CN116003453A - Reversible cyclized peptide based on double 1, 4-elimination reaction and application thereof - Google Patents

Reversible cyclized peptide based on double 1, 4-elimination reaction and application thereof Download PDF

Info

Publication number
CN116003453A
CN116003453A CN202211619035.4A CN202211619035A CN116003453A CN 116003453 A CN116003453 A CN 116003453A CN 202211619035 A CN202211619035 A CN 202211619035A CN 116003453 A CN116003453 A CN 116003453A
Authority
CN
China
Prior art keywords
peptide
polypeptide
resin
reversible
linker
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
CN202211619035.4A
Other languages
Chinese (zh)
Other versions
CN116003453B (en
Inventor
万阳
祝及宝
曾子珍
陈芝
何峰
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Jiangxi University of Traditional Chinese Medicine
Jiangxi Bencao Tiangong Technology Co Ltd
Original Assignee
Jiangxi University of Traditional Chinese Medicine
Jiangxi Bencao Tiangong Technology Co Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Jiangxi University of Traditional Chinese Medicine, Jiangxi Bencao Tiangong Technology Co Ltd filed Critical Jiangxi University of Traditional Chinese Medicine
Priority to CN202211619035.4A priority Critical patent/CN116003453B/en
Publication of CN116003453A publication Critical patent/CN116003453A/en
Application granted granted Critical
Publication of CN116003453B publication Critical patent/CN116003453B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02PCLIMATE CHANGE MITIGATION TECHNOLOGIES IN THE PRODUCTION OR PROCESSING OF GOODS
    • Y02P20/00Technologies relating to chemical industry
    • Y02P20/50Improvements relating to the production of bulk chemicals
    • Y02P20/55Design of synthesis routes, e.g. reducing the use of auxiliary or protecting groups

Landscapes

  • Peptides Or Proteins (AREA)

Abstract

The invention discloses a reversible cyclized peptide based on double 1, 4-elimination reaction and application thereof, wherein the reversible cyclized peptide passes through a linker 1 pairThe synthesis of the chain peptide and the synthesis process of the linker 1 and the reversible cyclized peptide are also disclosed. The synthesized reversible cyclized antibacterial peptide greatly reduces the hemolytic toxicity and the cytotoxicity of the antibacterial peptide and greatly enhances the stability of enzyme protein; at the same time, the reversible antibacterial peptide has no antibacterial activity per se, but is in H 2 O 2 The response factor can be used for cracking and releasing the antibacterial peptide and recovering the antibacterial activity.

Description

Reversible cyclized peptide based on double 1, 4-elimination reaction and application thereof
Technical Field
The invention belongs to the field of biochemistry, and in particular relates to a reversible cyclized peptide based on double 1, 4-elimination reaction and application thereof.
Background
Antibiotic resistance has become one of the major problems threatening public health. According to the report of the us center 2019 for disease control and prevention, more than 280 tens of thousands of people in the united states are infected with antibiotic-resistant bacteria each year, and more than 3.5 tens of thousands of people die directly from these infections. World health organization warns that by 2050, the number of deaths each year worldwide due to antibiotic-resistant pathogens is expected to reach 1000 tens of thousands. There have been reports of various antibiotic resistance mechanisms. Most conventional antibiotics bind to targets through site-specific binding mechanisms, thereby exerting pressure on the metabolism and proliferation of bacteria to inhibit or kill the bacteria. However, this site-specific binding mechanism is also affected by the rapid development of antibiotic resistance, as simple mutations in the binding site or modifications of the antibiotic structure inactivate the antibiotic. Furthermore, since many antibiotics act on intracellular targets, reduced membrane permeability and increased efflux pump activity are also important mechanisms of drug resistance. There is an urgent need for new potent antimicrobial agents that act through different mechanisms to cope with a wide range of antibiotic resistances.
In the search for new generation antibiotics, antibacterial peptides (AMPs) have received great attention over the last 30 years. AMPs coexist with microorganisms for millions of years, but have not been widely reported for resistance, which strongly suggests that they have unique antimicrobial mechanisms that may be able to evade the development of resistance. Unlike most traditional antibiotics, AMPs kill or inhibit bacteria primarily through a mechanism of membrane activity, neither involving site-specific binding nor interfering with bacterial metabolism. Although various drug resistance mechanisms have been reported, it is still very difficult for such membrane-active antimicrobial mechanisms to develop resistance to bacteria. Some AMPs have a broad range of biological activities against bacteria, fungi, parasites, insects, viruses and even cancer cells. Thus, antibacterial peptides and synthetic mimics of antibacterial peptides (SMAMPs) have been widely studied over the past 30 years as new generation antibiotics. However, current clinical transformation progress is limited, in part, due to the large toxicity, low in vivo efficiency and poor enzyme stability of the antibacterial peptides designed by the existing strategies.
Cyclization is an effective method for improving stability of antibacterial peptide proteases. The arrangement of the side chains of amino acids can be more compact through cyclization, and the steric hindrance is formed to stabilize the steric structure of the antibacterial peptide and slow down the cleavage effect of protease.
Inspired by the fact that cyclization strategies can disrupt the secondary structure of the antibacterial peptide, and in combination with the relevant reports, the present invention envisages whether or not the scientific assumption of the spiral conformation variable antibacterial peptide can be designed by reversible cyclization strategies. Briefly, the present study hopes to disrupt the secondary structure of the antimicrobial peptide by a reversible cyclization strategy, thereby obtaining a metabolically stable but biologically inactive reversible cyclized antimicrobial peptide (Reversibly Stapled AMPs, RStAMPs). At the bacterial infection site, RStAMPs can release active antibacterial peptide again under the stimulation of a stimulating factor, so that the aim of targeted administration is fulfilled. Designing a drug cascade release system or diagnostic reagent based on 1, 4-or 1, 6-benzyl elimination reaction (1, 4-or 1, 6-benzyl elimination) is a common synthetic means. Under the stimulation of the stimulation factors of the target positions, the system can spontaneously perform elimination/rearrangement reactions for multiple times, so as to achieve the purpose of releasing multiple compounds. Based on the above, the present invention is intended to construct a reversible cyclized antibacterial peptide system by a double 1, 4-benzyl elimination reaction.
Disclosure of Invention
The invention realizes the above purpose through the following technical scheme: the invention aims to construct a reversible cyclization antibacterial peptide system realized by double 1, 4-benzyl elimination reaction, which takes cheap and easily available 2, 6-bis (hydroxymethyl) p-cresol as a core, and constructs a polypeptide macrocyclic structure through forming a carbamate structure with the terminal of the antibacterial peptide or lysine side chain amino group.
The reversible cyclized antibacterial peptide (hereinafter referred to as cyclic peptide) is synthesized by coupling an antibacterial chain peptide with a linker 1, wherein the structural formula of the linker 1 is as follows:
Figure 82464DEST_PATH_IMAGE001
the antibacterial chain peptide and the reversible cyclized antibacterial peptide have the following sequences:
chain peptide TL: FVQWFSKFLGRIL-NH 2
Chain peptide TL-2: FVQWFSRFLGKIL-NH 2
Cyclic peptide RCTL: f- (linker 2) VQWFSKFLGRIL-NH 2
Cyclic peptide RCTL-2: f- (linker 2) VQWFSRFLGKIL-NH 2
Wherein F or F is Phe (phenylalanine), V is Val (valine), Q is Gln (glutamine), W is Trp (tryptophan), S is Ser (serine), K or K is Lys (lysine), L is Leu (leucine), G is Glu (glycine), R is Arg (arginine), and I is Ile (isoleucine). F and K in the cyclic peptides RCTL and RCTL-2 refer to the site of coupling of linker 1, and linker 1 cyclizes the polypeptide by coupling the two amino acids F and K.
Wherein linker 2 is a molecule obtained by cyclizing chain peptide TL and chain peptide TL-2 with linker 1 and then removing pinacol.
Linker 1 after coupling the chain peptide, at H 2 O 2 Under the action of the double 1, 4-elimination reaction, the chain peptide is released, and the active chain antibacterial peptide is obtained.
The invention also provides application of the reversible cyclized peptide in preparation of antibacterial targeted drugs.
The synthetic route of the linker 1 of the present invention is as follows:
Figure 693705DEST_PATH_IMAGE002
the synthesis method of the reversible cyclization antibacterial peptide comprises the following steps:
1. swelling of the resin: 110mg of amide resin with 0.1mmol substitution value of 0.91mmol/g is precisely weighed and put into a T-type polypeptide solid phase synthesis tube, 4-5ml of dry DMF is added, the mixture is sleeved on a cover by a self-sealing bag to be screwed up, and the mixture is put into a shaking table (220 rpm/min) to shake for 15 minutes.
2. Removal of Fmoc protecting groups of the resin: pumping the fully swelled resin, adding 4-5ml of prepared deprotection liquid (50% morpholine/DMF), placing into a shaking table, shaking for 10 minutes, pumping the deprotection liquid to dryness after the time is up, and adding 4-5ml of deprotection liquid, and shaking for 10 minutes.
3. Resin washing: taking the resin without Fmoc protecting group out of the shaking table, pumping the deprotected solution, washing with DMF once, methanol twice and DCM twice respectively, washing with DMF twice finally, pumping after the resin is washed completely, and carrying out the next step.
4. Condensation of amino acids: according to the antibacterial peptide sequence, 0.5mmol of the first required Fmoc protected amino acid is weighed and added into a 10 ml penicillin bottle, then 0.5mmol of HBTU, HOBT and 1 mmole of DiEA are weighed and added, then 4-5ml of dry DMF is added for dissolving, ultrasound is carried out for 2 minutes, so that the first required Fmoc protected amino acid is fully dissolved, and the solution is sealed for standby. And (3) washing the resin with the protective groups removed in the same way, adding the prepared amino acid into a polypeptide synthesis tube, and oscillating for 1 hour. When the steric hindrance of the amino acid is larger (the side chain of the amino acid has a larger protecting group such as Fmoc-Arg (Pbf) -OH), the amino acid should be condensed for 2 times, namely, the amino acid is pumped out after the first condensation is finished, and the condensation is repeated once again, so that the amino acid is completely coupled.
5. Synthesis of amino acid sequence: washing the resin with the first amino acid after condensation, adding deprotected solution to remove Fmoc on amino, washing the resin, adding the second amino acid, and repeating the synthesis according to the polypeptide sequence.
6. Polypeptide cleavage: the method comprises the steps of preparing a cutting reagent of 95% TFA, 2.5% Tis and 2.5% water in advance, removing Fmoc groups of the last amino acid synthesized, cleaning resin, adding 4-5 ml of the cutting reagent, placing into a shaking table to shake for 30 minutes, cutting twice, collecting the cutting liquid obtained by the two times into a 50 ml centrifuge tube, and drying with nitrogen.
7. Purifying polypeptide: adding 40 ml of methyl tertiary butyl ether into a centrifuge tube which is blown to a centrifuge tube with 1 ml of solvent left, blowing and beating uniformly by a plastic rubber head dropper, freezing and precipitating for 15 minutes in a refrigerator with the temperature of minus 20 ℃, centrifuging by a centrifuge, pouring out supernatant, dissolving and precipitating by an organic solvent, taking a trace sample, determining the molecular weight of the polypeptide by using mass spectrum, analyzing the purity of the polypeptide by using reverse high-efficiency analysis liquid phase full gradient, and purifying the polypeptide by using a preparation liquid phase.
8. Polypeptide curing: and freeze-drying the purified polypeptide by using a low-temperature freeze dryer to obtain polypeptide solid powder, namely chain peptide. And analyzing the purity of the polypeptide by analyzing the liquid phase.
9. Cyclization of polypeptide: precisely weighing chain peptide freeze-dried powder and a linker 1 (chain peptide: linker 1=1:1.2 molar ratio) to dissolve in DMSO, adding DiEA, reacting in a water bath kettle at 40 ℃ for 2 hours, freeze-drying the DMSO after the reaction is completed, adding a proper amount of methyl boric acid, dissolving with 5% TFA/DCM, reacting for 2 hours at normal temperature, removing a solvent by using a rotary evaporator, adding a proper amount of methanol for dissolving, purifying by using a preparation liquid phase, and solidifying the obtained cyclic peptide preparation liquid by using a low-temperature freeze-dryer to finally obtain white cyclic peptide solid powder.
The invention has the beneficial effects that:
1. the invention provides a connector 1 molecule based on double 1, 4-elimination reaction and a synthesis method thereof, and the method has the advantages of few synthesis steps, simple synthesis method, low manufacturing cost and the like.
2. The synthesized reversible cyclized antibacterial peptide greatly reduces the hemolytic toxicity of the antibacterial peptide (the test does not generate hemolysis at 100 mu M) and the cytotoxicity of the antibacterial peptide (the test does not generate cytotoxicity at 64 mu M), and greatly enhances the stability of enzyme protein; at the same time, the reversible antibacterial peptide has no antibacterial activity and is in H 2 O 2 The response factor can be used for cracking and releasing the antibacterial peptide and recovering the antibacterial activity.
Drawings
FIG. 1 is a mass spectrum of a chain peptide TL provided by the invention;
FIG. 2 is a mass spectrum of the chain peptide TL-2 provided by the invention;
FIG. 3 is a mass spectrum of the cyclic peptide RCTL provided by the invention;
FIG. 4 is a mass spectrum of the cyclic peptide RCTL-2 provided by the invention;
FIG. 5 is a graph showing the results of the antibacterial peptide liposome cleavage experiment provided by the invention;
FIG. 6 is a graph showing the results of an antibacterial peptide hemolysis experiment provided by the invention;
FIG. 7 is a graph showing the cytotoxicity test results of the antibacterial peptide provided by the invention;
FIG. 8 is a graph showing the experimental results of the stability of the antibacterial peptidase protein;
FIG. 9 is a graph of the experimental result of the antibacterial peptide scanning electron microscope provided by the invention;
fig. 10 is a graph showing the experimental result of the bacterial survival rate of the antibacterial peptide provided by the invention.
Detailed Description
The invention will be further described with reference to examples and with reference to the accompanying drawings. It will be apparent that the described embodiments are only some, but not all, embodiments of the invention. All other embodiments, which can be made by those skilled in the art based on the embodiments of the invention without making any inventive effort, are intended to be within the scope of the invention.
1. Synthesis of linker 1 molecule
The linker 1 molecule based on double 1, 4-elimination reaction is obtained by taking cheap and easily available 2, 6-bis (hydroxymethyl) p-cresol as a core and performing short synthesis in several steps, and the specific reaction flow is as follows:
Figure 830288DEST_PATH_IMAGE003
Synthesis of Compound 1: 2, 6-bis- (hydroxymethyl) -p-cresol (5.00 g, 29.7 mmol) and imidazole (4.65 g, 68.3 mmol) were dissolved in anhydrous DMF (30 mL) at 0deg.C. Addition of tert-butyl Di (butyl) dissolved in anhydrous DMF (15 mL)Methylchlorosilanes (9.85 g, 65.34 mmol). The solution was stirred at room temperature for 2 h, then diluted with diethyl ether and washed 3 times with water. After drying the organic phase over anhydrous MgSO4, filtering and concentrating. The crude product was purified by silica gel column (petroleum ether: ethyl acetate=90:10) to give compound S1 (11.01 g, yield 94%) as a colorless oil. 1 H NMR (600 MHz, CDCl 3 ): δ 8.09 (s, 1H), 6.98 (s, 2H), 4.90 (s, 4H), 2.33 (s, 3H), 1.03 (s, 18H), 0.21 (s, 12H). 13 C (150 MHz, CDCl 3 ): δ 151.0, 128.3, 126.3, 125.8, 63.1, 26.0, 20.7, 18.4, -5.4. ESI-MS: calcd. for C 21 H 40 NaO 3 Si 2 [M + Na] + 419.24; found 419.24。
Synthesis of Compound 2: compound 1 (2.00 g, 5.0 mmol) was dissolved in dry DMF (15 mL) and K2CO3 (0.84 g, 6.0 mmol) was added. The mixed solution was stirred at 0℃for 15 min, followed by the addition of pinacol 4-bromomethylphenyl borate (1.49, g, 5.0 mmol). The resulting mixture was stirred at room temperature overnight. After completion of the reaction, the reaction mixture was diluted with diethyl ether and washed with saturated NH4Cl and saturated brine, respectively. The organic phase was collected, dried over anhydrous MgSO4, filtered and concentrated. The residue was purified by chromatography on a silica gel column (petroleum ether: ethyl acetate=90:10) to give pure compound S2 (2.51 g, yield 82%) as a colorless oil. 1 H NMR (600 MHz, CDCl 3 ): δ7.92 (d, J = 7.6 Hz, 2H), 7.50 (d, J = 7.6 Hz, 2H), 7.25 (s, 2H), 4.97 (s, 2H), 4.78 (s, 4H), 2.41 (s, 3H), 1.42 (s, 12H), 0.99 (s, 18H), 0.14 (s, 12H). 13 C (150 MHz, CDCl 3 ): δ151.2, 140.8, 135.1, 133.8, 133.7, 128.0, 127.0, 83.8, 76.2, 60.4, 26.1, 24.9, 21.3, 18.5, -5.2. ESI-MS: calcd. for C 34 H 57 BNaO 5 Si 2 [M + Na] + 635.37; found 635.71。
Synthesis of Compound 3: to a solution of compound 2 (1.20 g, 3.0 mmol) in methanol (10 mL) was added a catalytic amount of p-toluenesulfonic acid monohydrate (0.10 g, 0.6 mmol). The resulting mixture was stirred at room temperature for 1 h. After completion, the mixture was removed under reduced pressureAn organic solvent. The residue was purified by chromatography on a silica gel column (petroleum ether: ethyl acetate=50:50) to give pure compound S3 (1.02 g, yield 89%) as a white solid. 1 H NMR (600 MHz, CDCl 3 ): δ7.82 (d, J = 7.9 Hz, 2H), 7.37 (d, J = 7.9 Hz, 2H), 7.11 (s, 2H), 4.81 (s, 2H), 4.58 (s, 4H), 3.10 (s, 2H), 2.26 (s, 3H), 1.36 (s, 12H). 13 C (150 MHz, CDCl 3 ): δ152.2, 140.1, 135.1, 134.2, 133.8, 129.3, 127.2, 84.0, 76.6, 60.4, 24.9, 20.9. ESI-MS: calcd. for C 22 H 29 BNaO 5 [M + Na] + 407.20; found 407.73。
Synthesis of linker 1: compound 3 (0.72 g, 1.9 mmol), DMAP (22.9 mg, 0.19 mmol) and DiEA (1.21 g, 9.4 mmol) were dissolved in anhydrous DCM and cooled to 0 ℃. Then, p-nitrobenzoate (0.83 g, 4.1 mmol) was added. The resulting solution was stirred at 0 ℃ for 10 min and then at room temperature for 4 h. After completion of the reaction, the reaction mixture was diluted with DCM and washed with 1N HCl and brine. The organic phase was separated, dried over MgSO4, filtered and concentrated. The residue was purified by chromatography on a silica gel column (petroleum ether: ethyl acetate=80:20) to give protected linker (linker 1) as a white solid (0.82 g, 61% yield). 1 H NMR (600 MHz, CDCl 3 ): δ8.27 (d, J = 9.0 Hz, 4H), 7.87 (d, J = 7.6 Hz, 2H), 7.50 (d, J = 7.6 Hz, 2H), 7.37 (s, 2H), 7.32 (d, J = 9.0 Hz, 4H), 5.36 (s, 4H), 5.09 (s, 2H), 2.41 (s, 3H), 1.39 (s, 12H). 13 C (150 MHz, CDCl 3 ): δ155.5, 154.6, 152.4, 145.4, 139.7, 135.2, 134.8, 132.8, 128.2, 126.7, 125.3, 121.8, 84.0, 77.7, 66.3, 24.9, 20.8.ESI-MS: calcd. for C 36 H 35 BN 2 NaO 13 [M + Na] + 737.21; found 737.48。
2. Synthesis of chain antibacterial peptide and cyclized antibacterial peptide
The chain antibacterial peptide and the cyclized antibacterial peptide synthesized by the invention have the following structures:
Figure 669806DEST_PATH_IMAGE004
Basic parameters of the polypeptides of Table 1
Figure 600853DEST_PATH_IMAGE005
The mass spectrograms of the four polypeptides are shown in figures 1, 2, 3 and 4.
The polypeptide synthesized by the invention adopts a solid-phase and liquid-phase combined mode, wherein the solid-phase synthesis part extends a chain in a polypeptide solid-phase synthesis tube according to a sequence by manually shaking a table from resin. Linker 1 after coupling the chain peptide, at H 2 O 2 Under the action of the above-mentioned linker 1 releases the chain peptide by means of double 1, 4-elimination reaction so as to obtain the active chain peptide.
Synthesis of (a) chain peptide TL:
1. swelling of the resin: 110mg of amide resin with 0.1mmol substitution value of 0.91mmol/g is precisely weighed and put into a T-type polypeptide solid phase synthesis tube, 4-5ml of dry DMF is added, the mixture is sleeved on a cover by a self-sealing bag to be screwed up, and the mixture is put into a shaking table (220 rpm/min) to shake for 15 minutes.
2. Removal of Fmoc protecting groups of the resin: pumping the fully swelled resin, adding 4-5ml of prepared deprotection liquid (50% morpholine/DMF), placing into a shaking table, shaking for 10 minutes, pumping the deprotection liquid to dryness after the time is up, and adding 4-5ml of deprotection liquid, and shaking for 10 minutes.
3. Resin washing: taking the resin without Fmoc protecting group out of the shaking table, pumping the deprotected solution, washing with DMF once, methanol twice and DCM twice respectively, washing with DMF twice finally, pumping after the resin is washed completely, and carrying out the next step.
4. Condensation of amino acids: according to the antibacterial peptide sequence (FVQWFSKFLGRIL-NH 2 ) 0.5mmol of the first required Fmoc protected amino acid is weighed into a 10 ml penicillin bottle, then 0.5mmol of HBTU, HOBT and 1 mmole of DiEA are weighed and added, then 4-5ml of dry DMF is added for dissolution, ultrasound is carried out for 2 minutes, so that the first required Fmoc protected amino acid is fully dissolved, and the solution is sealed for standby. The protective group-removed resin is prepared by the same methodThe method is used for washing cleanly, and the prepared amino acid is added into a polypeptide synthesis tube and oscillated for 1 hour. When the steric hindrance of the amino acid is larger (the side chain of the amino acid has a larger protecting group such as Fmoc-Arg (Pbf) -OH), the amino acid should be condensed for 2 times, namely, the amino acid is pumped out after the first condensation is finished, and the condensation is repeated once again, so that the amino acid is completely coupled.
5. Synthesis of amino acid sequence: washing the resin with the first amino acid after condensation, adding deprotected solution to remove Fmoc on amino, washing the resin, adding the second amino acid, and repeating the synthesis according to the polypeptide sequence.
6. Polypeptide cleavage: the method comprises the steps of preparing a cutting reagent of 95% TFA, 2.5% Tis and 2.5% water in advance, removing Fmoc groups of the last amino acid synthesized, cleaning resin, adding 4-5ml of the cutting reagent, placing into a shaking table to shake for 30 minutes, cutting twice, collecting the cutting liquid obtained by the two times into a 50 ml centrifuge tube, and drying with nitrogen.
7. Purifying polypeptide: adding 40 ml of methyl tertiary butyl ether into a centrifuge tube which is blown to a centrifuge tube with 1 ml of solvent left, blowing and beating uniformly by a plastic rubber head dropper, freezing and precipitating for 15 minutes in a refrigerator with the temperature of minus 20 ℃, centrifuging by a centrifuge, pouring out supernatant, dissolving and precipitating by an organic solvent, taking a trace sample, determining the molecular weight of the polypeptide by using mass spectrum, analyzing the purity of the polypeptide by using reverse high-efficiency analysis liquid phase full gradient, and purifying the polypeptide by using a preparation liquid phase.
8. Polypeptide curing: and freeze-drying the purified polypeptide by using a low-temperature freeze dryer to obtain polypeptide solid powder. And analyzing the purity of the polypeptide by analyzing the liquid phase.
Synthesis of (II) chain peptide TL-2:
1. swelling of the resin: 110mg of amide resin with 0.1mmol substitution value of 0.91mmol/g is precisely weighed and put into a T-type polypeptide solid phase synthesis tube, 4-5ml of dry DMF is added, the mixture is sleeved on a cover by a self-sealing bag to be screwed up, and the mixture is put into a shaking table (220 rpm/min) to shake for 15 minutes.
2. Removal of Fmoc protecting groups of the resin: pumping the fully swelled resin, adding 4-5ml of prepared deprotection liquid (50% morpholine/DMF), placing into a shaking table, shaking for 10 minutes, pumping the deprotection liquid to dryness after the time is up, and adding 4-5ml of deprotection liquid, and shaking for 10 minutes.
3. Resin washing: taking the resin without Fmoc protecting group out of the shaking table, pumping the deprotected solution, washing with DMF once, methanol twice and DCM twice respectively, washing with DMF twice finally, pumping after the resin is washed completely, and carrying out the next step.
4. Condensation of amino acids: according to the antibacterial peptide sequence (FVQWFSRFLGKIL-NH 2 ) 0.5mmol of the first required Fmoc protected amino acid is weighed into a 10 ml penicillin bottle, then 0.5mmol of HBTU, HOBT and 1 mmole of DiEA are weighed and added, then 4-5ml of dry DMF is added for dissolution, ultrasound is carried out for 2 minutes, so that the first required Fmoc protected amino acid is fully dissolved, and the solution is sealed for standby. And (3) washing the resin with the protective groups removed in the same way, adding the prepared amino acid into a polypeptide synthesis tube, and oscillating for 1 hour. When the steric hindrance of the amino acid is larger (the side chain of the amino acid has a larger protecting group such as Fmoc-Arg (Pbf) -OH), the amino acid should be condensed for 2 times, namely, the amino acid is pumped out after the first condensation is finished, and the condensation is repeated once again, so that the amino acid is completely coupled.
5. Synthesis of amino acid sequence: washing the resin with the first amino acid after condensation, adding deprotected solution to remove Fmoc on amino, washing the resin, adding the second amino acid, and repeating the synthesis according to the polypeptide sequence.
6. Polypeptide cleavage: the method comprises the steps of preparing a cutting reagent of 95% TFA, 2.5% Tis and 2.5% water in advance, removing Fmoc groups of the last amino acid synthesized, cleaning resin, adding 4-5ml of the cutting reagent, placing into a shaking table to shake for 30 minutes, cutting twice, collecting the cutting liquid obtained by the two times into a 50 ml centrifuge tube, and drying with nitrogen.
7. Purifying polypeptide: adding 40 ml of methyl tertiary butyl ether into a centrifuge tube which is blown to a centrifuge tube with 1 ml of solvent left, blowing and beating uniformly by a plastic rubber head dropper, freezing and precipitating for 15 minutes in a refrigerator with the temperature of minus 20 ℃, centrifuging by a centrifuge, pouring out supernatant, dissolving and precipitating by an organic solvent, taking a trace sample, determining the molecular weight of the polypeptide by using mass spectrum, analyzing the purity of the polypeptide by using reverse high-efficiency analysis liquid phase full gradient, and purifying the polypeptide by using a preparation liquid phase.
8. Polypeptide curing: and freeze-drying the purified polypeptide by using a low-temperature freeze dryer to obtain polypeptide solid powder. And analyzing the purity of the polypeptide by analyzing the liquid phase.
(III) Synthesis of the cyclic peptide RCTL:
1. swelling of the resin: 110mg of amide resin with 0.1mmol substitution value of 0.91mmol/g is precisely weighed and put into a T-type polypeptide solid phase synthesis tube, 4-5ml of dry DMF is added, the mixture is sleeved on a cover by a self-sealing bag to be screwed up, and the mixture is put into a shaking table (220 rpm/min) to shake for 15 minutes.
2. Removal of Fmoc protecting groups of the resin: pumping the fully swelled resin, adding 4-5ml of prepared deprotection liquid (50% morpholine/DMF), placing into a shaking table, shaking for 10 minutes, pumping the deprotection liquid to dryness after the time is up, and adding 4-5ml of deprotection liquid, and shaking for 10 minutes.
3. Resin washing: taking the resin without Fmoc protecting group out of the shaking table, pumping the deprotected solution, washing with DMF once, methanol twice and DCM twice respectively, washing with DMF twice finally, pumping after the resin is washed completely, and carrying out the next step.
4. Condensation of amino acids: according to the antibacterial peptide sequence (F- (linker 2) VQWFSKFLGRIL-NH) 2 ) 0.5mmol of the first required Fmoc protected amino acid is weighed into a 10 ml penicillin bottle, then 0.5mmol of HBTU, HOBT and 1 mmole of DiEA are weighed and added, then 4-5ml of dry DMF is added for dissolution, ultrasound is carried out for 2 minutes, so that the first required Fmoc protected amino acid is fully dissolved, and the solution is sealed for standby. And (3) washing the resin with the protective groups removed in the same way, adding the prepared amino acid into a polypeptide synthesis tube, and oscillating for 1 hour. When the steric hindrance of the amino acid is larger (the side chain of the amino acid has a larger protecting group such as Fmoc-Arg (Pbf) -OH), the amino acid should be condensed for 2 times, namely, the amino acid is pumped out after the first condensation is finished, and the condensation is repeated once again, so that the amino acid is completely coupled.
5. Synthesis of amino acid sequence: washing the resin with the first amino acid after condensation, adding deprotected solution to remove Fmoc on amino, washing the resin, adding the second amino acid, and repeating the synthesis according to the polypeptide sequence.
6. Polypeptide cleavage: the method comprises the steps of preparing a cutting reagent of 95% TFA, 2.5% Tis and 2.5% water in advance, removing Fmoc groups of the last amino acid synthesized, cleaning resin, adding 4-5 ml of the cutting reagent, placing into a shaking table to shake for 30 minutes, cutting twice, collecting the cutting liquid obtained by the two times into a 50 ml centrifuge tube, and drying with nitrogen.
7. Purifying polypeptide: adding 40 ml of methyl tertiary butyl ether into a centrifuge tube which is blown to a centrifuge tube with 1 ml of solvent left, blowing and beating uniformly by a plastic rubber head dropper, freezing and precipitating for 15 minutes in a refrigerator with the temperature of minus 20 ℃, centrifuging by a centrifuge, pouring out supernatant, dissolving and precipitating by an organic solvent, taking a trace sample, determining the molecular weight of the polypeptide by using mass spectrum, analyzing the purity of the polypeptide by using reverse high-efficiency analysis liquid phase full gradient, and purifying the polypeptide by using a preparation liquid phase.
8. Polypeptide curing: and freeze-drying the purified polypeptide by using a low-temperature freeze dryer to obtain polypeptide solid powder. And analyzing the purity of the polypeptide by analyzing the liquid phase.
9. Cyclization of polypeptide: precisely weighing chain peptide freeze-dried powder and a linker 1 (chain peptide: linker 1=1:1.2 molar ratio) to dissolve in DMSO, adding DiEA, reacting in a water bath kettle at 40 ℃ for 2 hours, freeze-drying the DMSO after the reaction is completed, adding a proper amount of methyl boric acid, dissolving with 5% TFA/DCM, reacting for 2 hours at normal temperature, removing a solvent by using a rotary evaporator, adding a proper amount of methanol for dissolving, purifying by using a preparation liquid phase, and solidifying the obtained cyclic peptide preparation liquid by using a low-temperature freeze-dryer to finally obtain white cyclic peptide solid powder.
(IV) Synthesis of the cyclic peptide RCTL-2:
1. swelling of the resin: 110mg of amide resin with 0.1mmol substitution value of 0.91mmol/g is precisely weighed and put into a T-type polypeptide solid phase synthesis tube, 4-5ml of dry DMF is added, the mixture is sleeved on a cover by a self-sealing bag to be screwed up, and the mixture is put into a shaking table (220 rpm/min) to shake for 15 minutes.
2. Removal of Fmoc protecting groups of the resin: pumping the fully swelled resin, adding 4-5ml of prepared deprotection liquid (50% morpholine/DMF), placing into a shaking table, shaking for 10 minutes, pumping the deprotection liquid to dryness after the time is up, and adding 4-5ml of deprotection liquid, and shaking for 10 minutes.
3. Resin washing: taking the resin without Fmoc protecting group out of the shaking table, pumping the deprotected solution, washing with DMF once, methanol twice and DCM twice respectively, washing with DMF twice finally, pumping after the resin is washed completely, and carrying out the next step.
4. Condensation of amino acids: according to the antibacterial peptide sequence (F- (linker 2) VQWFSRFLGKIL-NH) 2 ) 0.5mmol of the first required Fmoc protected amino acid is weighed into a 10 ml penicillin bottle, then 0.5mmol of HBTU, HOBT and 1 mmole of DiEA are weighed and added, then 4-5ml of dry DMF is added for dissolution, ultrasound is carried out for 2 minutes, so that the first required Fmoc protected amino acid is fully dissolved, and the solution is sealed for standby. And (3) washing the resin with the protective groups removed in the same way, adding the prepared amino acid into a polypeptide synthesis tube, and oscillating for 1 hour. When the steric hindrance of the amino acid is larger (the side chain of the amino acid has a larger protecting group such as Fmoc-Arg (Pbf) -OH), the amino acid should be condensed for 2 times, namely, the amino acid is pumped out after the first condensation is finished, and the condensation is repeated once again, so that the amino acid is completely coupled.
5. Synthesis of amino acid sequence: washing the resin with the first amino acid after condensation, adding deprotected solution to remove Fmoc on amino, washing the resin, adding the second amino acid, and repeating the synthesis according to the polypeptide sequence.
6. Polypeptide cleavage: the method comprises the steps of preparing a cutting reagent of 95% TFA, 2.5% Tis and 2.5% water in advance, removing Fmoc groups of the last amino acid synthesized, cleaning resin, adding 4-5ml of the cutting reagent, placing into a shaking table to shake for 30 minutes, cutting twice, collecting the cutting liquid obtained by the two times into a 50 ml centrifuge tube, and drying with nitrogen.
7. Purifying polypeptide: adding 40 ml of methyl tertiary butyl ether into a centrifuge tube which is blown to a centrifuge tube with 1 ml of solvent left, blowing and beating uniformly by a plastic rubber head dropper, freezing and precipitating for 15 minutes in a refrigerator with the temperature of minus 20 ℃, centrifuging by a centrifuge, pouring out supernatant, dissolving and precipitating by an organic solvent, taking a trace sample, determining the molecular weight of the polypeptide by using mass spectrum, analyzing the purity of the polypeptide by using reverse high-efficiency analysis liquid phase full gradient, and purifying the polypeptide by using a preparation liquid phase.
8. Polypeptide curing: and freeze-drying the purified polypeptide by using a low-temperature freeze dryer to obtain polypeptide solid powder. And analyzing the purity of the polypeptide by analyzing the liquid phase.
9. Cyclization of polypeptide: precisely weighing chain peptide freeze-dried powder and a linker 1 (chain peptide: linker 1=1:1.2 molar ratio) to dissolve in DMSO, adding DiEA, reacting in a water bath kettle at 40 ℃ for 2 hours, freeze-drying the DMSO after the reaction is completed, adding a proper amount of methyl boric acid, dissolving with 5% TFA/DCM, reacting for 2 hours at normal temperature, removing a solvent by using a rotary evaporator, adding a proper amount of methanol for dissolving, purifying by using a preparation liquid phase, and solidifying the obtained cyclic peptide preparation liquid by using a low-temperature freeze-dryer to finally obtain white cyclic peptide solid powder.
3. In vitro application of reversible cyclized antibacterial peptide in preparation of clinical antibacterial drugs
1. Liposome lysis assay
The calcein powder was weighed into a volumetric flask, added with 3/4 volume of Tris-HCl buffer (100 mM Tris-HCl 150 mM NaCl pH 7.4) and placed in an ultrasonic water bath for 10 minutes at 37 ℃. After the calcium carbonate is completely dissolved, the volume is fixed to obtain 80 mM calcein buffer solution for use in light shielding.
1, 2-dioleoyl-SN-propan-3-phosphatidylethanolamine (DOPE) and 4 mg of 1, 2-dioleoyl-SN-glyceryl-3-phosphate-RAC-glycerinum sodium salt (75/25, w/w) were weighed in amounts of 12 mg, dissolved in 3 mL chloroform/methanol (9:1, V/V) to give a colorless transparent solution, and the solvent was removed by rotary evaporation of 1 h at 37℃and the liposomes were dried, while forming a translucent film in a low pressure state. Adding calcein buffer solution 4 mL, water bathing at 37deg.C, and ultrasonic treating for 10 min. The cycle was then repeated for about 1 h by hydrating for 5 min in a 37℃water bath and vortexing for 30 min s. And (5) performing ultrasonic treatment for 30 min by using an ultrasonic cell disruption instrument to obtain the liposome with uniform diameter. Centrifuging for 15 min by using an ultrafiltration centrifuge tube (centrifugal force of 5000 xg), removing centrifugate, diluting with Tris-HCl buffer solution, cleaning liposome, centrifuging for 15 min, repeating for 3-4 times until the centrifugate has no obvious calcein, and collecting liposome in the centrifuge tube for use in dark place. Lipid concentrations were determined by the phosphorus assay.
Liposomes were diluted with Tris buffer (final lipid concentration: 0.5 mM) and the liposome solution (95. Mu.L) was then mixed with drug solution (5. Mu.L) on 96-well black microwell plates (grey, flat bottom); the final drug concentrations were made 2. Mu.M, 1. Mu.M, 0.5. Mu.M, 0.25. Mu.M, 0.125. Mu.M, 0.0625. Mu.M and 0.03125. Mu.M with 5% DMSO/Tris solution and Triton-X/Tris solution (0.1% v/v) as negative and positive controls, respectively. After incubation for 1 hour at 37 ℃ in the dark, the fluorescence intensity in each well was recorded using the instrument. The release rate of calcein was calculated based on the measured fluorescence intensity, thereby illustrating the destructive effect of the drug on the liposomes. The excitation and emission wavelengths of calcein were 490nm and 515 nm, respectively, and the calcein leakage rates were as follows: leakage (%) = [ (F-F0)/(FTX-F0) ] ×100%, F being fluorescence intensity; f0 Fluorescence intensity as negative control; FTX is the fluorescence intensity of the positive control. The detailed results are shown in FIG. 5.
As can be seen from FIG. 5, either the cyclic peptide RCTL or the cyclic peptide RCTL-2 requires H 2 O 2 Cleavage releases biological activity when no H is added 2 O 2 At this time, none of the calcein leaked in the concentrations tested, indicating no H 2 O 2 In this case, the cyclic peptides RCTL and RCTL-2 are not destroyed and have no biological activity. Both chain peptide TL and TL-2 show strong cleavage activity, the chain peptide TL and chain peptide RCTL-2 show cleavage activity at 0.125 mu M, calcein is completely released at 0.25 mu M, and the liposome is almost completely destroyed; and cyclized peptides RCTL and RCTL-2, upon addition of H 2 O 2 After incubation, the cleavage activity was shown to be almost completely equivalent to that of its parent peptide. It was shown that in liposomes, this concentration of H 2 O 2 Can completely hydrolyze cyclized peptide and release corresponding polypeptide, and cyclized peptide is cleaved by H 2 O 2
2. Hemolysis experiment
The blood used in this experiment was fresh human blood, 2 mL blood was drawn into a 15 mL centrifuge tube, 3 mL PBS (pH 7.4) was added, gently blown (to avoid vigorous blowing, causing red blood cell disruption) with a pipette, the supernatant was centrifuged with a centrifuge for 15 min (1000 rpm), and then the supernatant was washed with PBS again and clarified. The blood cells were dispersed in PBS solution to prepare a 4% (V/V) blood cell solution. The drug was precisely weighed and dissolved in DMSO to a mother liquor of 2 mM, half-diluted to the desired concentration (2 mM, 1 mM, 0.5 mM, 0.25 mM, 0.125 mM and 0.0625 mM, respectively). With 5% DMSO as a blank negative control, 2% sds solution as a positive control, 95 uL blood cell solution, 5 uL drug solution were pipetted into 96 well plates at final drug concentrations of 100 μm, 50 μm, 25 μm, 12.5 μm, 6.25 μm and 3.125 μm, three wells per level. Placing the sample in a constant temperature incubator at 37 ℃ for incubation for 1 hour, transferring the incubated sample into an EP tube (the 96-well plate is inclined and the bottom of the sample cannot be touched by a gun head to avoid red blood cell rupture), centrifuging for 15 min (1000 rpm), sucking 50 uL supernatant into the corresponding 96-well plate, and measuring the absorbance (450 nm) by using an enzyme-labeled instrument. Hemolysis (%) = [ (base:Sub>A-base:Sub>A yin)/(base:Sub>A-base:Sub>A yang-base:Sub>A yin) ]×100%base:Sub>A is absorbance intensity,base:Sub>A yin is absorbance of negative control, andbase:Sub>A yang is absorbance of positive control.
The results of the hemolysis experiments are shown in FIG. 6, in which the linear native peptides TL and TL-2 exhibit strong hemolytic activity, with TL exhibiting hemolytic activity at 6.25. Mu.M and hemolyzing up to 85% at 12.5. Mu.M; the TL-2 hemolysis activity was stronger, and started to be present at 3.125. Mu.M, and was as high as 80% at 12.5. Mu.M. Whereas, after cyclizing the polypeptides, the cyclized polypeptides RCTL and RCTL-2 showed a very large decrease in hemolytic activity, even at 100 μm. The method shows that the hemolytic activity of the polypeptide can be greatly reduced after cyclization, and the method verifies the concept of the invention, thereby achieving the purpose of reducing the hemolytic toxic and side effects of the expected polypeptide after cyclization.
3. Cytotoxicity test
The cells selected in the experiment are LO2 cells and HUVEC cells with good cell growth state, and the specific experimental steps are as follows: LO2 cells and HUVEC cells were resuscitated and passaged to normal cell morphology, after vigorous growth (at least 2-3 passages), 10% FBS in 1640 medium cell suspension and transferred to 96 well plates for growth, 100 μl of each microwell cell suspension (cell numberFor purposes of 5X 10 3 ) 24 h was cultured in a carbon dioxide incubator (37 ℃). Dissolving polypeptide medicine with DMSO to prepare mother solution with concentration of 10 mM; the 1640 medium was warmed to 37℃in a water bath in advance, and the polypeptides (TL, TL-2, RCTL and RCTL-2) were formulated with 1640 medium in half-fold dilution to drug solutions of different concentrations (64. Mu.M, 32. Mu.M, 16. Mu.M, 8. Mu.M, 4. Mu.M and 2. Mu.M) (highest concentration DMOS content 0.64%). The original culture medium in the 96-well plate is carefully sucked out (avoiding sucking into cells) by a pipette, the prepared drug solution (100 mu L of each microwell) is added, meanwhile, 0.64% DMSO/1640 solution and 20% DMSO/1640 solution are respectively used as a negative control and a positive control, three compound wells are arranged at each level, and the culture medium is put into a carbon dioxide incubator for incubation (37 ℃) for 24 hours after the drug is added. The drug solution was aspirated, 100. Mu.L of CCK-8/1640 medium solution (10%) was added to each microwell, incubated in a carbon dioxide incubator for 4 hours, and absorbance at 450nm was measured by a microplate reader, reflecting the number of living cells.
As shown in FIG. 7, the cytotoxicity results are strong for the linear peptides TL and TL-2, wherein TL and TL-2 have almost 0% cell viability at 16 μM, 32 μM, 64 μM for LO2 cells and HUVEC cells, i.e., strong cytotoxicity, whereas TL has 63% HUVEC viability, 70% LO2 cell viability, 45% HUVEC viability and 62% LO2 cell viability at 8 μM; TL and TL-2 had almost 100% cell viability at LO2 cells and HUVEC cells at concentrations of 2. Mu.M, 4. Mu.M, with no cytotoxicity. In contrast, after cyclization of the polypeptide, the cell viability of the cyclized polypeptides RCTL and RCTL-2, both LO2 cells and HUVEC cells, was nearly 100% at the tested concentration conditions, without cytotoxicity. In summary, the linear peptides TL, TL-2 were non-cytotoxic at concentrations below 4. Mu.M, starting to exhibit cytotoxicity above 4. Mu.M, and exhibited strong cytotoxicity above 16. Mu.M; and after cyclizing the peptide, no cytotoxicity was exhibited at the concentrations tested; the polypeptide can reduce the toxic and side effects of hemolysis and reduce the cytotoxicity at the same time after cyclization.
4. Enzyme protein stability assay
Trypsin (Trypsin), pepsin (Pepsin), chymotrypsin (Chymotorypsin), proteinase-K (proteinase K) and FBS (fetal bovine serum) were precisely weighed and dissolved in PBS to prepare a Trypsin solution with a concentration of 0.25. Mu.g/ml, a Chymotrypsin solution with a concentration of 0.1. Mu.g/ml, a pH=2 Pepsin solution with a concentration of 10. Mu.g/ml, a proteinase-K solution with a concentration of 0.05. Mu.g/ml and a 30% FBS solution. The polypeptides (TL-2 and RCTL-2) were precisely weighed and dissolved in DMSO to prepare a stock solution at a concentration of 2 mM. To an EP tube of 1.5. mL, 940. Mu.L of PBS solution, 50. Mu.L of drug mother liquor, 10. Mu.L/mL of enzyme solution and FBS solution were added so that the final concentration of the drug was 100. Mu.M and the DMSO content was 5%, and the mixture was homogenized by pipetting and incubated in a water bath at 37 ℃. Samples were taken after incubation for 15 min, 30 min, 1 h, 2 h, 4 h, 8 h, 16 h, 24 h, respectively. Mixing well before sampling, sampling 100 uL each time, immediately adding equal volume of glacial acetonitrile to terminate the reaction and mixing well, placing the mixture in an environment of-20 ℃ for 15 min, centrifuging the mixture for 15 min by using a 10000 rpm centrifuge, absorbing supernatant, and observing the change of TL peak areas before and after hydrolysis by RP-HPLC to judge the stability of TL in trypsin. The detailed results are shown in FIG. 8.
As can be seen from FIG. 8, the natural antibacterial peptide has stronger stability against enzymolysis after cyclization. In summary, the natural linear antibacterial peptide can greatly improve the enzymolysis resistance stability after being cyclized by the reversible cyclization strategy.
5. Minimum inhibitory concentration assay
10 mL glass test tubes were taken, 6 mL LB liquid medium solution (pH 7.4) was added, normal bacteria (ATCC 25923, ATCC 29213 and MRSA) were inoculated into the glass test tubes with an inoculating loop, rubber stoppers were added and sealed with a sealing film. Shaking at constant temperature (220 rpm,37 ℃) for 2-3 hr, culturing to logarithmic phase, collecting bacteria by centrifuge (4000 rpm,15 min), and diluting bacterial solution to bacterial concentration of 2×10 by adding LB liquid culture medium solution 5 CFU/mL, blow-down to suspend the bacterial suspension in a refrigerator at 4deg.C (maximum cannot exceed 1 h). Polypeptide medicine (TL, TL-2, RCTL-2) is dissolved in DMSO to prepare 10 mM mother solution, H 2 O 2 The solution was diluted with LB liquid medium to a mother liquor of 2 mM. By half-fold dilution, addingThe polypeptide drugs are diluted into polypeptide liquid medicine of 640 mu M, 320 mu M, 160 mu M, 80 mu M, 40 mu M and 20 mu M in DMSO solution. In the experiments for investigating the antibacterial activities of TL, TL-2, RCTL and RCTL-2, 95 mu L of bacterial liquid and 5 uL drugs are added into each microwell in a sterile 96-well plate, so that the final concentration of polypeptide drugs is respectively 32 mu M, 16 mu M, 8 mu M, 4 mu M, 2 mu M and 1 mu M, and the content of DMSO is 5%. In the investigation of 100 mu M H 2 O 2 +polypeptide drugs (TL, TL-2, RCTL-2) antibacterial Activity experiments, 90. Mu.L of bacterial liquid, 5 uL drug, 5 uL H were added to each microwell 2 O 2 Solution, ensuring the final concentration of polypeptide to be the required concentration, H 2 O 2 The concentration of the solution was 100. Mu.M and the DMSO content was 5%. Each 96-well plate was incubated in a water bath thermostatted shaker (37 ℃) for 24 h and its absorbance at 620nm was measured using a microplate reader. The detailed results are shown in Table 2 below.
Table 2 minimum inhibitory concentration results
Figure 89603DEST_PATH_IMAGE006
As can be seen from Table 2, the antimicrobial activity of the cyclized peptides RCTL and RCTL-2 was inactive at the highest concentration of 32. Mu.M for the three bacteria tested in this experiment, indicating that the cyclized strategy can greatly reduce (at least 8-fold) the antimicrobial activity of the linear antimicrobial peptide drug, even without activity, as compared to the antimicrobial activity of the linear peptides TL and TL-2. When the polypeptides TL, TL-2, RCTL-2 are 100 mu M H 2 O 2 Under action, the linear peptides TL and TL-2 showed no change in activity in three bacteria, indicating H 2 O 2 Has no influence on the activity of the linear peptide, and the cyclized peptide RCTL, RCTL-2 is shown in H 2 O 2 The activity of the peptide is reduced by one time compared with that of the linear peptide. The experiment shows that the reversible cyclization strategy can greatly reduce the antibacterial activity of the antibacterial peptide, and meanwhile, the antibacterial peptide prodrug can be replaced by H 2 O 2 "release" biological activity, but its activity is doubled compared to a linear peptide.
6. Scanning electron microscope experiment
MRSA bacteria were inoculated from a bacterial culture dish with an inoculating loop into LB liquid medium (PH=7.4)Is incubated in a logarithmic growth phase (OD 620 = 0.4-0.6) by shaking at 220ppm in a constant temperature air shaker at 37℃for 3 h. Transferring the bacterial liquid into a centrifuge tube, centrifuging the bacterial liquid by using a low-temperature centrifuge (4000 ppm,10 min), removing the supernatant, adding an equal volume of PBS buffer (PH=7.4), blowing uniformly, standing for 15s, centrifuging again, and repeating washing three times. Concentrating bacteria into 1 x 10 x 9CFU/mL, and suspending with PBS to obtain bacterial liquid for later use. The bacterial liquid without drug treatment was set as a negative control group. 950uL of bacterial liquid and 50uL of the required drug (containing 5% DMSO) were added to a 2xMIC final concentration in a 1.5ml centrifuge tube, and then H was added separately 2 O 2 (100. Mu.M) or no addition of H 2 O 2 Then incubated at 37℃for 1h. After the incubation, the bacterial solution (4000 ppm,10 min) was centrifuged with a centrifuge, the supernatant was discarded, an equal volume of PBS buffer (ph=7.4) was added and blown up evenly, and after 15s standing, it was centrifuged again and washing was repeated three times. After the last washing, the supernatant was removed, and 1mL of an electron microscope fixing solution (2.5% glutaraldehyde) (the bacterial mass was not dispersed) was directly added, and the mixture was fixed in a refrigerator at 4℃overnight. The fixed sample was washed three times with PBS buffer for 10min each time and the fixative was removed thoroughly. Then dehydrated with ethanol gradient (30%, 50%, 60%, 70%, 80%, 90% and 100%), incubated at 4℃for 15min, and centrifuged for 15min (4000 ppm,4 ℃). And uniformly dispersing the dehydrated sample with absolute ethyl alcohol, sucking 10uL of bacterial liquid onto a slide, and baking the slide on an alcohol lamp. The surface of the sample is plated with gold for 30s, and finally observed under a scanning electron microscope.
The experimental results are shown in fig. 9, and the bacteria of the negative control group (control group in fig. 9) are normal in morphology, the bacterial cell membranes are kept intact, and the cells are full spheres without leakage of contents. At this concentration, however, the linear peptides TL, TL-2 of the dosing group were severely deformed in part of the bacterial morphology (at the white arrow in fig. 9), their cell membranes were broken, the contents were released, and no globular structure was observed, indicating that they were bacterial cell membrane destruction to kill bacteria. At this concentration, the cyclized peptides RCTL, RCTL-2 were normal in bacterial morphology, indicating that their direct interaction with the bacteria did not affect the bacteria. When cyclized peptide RCTL, RCTL-2 is added to H 2 O 2 After that, the bacterial morphology was severely deformed (at the white arrow in FIG. 9), and the results were consistent with TL, TL-2. In summary, the antibacterial peptides TL and TL-2 kill bacteria by destroying bacterial membranes, and the cyclized peptides RCTL and RCTL-2 directly act without affecting bacteria; and only add H 2 O 2 After that, the biological activity, namely the activity dependence H of the cyclized peptide RCTL and RCTL-2, can be displayed 2 O 2
7. Bacterial viability experiment
An LB nutrient agar ultrapure water solution was prepared according to the formulation, and the solution and a clean glass petri dish were sterilized in an autoclave (121 ℃) for 30 minutes. The glass culture dish is put into a baking oven to dry water, LB agar is carefully poured into the glass culture dish (20 mL each) after the temperature is reduced to about 60 ℃, the glass culture dish is irradiated under an ultraviolet lamp and naturally cooled, and the glass culture dish is cooled and solidified after 2-3 h for later use. Precisely weighing trypsin 1 mg, preparing into trypsin solution with concentration of 0.025 mg/mL by PBS solution, and preserving at low temperature (4 ℃) for standby. Taking a mother solution of TL-2 or RCTL-2 of 2 mM (DMSO dissolution), adding 940 mu L of PBS solution, 50 mu L of TL-2 or RCTL-2 mother solution of uL and 10 mu L/mL of trypsin solution (0.025 mg/mL) into an EP tube of 1.5 mL, so that the final concentration of TL-2 or RCTL-2 medicine is 100 mu M, the DMSO content is 5 percent, taking 5 percent DMSO PBS solution as a blank, blowing uniformly, and then incubating the mixed solution for 1 hour under the water bath condition of 37 ℃. Immediately placing the EP tube into a water bath kettle at 60 ℃ for 15 min to deactivate trypsin and terminate the reaction, obtaining enzymolysis liquid medicine or blank solution, and preserving the enzymolysis liquid medicine or blank solution in a refrigerator at 4 ℃ for later use. Selecting MRSA bacteria, culturing with LB culture medium to logarithmic phase, centrifuging with centrifuge to collect bacterial liquid (4000 rpm,15 min), diluting bacterial liquid with LB culture medium to 2×10 6 CFU/mL; diluting enzymolysis agent and H with LB culture medium 2 O 2 Solution, 490. Mu.L of bacterial liquid and 500. Mu.L of enzymatic drug solution 10. Mu.L of H were added to a 1.5 mL EP tube 2 O 2 The solution is such that the final concentration of the bacterial liquid is 1 multiplied by 10 6 CFU/mL, the final concentration of the enzymolysis liquid medicine is 25 mu M, 10 mu M and 5 mu M respectively, the final concentration of the H2O2 solution is 100 mu M, and the three are evenly mixed and incubated in a constant temperature incubator at 37 ℃ for 1H. At the same time dilute the bacteria 10 2 、10 3 、10 4 Fold, 100. Mu.L of incubation liquid was aspirated and carefully spread on LB agar plates at room temperature. Culturing in a constant temperature incubator for 24 hours, counting the colonies on the plate, and reflecting the inhibition condition of the drug on bacteria by the colony count.
The results of the experiment are shown in FIG. 10, in which TL-2 does not exhibit biological activity at each concentration, indicating that TL-2 is completely hydrolyzed by trypsin, whereas RCTL-2 completely kills bacteria at 25. Mu.M and exhibits killing of bacteria at both 10. Mu.M and 5. Mu.M.

Claims (7)

1. A double 1, 4-elimination reaction-based linker 1, characterized in that: the connector 1 has the following structure:
Figure 900509DEST_PATH_IMAGE001
2. a reversible cyclized peptide synthesized by coupling of linker 1 according to claim 1, wherein: the linker 1 is coupled with a chain peptide to synthesize a reversible cyclized peptide, and the sequences of the chain peptide and the reversible cyclized peptide are as follows:
Chain peptide TL: FVQWFSKFLGRIL-NH 2
Chain peptide TL-2: FVQWFSRFLGKIL-NH 2
Reversible cyclization peptide RCTL: f- (linker 2) VQWFSKFLGRIL-NH 2
Reversible cyclization peptide RCTL-2: f- (linker 2) VQWFSRFLGKIL-NH 2
Wherein, the linker 2 is a molecule obtained by removing pinacol after cyclizing the chain peptide TL and the chain peptide TL-2 by the linker 1.
3. Connector 1 according to claim 1 or 2, characterized in that: the linker 1 is attached to the chain peptide at H 2 O 2 Under the influence of (a) the chain peptide can be released by a double 1, 4-elimination reaction.
4. A method of synthesizing the linker 1 according to claim 1, characterized in that: the synthetic route is as follows:
Figure DEST_PATH_IMAGE002
5. a method of synthesizing a reversible cyclized peptide according to claim 2, characterized in that: the method comprises the following steps:
(1) Swelling of the resin: precisely weighing 110mg of amide resin with 0.1mmol substitution value of 0.91mmol/g, placing into a T-type polypeptide solid-phase synthesis tube, adding 4-5ml dry DMF, sleeving a self-sealing bag on a cover, tightening, and placing into a shaking table to shake for 15 minutes;
(2) Removal of Fmoc protecting groups of the resin: pumping the fully swelled resin, adding 4-5ml of the prepared deprotection liquid, putting into a shaking table, shaking for 10 minutes, pumping the deprotection liquid to dryness after the time is up, and adding 4-5ml of the deprotection liquid, and shaking for 10 minutes;
(3) Resin washing: taking the resin without Fmoc protecting groups out of a shaking table, pumping the deprotected solution, washing with DMF once, methanol twice and DCM twice respectively, washing with DMF twice finally, pumping after the resin is washed completely, and then carrying out the next step;
(4) Condensation of amino acids: weighing 0.5mmol of first required Fmoc protected amino acid according to a target polypeptide sequence, adding into a 10 ml penicillin bottle, weighing and adding 0.5mmol of HBTU, HOBT and 1 mmole of DiEA, adding 4-5ml of dry DMF for dissolving, performing ultrasonic treatment for 2 minutes to fully dissolve, sealing for standby, washing the resin with the protective groups removed in the same manner, adding the prepared amino acid into a polypeptide synthesis tube, and vibrating for 1 hour;
(5) Synthesis of amino acid sequence: washing the resin with condensed first amino acid according to the steps, adding deprotection solution to remove Fmoc on amino, washing the resin, adding second amino acid, and synthesizing repeatedly according to polypeptide sequence;
(6) Polypeptide cleavage: pre-preparing a cutting reagent of 95% TFA, 2.5% Tis and 2.5% water, removing Fmoc groups of the last amino acid synthesized, cleaning resin, adding 4-5ml cutting reagent, putting into a shaking table for shaking for 30 minutes, cutting twice, collecting the cutting liquid of the two times in a 50 ml centrifuge tube, and drying with nitrogen;
(7) Purifying polypeptide: adding 40 ml of methyl tertiary butyl ether into a centrifuge tube which is blown to a centrifuge tube with 1 ml of solvent left, blowing and beating uniformly by a plastic rubber head dropper, putting into a refrigerator with the temperature of minus 20 ℃ for freezing and precipitating for 15 minutes, centrifuging by using a centrifuge, pouring out supernatant, dissolving and precipitating by using an organic solvent, taking a trace sample, determining the molecular weight of the polypeptide by using mass spectrum, analyzing the purity of the polypeptide by using a reverse high-efficiency analysis liquid phase full gradient, and purifying the polypeptide by using a preparation liquid phase;
(8) Polypeptide curing: freeze-drying the purified polypeptide by using a low-temperature freeze dryer to obtain polypeptide freeze-dried powder, namely chain peptide;
(9) Cyclization of polypeptide: precisely weighing chain peptide freeze-dried powder and a connector 1, dissolving in DMSO, adding DiEA, reacting in a water bath kettle at 40 ℃ for 2 hours, freeze-drying the DMSO after the reaction is completed, adding a proper amount of methyl boric acid, dissolving with 5% TFA/DCM, reacting for 2 hours at normal temperature, removing a solvent by using a rotary evaporator, adding a proper amount of methanol for dissolving, purifying by using a preparation liquid phase, and solidifying the obtained cyclic peptide preparation liquid by using a low-temperature freeze-dryer to finally obtain white cyclic peptide solid powder.
6. The method for synthesizing a reversible cyclized peptide according to claim 5, wherein: the molar ratio of the chain peptide to the linker 1 is 1:1.2.
7. Use of a reversible cyclic peptide according to claim 2 for the preparation of an antibacterial targeted drug.
CN202211619035.4A 2022-12-16 2022-12-16 Reversible cyclized peptide based on double 1, 4-elimination reaction and application thereof Active CN116003453B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202211619035.4A CN116003453B (en) 2022-12-16 2022-12-16 Reversible cyclized peptide based on double 1, 4-elimination reaction and application thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202211619035.4A CN116003453B (en) 2022-12-16 2022-12-16 Reversible cyclized peptide based on double 1, 4-elimination reaction and application thereof

Publications (2)

Publication Number Publication Date
CN116003453A true CN116003453A (en) 2023-04-25
CN116003453B CN116003453B (en) 2024-07-30

Family

ID=86034603

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202211619035.4A Active CN116003453B (en) 2022-12-16 2022-12-16 Reversible cyclized peptide based on double 1, 4-elimination reaction and application thereof

Country Status (1)

Country Link
CN (1) CN116003453B (en)

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5811392A (en) * 1994-06-08 1998-09-22 Yissum Research Development Co. Of The Hebrew University Conformationally constrained backbone cyclized peptide analogs
CN107400161A (en) * 2017-09-19 2017-11-28 中国工程物理研究院核物理与化学研究所 Equal cyclic peptide Cyclo [(Asp)5Gly] preparation method
CN108315308A (en) * 2018-02-26 2018-07-24 中南民族大学 A kind of biologically active COX18 polypeptides and its synthetic method and purposes
CN112625092A (en) * 2021-01-13 2021-04-09 兰州大学 Antibacterial polypeptide compound based on polybia-MPI and synthesis and application thereof
CN112625106A (en) * 2021-01-13 2021-04-09 兰州大学 Antibacterial polypeptide compound, synthesis method and application thereof

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5811392A (en) * 1994-06-08 1998-09-22 Yissum Research Development Co. Of The Hebrew University Conformationally constrained backbone cyclized peptide analogs
CN107400161A (en) * 2017-09-19 2017-11-28 中国工程物理研究院核物理与化学研究所 Equal cyclic peptide Cyclo [(Asp)5Gly] preparation method
CN108315308A (en) * 2018-02-26 2018-07-24 中南民族大学 A kind of biologically active COX18 polypeptides and its synthetic method and purposes
CN112625092A (en) * 2021-01-13 2021-04-09 兰州大学 Antibacterial polypeptide compound based on polybia-MPI and synthesis and application thereof
CN112625106A (en) * 2021-01-13 2021-04-09 兰州大学 Antibacterial polypeptide compound, synthesis method and application thereof

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
OZLEM SEVEN ET.AL: ""Self Immolative Dioxetane Based Chemiluminescent Probe for H2O2Detection"", 《SENSORS AND ACTUATORS B》, 20 September 2016 (2016-09-20), pages 16 *
TATSUYUKI YOSHII ET.AL: ""Chemically Reactive Supramolecular Hydrogel Coupled with a Signal Amplification System for Enhanced Analyte Sensitivity"", 《JOURAL OF AMERICAN CHEMICAL SOCIETY》, 13 February 2015 (2015-02-13), pages 3363 *

Also Published As

Publication number Publication date
CN116003453B (en) 2024-07-30

Similar Documents

Publication Publication Date Title
CN109232719B (en) PH-responsive antibacterial peptide and preparation method and application thereof
Jiang et al. Self-assembly of cationic multidomain peptide hydrogels: supramolecular nanostructure and rheological properties dictate antimicrobial activity
WO2006058436A1 (en) Antimicrobial peptides with reduced hemolysis and methods of their use
CN105566452A (en) Antibacterial peptide with annular structure and preparation method and application thereof
CN103421090B (en) A kind of novel antimicrobial peptide
CN116874613B (en) Broad-spectrum efficient antibacterial polypeptide APH143 and preparation method and application thereof
CN111363010A (en) Symmetric short-sequence antibacterial peptide analogue and application thereof
RU2415868C1 (en) Hemin derivatives, having antimicrobial activity or pharmaceutically acceptable salts thereof, synthesis method, pharmaceutical composition and use
JP6124139B2 (en) Low erythrocyte lytic antimicrobial peptide, pharmaceutical composition and use thereof
US9090655B2 (en) Low hemolytic antimicrobial peptide, pharmaceutical composition and use thereof
CN116003453B (en) Reversible cyclized peptide based on double 1, 4-elimination reaction and application thereof
CN112625092B (en) Antibacterial polypeptide compound based on polybia-MPI and synthesis and application thereof
CN110054664B (en) Side chain fatty acid modified antibacterial peptide analogue containing D-type amino acid and synthesis and application thereof
CN110066320B (en) Cyclic peptide resisting multiple drug-resistant bacteria and preparation method and application thereof
CN112625106A (en) Antibacterial polypeptide compound, synthesis method and application thereof
CN116947999A (en) Small molecular antibacterial polypeptide, preparation method and application thereof
CN104672307B (en) A kind of method for improving cation small peptide antibiotic property and stability
JP2005247721A (en) Antimicrobial peptide having reduced hemolysis, and method for using the same
WO2020119773A1 (en) Amphotericin b peptide derivative
CN107033219A (en) A kind of self-assembling peptides and its application as DNA agglomeration reagents
RU2758060C1 (en) Bacillus subtilis strain - producer of a low-molecular antimicrobial peptide and method for producing a low-molecular antimicrobial peptide
CN110437308A (en) One kind has antibacterial peptide analogues containing beta amino acids and its application of specific activity to pseudomonas aeruginosa
CN112341524B (en) Cyclic antibacterial peptide analogue rich in positive charges and application thereof
CN118480089A (en) Gemini type surfactant-like antibacterial peptide resistant to enzymolysis, and preparation method and application thereof
CN118307634A (en) Antibacterial peptide derivative, synthesis method thereof and application thereof in resisting drug-resistant bacteria

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
GR01 Patent grant
GR01 Patent grant