogen channel is quite different from the hopping process for 5-Fu bonding interactions with carbonyl group and amide group transported by ortho-CPN. The slower transporting of Na of each MPR. The characteristics of the pulling force profile and K along the octapeptide channel was attributed to the of SMD simulations and the potential energy profile of the electrostatic interactions of Na or k with the negatively ortho-CPN confirmed the hopping style of 5-FU transporting harged carbonyl at the wall of the octapeptide nanotube In Finally, 5-FU was transported to the outer end of the addition, many more water molecules were found surrounding nanotube by escaping from the energy well located around the Na and k along the octapeptide channel, while the 5-FU the kink region at subunits 7 and 8 of ortho-CPN. Both our molecule formed a hydrogen bonding network with the interior experimental and computational results demonstrated that wall of the MPR region of ortho-CPN in the present stud ortho-CPN is an ideal transporter for the delivery of Conclusion antitumor drug 5-fu to tumor cells In this work, we synthesized cyclo[(Trp-D-Leu)4-GIn-D- Acknowledgment. The authors gratefully acknowledge Leul peptide subunits. The studies on drug loaded liposomes Dr. M. R. Ghadiri for providing us the initial molecular showed that 5-FU could quickly be transported across the model of the cyclic peptide nanotube and Dr. Weihua Li for lipid bilayers after adding cyclic peptides into the aqueous his helpful discussions upon SMD simulation. This work is suspensions of large unilamellar liposomes. The result combined financially supported by the National Science Foundation of Investigations confirmed China(NsFc)(no. 30672546, 20702009)and partly by the of 5-FU was facilitated by ortho-CPN in a lipid environment. National Key Basic Research Program of China(No Computational studies have brought insights on the 2007CB935800, 2009CB930300) structural and dynamical characteristics of the synthetic CPN, Supporting Information Available: Figures Sl-S4 as discussed in the text. This material is available free of charge (34)asthagiri,D.;Bashford,D.ContinuumandatomisticmodelingviatheInternetathttp://pubs.acs.org (3),1176-1189 MP100274F 1994 MOLECULAR PHARMACEUTICS VOL 7. NO 6

above, the ortho-CPN is able to transport water molecules and small drug molecules, and the transporting process should be much faster than that of the gramicidin channel or octapeptide channel. The slower walking process of 5-FU at the kink region as well as the relatively large “wiggle” space of the tube (10 Å in the diameter) at this local region enabled more water molecules surrounding the polar 5-FU (Figure 4). Because of the dynamic nature of water molecules (Figure 4), the entropic effects may help the 5-FU molecule escape from the kink region and move further along the nanotube Ghadiri et al.15 tested the glucose transport of the cyclo￾octapeptide tube by glucose-entrapped unilamellar lipid vesicle studies, but the molecular mechanism of glucose transporting has not been well addressed. The reported transporting mechanism of Na+ and K+ along the octapeptide channel is quite different from the hopping process for 5-FU transported by ortho-CPN.6,8 The slower transporting of Na+ and K+ 8 along the octapeptide channel was attributed to the electrostatic interactions of Na+ or K+ with the negatively charged carbonyl at the wall of the octapeptide nanotube. In addition, many more water molecules were found surrounding the Na+ and K+ along the octapeptide channel, while the 5-FU molecule formed a hydrogen bonding network with the interior wall of the MPR region of ortho-CPN in the present study.34 Conclusion In this work, we synthesized cyclo[(Trp-D-Leu)4-Gln-D￾Leu] peptide subunits. The studies on drug loaded liposomes showed that 5-FU could quickly be transported across the lipid bilayers after adding cyclic peptides into the aqueous suspensions of large unilamellar liposomes. The result combined with earlier investigations confirmed that the actual transport of 5-FU was facilitated by ortho-CPN in a lipid environment. Computational studies have brought insights on the structural and dynamical characteristics of the synthetic CPN, and molecular mechanism of 5-FU transportation by the nanotube by CMD simulations and SMD simulations. Detailed analyses of the whole CMD trajectory revealed that the -sheetlike cylindrical tube conserved its hollow tubular structure by means of intersubunit hydrogen bond network between the carbonyl and amide groups on the backbone of the tube. The CMD simulation demonstrated that CPN tilted about 50° with respect to the normal of the bilayer. The value of this tilting angle is close to the reported tilt angle of 39° for octapeptide nanotube by ATR-IR spectroscopy measurements. The robust hydrogen bond network formed by the carbonyl and amide group of the backbone in CPN, the inner diffusing water and the oxygen of the phosphate on surrounding lipid contributed to the stability and tilt of the tube. On average, 38 diffusing water molecules were found in the tube and most of them preferred to reside in the middle zone (MPR) of neighboring cyclic peptide subunits. Analysis of the motion of diffusing water molecules in the tube gives a diffusion con￾stant of 1.068 × 10-5 cm2 ·s-1 . This value of diffusion coefficient is approximately half of the value of self-diffusion coefficdient of bulk water (2.3 × 10-5 cm2 ·s-1 ), 2.4 times the value for octapeptide tube, and 42 times the value of water molecules diffusing inside the gramicidin A channel. Such a large value of diffusion coefficient indicates a bulk-like behavior of the inner diffusing water. SMD simulation was conducted to uncover the transport process of 5-FU across the lipid bilayer. Based on our simulation results and analyses, we proposed a molecular mechanism for the transport of 5-FU across CPN. During the transporting process, 5-FU hopped over each APR and resided in each MPR by switching from hydrophobic interactions with the interior wall of the nanotube to hydrogen bonding interactions with carbonyl group and amide group of each MPR. The characteristics of the pulling force profile of SMD simulations and the potential energy profile of the ortho-CPN confirmed the hopping style of 5-FU transporting. Finally, 5-FU was transported to the outer end of the nanotube by escaping from the energy well located around the kink region at subunits 7 and 8 of ortho-CPN. Both our experimental and computational results demonstrated that ortho-CPN is an ideal transporter for the delivery of antitumor drug 5-FU to tumor cells. Acknowledgment. The authors gratefully acknowledge Dr. M. R. Ghadiri for providing us the initial molecular model of the cyclic peptide nanotube and Dr. Weihua Li for his helpful discussions upon SMD simulation. This work is financially supported by the National Science Foundation of China (NSFC) (No. 30672546, 20702009) and partly by the National Key Basic Research Program of China (No. 2007CB935800, 2009CB930300). Supporting Information Available: Figures S1-S4 as discussed in the text. This material is available free of charge via the Internet at http://pubs.acs.org. MP100274F (34) Asthagiri, D.; Bashford, D. Continuum and atomistic modeling of ion partitioning into a peptide nanotube. Biophys. J. 2002, 82 (3), 1176–1189. Figure 8. The pulling force and calculated binding free energy profile for 5-FU interacting with ortho-CPN. articles Liu et al. 1994 MOLECULAR PHARMACEUTICS VOL. 7, NO. 6

点击下载完整版文档(PDF)VIP每日下载上限内不扣除下载券和下载次数;
按次数下载不扣除下载券;
24小时内重复下载只扣除一次;
顺序:VIP每日次数-->可用次数-->下载券;
已到末页,全文结束
相关文档

关于我们|帮助中心|下载说明|相关软件|意见反馈|联系我们

Copyright © 2008-现在 cucdc.com 高等教育资讯网 版权所有