CN111793111B - Application of cryopreservation liquid containing peptide compounds in stem cell cryopreservation - Google Patents

Application of cryopreservation liquid containing peptide compounds in stem cell cryopreservation Download PDF

Info

Publication number
CN111793111B
CN111793111B CN202010172545.6A CN202010172545A CN111793111B CN 111793111 B CN111793111 B CN 111793111B CN 202010172545 A CN202010172545 A CN 202010172545A CN 111793111 B CN111793111 B CN 111793111B
Authority
CN
China
Prior art keywords
solution
cryopreservation
use according
thr
pva
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.)
Active
Application number
CN202010172545.6A
Other languages
Chinese (zh)
Other versions
CN111793111A (en
Inventor
乔杰
严杰
闫丽盈
李蓉
王健君
金晟琳
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.)
Institute of Chemistry CAS
Peking University Third Hospital Peking University Third Clinical Medical College
Original Assignee
Institute of Chemistry CAS
Peking University Third Hospital Peking University Third Clinical Medical College
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 Institute of Chemistry CAS, Peking University Third Hospital Peking University Third Clinical Medical College filed Critical Institute of Chemistry CAS
Publication of CN111793111A publication Critical patent/CN111793111A/en
Application granted granted Critical
Publication of CN111793111B publication Critical patent/CN111793111B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/06Dipeptides
    • C07K5/06008Dipeptides with the first amino acid being neutral
    • C07K5/06017Dipeptides with the first amino acid being neutral and aliphatic
    • C07K5/0606Dipeptides with the first amino acid being neutral and aliphatic the side chain containing heteroatoms not provided for by C07K5/06086 - C07K5/06139, e.g. Ser, Met, Cys, Thr
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01NPRESERVATION OF BODIES OF HUMANS OR ANIMALS OR PLANTS OR PARTS THEREOF; BIOCIDES, e.g. AS DISINFECTANTS, AS PESTICIDES OR AS HERBICIDES; PEST REPELLANTS OR ATTRACTANTS; PLANT GROWTH REGULATORS
    • A01N1/00Preservation of bodies of humans or animals, or parts thereof
    • A01N1/02Preservation of living parts
    • A01N1/0205Chemical aspects
    • A01N1/021Preservation or perfusion media, liquids, solids or gases used in the preservation of cells, tissue, organs or bodily fluids
    • A01N1/0221Freeze-process protecting agents, i.e. substances protecting cells from effects of the physical process, e.g. cryoprotectants, osmolarity regulators like oncotic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/06Dipeptides
    • C07K5/06086Dipeptides with the first amino acid being basic
    • C07K5/06095Arg-amino acid
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/06Dipeptides
    • C07K5/06139Dipeptides with the first amino acid being heterocyclic
    • C07K5/06165Dipeptides with the first amino acid being heterocyclic and Pro-amino acid; Derivatives thereof
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/08Tripeptides
    • C07K5/0802Tripeptides with the first amino acid being neutral
    • C07K5/0804Tripeptides with the first amino acid being neutral and aliphatic
    • C07K5/0806Tripeptides with the first amino acid being neutral and aliphatic the side chain containing 0 or 1 carbon atoms, i.e. Gly, Ala
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/08Tripeptides
    • C07K5/0802Tripeptides with the first amino acid being neutral
    • C07K5/0804Tripeptides with the first amino acid being neutral and aliphatic
    • C07K5/081Tripeptides with the first amino acid being neutral and aliphatic the side chain containing O or S as heteroatoms, e.g. Cys, Ser

Abstract

The invention discloses application of cryopreservation liquid containing peptide compounds in stem cell cryopreservation. The peptide compound consists of the ice-philic amino acid and the hydrophilic group, has good capability of inhibiting the growth of ice crystals and modifying the appearance of the ice crystals in aqueous solution, has no thermal hysteresis and good biocompatibility, and is an ideal ice control material. The cryopreservation solution containing the peptide compound contains the peptide compound, polyhydric alcohol, water-soluble sugar and other components, and can keep good survival rate when being used for cryopreservation of stem cells.

Description

Application of cryopreservation liquid containing peptide compounds in stem cell cryopreservation
The present application claims the priority of a prior application entitled "a peptide compound and a cryopreservation solution containing the same" filed on the national intellectual property office of china with patent application number 201910281986.7, filed on 9/4/9/2019. The entire contents of this prior application are incorporated by reference into this application.
Technical Field
The invention belongs to the technical field of biomedical materials, and particularly relates to application of cryopreservation solution containing peptide compounds in stem cell cryopreservation.
Background
Cryopreservation refers to keeping the biological material at ultralow temperature to slow down or stop cell metabolism and division, and to continue to develop once normal physiological temperature is restored. Since the advent, this technique has become one of indispensable research methods in the field of natural science, and has been widely adopted. In recent years, with the increasing pressure of life, human fertility tends to decrease year by year, and fertility preservation is receiving more and more attention, and cryopreservation of human germ cells (sperm and oocyte), gonadal tissue, and the like is an important means for fertility preservation. In addition, as the world population ages, the need for cryopreservation of donated human cells, tissues or organs available for regenerative medicine and organ transplantation is also increasing dramatically. Therefore, how to efficiently store precious cells, tissues and organ resources in a freezing way becomes a scientific and technical problem to be solved urgently.
The most common cryopreservation method currently used is vitrification freezing. The vitrification freezing technology can ensure that liquid inside and outside cells is in a vitrification state in the rapid freezing process, thereby avoiding damage in the freezing process. However, prior art cryopreservation reagents are not effective in controlling ice crystal growth during rewarming, thereby damaging the cells. The high concentration (more than or equal to 15 percent) of toxic organic solvents used by the prior vitrification freezing method, such as: DMSO causes toxic and side effects of the cryopreservation reagent on cells, and seriously influences survival rate and even (offspring) safety and function expression of the cryopreserved objects after resuscitation. In conclusion, the currently adopted freezing preservation reagent does not have the capability of effectively controlling the growth of ice crystals in the rewarming process, and simultaneously has the problem of poor safety of the reagent.
Disclosure of Invention
In order to overcome the defects of the prior art, the invention provides a peptide compound, a synthesis method thereof, a cryopreservation solution containing the peptide compound, and application of the cryopreservation solution.
The invention is realized by the following technical scheme:
a peptide compound consisting of an ice-philic amino acid, such as: threonine (L-Thr), glutamine (L-Gln), aspartic acid (L-Asn), etc. with other hydrophilic amino acids selected from arginine, proline, alanine, etc. or Gluconolactone (GDL) or saccharides.
According to the invention, the peptide compound is a polypeptide compound formed by more than two amino acid units, such as: 2-8 amino acid units, specifically 2, 3, 4, 5, 6 amino acid units; the polypeptide compound is composed of more than two different amino acids.
As an exemplary technical scheme, the molar ratio of the ice-philic amino acid to other hydrophilic amino acids in the peptide compound is (0.1-3): 1, preferably (0.5-2): 1.
according to the present invention, the arrangement of the hydrophilic amino acid and the hydrophilic amino acid in the peptide compound is not particularly limited, and the amino acid and the hydrophilic amino acid may be connected by amino acid linking groups or chemical bonds known in the art, for example, the hydrophilic amino acid and the hydrophilic amino acid may be arranged in a single interval, or a plurality of hydrophilic amino acids may be linked to form a hydrophilic amino acid fragment or a hydrophilic amino acid fragment, and then linked to the hydrophilic amino acid (or fragment) and the hydrophilic amino acid (or fragment), respectively.
According to the invention, the peptidic compound is at least one of L-Thr-L-Arg (TR), L-Thr-L-Pro (TP), L-Arg-L-Thr (RT), L-Pro-L-Thr (PT), L-Thr-L-Arg-L-Thr (TRT), L-Thr-L-Pro-L-Thr (TPT), L-Ala-L-Ala-L-Thr (AAT), L-Thr-L-Cys-L-Thr (TCT).
According to the invention, the peptide compound is a molecule formed by chemically bonding glucolactone or other saccharides and an ice-philic amino acid, such as: GDL-L-Thr, GDL-L-Gln, GDL-L-Asn, GDL-L-Phe, GDL-L-Tyr, -GDL-L-Val, GDL-L-Ser.
According to the invention, the peptide compound has any one structure shown in formula (1) to formula (8):
Figure BDA0002409683710000031
Figure BDA0002409683710000041
the peptide compounds can be prepared by peptide synthesis methods known in the art, such as solid phase synthesis.
The preparation method comprises the following steps of swelling resin, covalently connecting amino acid protected by one amino group to the swelled resin, deprotecting, adding another amino acid protected by the other amino group for condensation reaction, deprotecting, cutting and purifying.
According to the preparation method of the present invention, the glycopeptide derivative may be prepared by a reaction method of an amino acid and a saccharide known in the present invention, for example, a reaction method of gluconolactone or other saccharides with an amino acid in an organic solvent, or a solid phase synthesis method. In one embodiment, the glycopeptide derivative is synthesized by dissolving Gluconolactone (GDL) in an organic solvent, and adding an amino acid and a basic catalyst to the organic solvent to perform a reaction.
According to the preparation method of the present invention, the organic solvent may be selected from methanol, ethanol, and the like. According to the preparation method, the GDL-L-Thr is prepared by adopting the following synthetic route:
Figure BDA0002409683710000051
the invention also provides application of the peptide compound in controlling ice crystal growth in an aqueous solution and application of the peptide compound in preparing a cell or tissue cryopreservation solution.
In one embodiment, the glycopeptide derivative is prepared using a solid phase synthesis method comprising: swelling resin, covalently linking an amino acid protected by an amino group to the swollen resin, deprotecting, adding a saccharide compound (such as gluconolactone) for condensation reaction, cleavage, and purification. Methods for the synthesis of GDL-L-Val and GDL-L-Ser refer to the method for the synthesis of GDL-L-Thr.
The present invention also provides a peptide compound represented by formula (9):
Figure BDA0002409683710000052
wherein R is selected from substituted or unsubstituted alkyl, and the substituent can be selected from-OH, -NH 2 、-COOH、-CONH 2 Etc., e.g., R is substituted or unsubstituted C 1-6 Alkyl, preferably R is-CH 3 、-CH 2 CH 3 、-CH 2 CH 2 COOH; n is an integer of 1 to 1000 inclusive, and may be, for example, an integer in the range of 1 to 100. In some embodiments of the invention, n is an integer of 2, 3, 4, 5, 6, 7, 8, 9, 10.
As an embodiment of the present invention, the compound represented by the formula (9) has a structure represented by any one of the following:
Figure BDA0002409683710000061
according to the invention, the compound of formula (9) is prepared using the following synthetic route:
Figure BDA0002409683710000062
the invention also provides application of the compound of the formula (9) in controlling ice crystal growth in aqueous solution, and application of the compound of the formula (9) in preparing a cell or tissue cryopreservation solution.
According to the application of the invention, the peptide compound can be combined with other ice control materials, and the ice control materials are selected from at least one of PVA, amino acid, polyamino acid and the like.
The invention also provides a cryopreservation solution containing the peptide compound.
The cryopreservation solution according to the present invention contains 0.1 to 50g of the peptide compound per 100 mL.
The cryopreservation solution of the present invention further contains one or more of PVA, amino acid, polyamino acid, DMSO, and polyhydric alcohol.
The cryopreservation solution of the present invention may further contain serum.
The cryopreservation solution comprises 0.1-50g of the peptide compound, 0-6.0g of PVA,0-9.0g of polyamino acid, 0-15mL of DMSO,5-45mL of polyol and 0.1-1.0mol of L per 100mL -1 0-30mL of serum, and the balance of buffer solution.
In one embodiment of the present invention, the cryopreservation solution comprises, per 100mL, 0.1 to 50g of the peptide compound, 0.1 to 15mL of DMSO,5.0 to 45mL of a polyol, and 0.1 to 1.0mol L of a buffer solution -1 Water-soluble sugar, 0-30mL of serum, no PVA and polyamino acid, and the balance of buffer solution.
As an embodiment of the present invention, the cryopreservation solution comprises 0.1 to 50g of the peptide compound, 0.1 to 6.0g of PVA,0 to 9.0g of polyamino acid, 0 to 15mL of DMSO,5.0 to 45mL of polyol, 0.1 to 1.0mol L per 100mL -1 0-30mL of serum, and the balance of buffer solution.
In one embodiment of the present invention, the cryopreservation solution comprises, per 100mL, 0.1 to 50g of the peptide compound, 0.1 to 6.0g of PVA,0.1 to 9.0g of the polyamino acid, 0.1 to 7.5mL of DMSO,5.0 to 45mL of the polyhydric alcohol, and 0.1 to 1.0mol L of the polyhydric alcohol -1 The balance being buffer solution.
In one embodiment, the cryopreservation solution contains 0.1-10mL of DMSO, e.g., 0.1-7.5mL of DMSO.
In one embodiment, the cryopreservation solution contains 0.1 to 6.0g of PVA, for example 0.1 to 4.0g of PVA.
In one embodiment, the cryopreservation solution contains 5 to 30mL of the polyol, e.g., 8.0 to 25mL, 20mL, 15mL.
In one embodiment, the cryopreservation solution contains 0.1 to 0.8mol L -1 Water-soluble sugars of (2), e.g. 0.2-0.6mol L -1 The sucrose of (1).
In one embodiment, the cryopreservation solution contains 5.0-30mL of serum, such as 5.0-20mL of fetal bovine serum; in another embodiment, the cryopreservation solution does not contain fetal bovine serum.
According to the invention, the PVA is selected from one or a combination of two or more of isotactic PVA, syndiotactic PVA and atactic PVA, for example the PVA has a syndiotacticity of from 15% to 60%, in particular for example from 50% to 60%, from 50% to 55%.
According to the invention, the PVA may be chosen from the group comprising PVA with a molecular weight of 10-500kDa, for example with a molecular weight of 10-30kDa, 30-50kDa, 80-90kDa, 200-500kDa.
According to the present invention, the polyamino acid may be selected from at least one homopolymer of lysine, arginine, proline, threonine, histidine, and the like, or a copolymer of two or more amino acids.
According to the invention, the polyol may be a polyol having from 2 to 5 carbon atoms, preferably a diol having from 2 to 3 carbon atoms, and/or a triol, such as any of ethylene glycol, propylene glycol, glycerol; ethylene glycol is preferred.
According to the invention, the water-soluble sugar may be at least one of non-reducing disaccharides, water-soluble polysaccharides, sugar anhydrides, for example selected from sucrose, trehalose, water-soluble celluloses (e.g. hydroxypropyl methylcellulose, etc.), polysucrose; sucrose and hydroxypropylmethyl cellulose are preferable. The water-soluble sugar can protect cell membranes and prevent cell sedimentation.
According to the present invention, the buffer may be selected from at least one of DPBS or hepes-buffered HTF buffer, or other cell culture buffer.
According to the invention, the serum can be selected from human serum albumin or analogues thereof, such as Sodium Dodecyl Sulfate (SDS); fetal bovine serum or bovine serum albumin can be selected for non-human cryopreservation subjects.
The preparation method of the frozen preservation solution comprises the steps of dissolving the peptide compound in a buffer solution, cooling to room temperature, adjusting pH, dissolving other components in another buffer solution, cooling, mixing, adjusting pH, fixing the volume to a preset volume by using the buffer solution, and optionally adding serum when in use.
The preparation method comprises the following steps:
(1) Dissolving a peptide compound in a part of buffer solution, cooling to room temperature, and adjusting the pH value to obtain a solution 1;
(2) Dissolving water-soluble sugar in a part of buffer solution, and adding other components after the water-soluble sugar is completely dissolved to prepare a solution 2;
(3) Optionally, dissolving PVA and/or a polyamino acid in the other part of buffer solution, cooling to room temperature, and adjusting the pH value to prepare a solution 3;
(4) And (3) cooling the solution 1, the solution 2 and optionally the solution 3 to room temperature, mixing, adjusting the pH value, and fixing the volume to a preset volume by using a buffer solution to obtain the cryopreservation liquid.
According to the preparation method of the invention, in the step (3), the PVA is heated and dissolved in a warm bath; for example, the bath temperature is 60 to 85 ℃ and preferably 80 ℃. In the step (3), the dissolving includes a stirring step.
According to the preparation method of the present invention, in the step (2), the dissolution is ultrasound-assisted dissolution.
In the present invention, hydrophilic means capable of forming a non-covalent interaction with a water molecule, for example, capable of forming a hydrogen bond, van der waals interaction, electrostatic interaction, hydrophobic interaction or pi-pi interaction with water;
ice-philic refers to a non-covalent interaction with ice, such as hydrogen bonding, van der Waals interactions, electrostatic interactions, hydrophobic interactions, or pi-pi interactions with ice.
In the invention, when the cryopreservation solution is used, a cryopreservation equilibrium solution known in the field can be used, preferably, the DMSO content in the cryopreservation solution is 0, and the cryopreservation solution contains 7.5-15mL of polyalcohol, 10-20mL of serum and the balance of buffer solution per 100 mL; preferably, when the serum content in the cryopreservation solution is also 0, the cryopreservation solution contains 1.0 to 5.0g of PVA, 7.5 to 15mL of polyol and the balance of buffer solution per 100 mL.
The components in the frozen equilibrium solution have the same meaning as in the frozen stock solution.
The invention also provides application of the cryopreservation liquid or the cryopreservation equilibrium liquid or the cryopreservation reagent, which is used for cryopreservation of cells, tissues or organs; for example for cryopreservation of oocytes, sperm or stem cells, ovarian tissue, embryos or ovarian organs.
Further, the present invention provides an application of the cryopreservation liquid to cryopreservation of stem cells.
According to an embodiment of the present invention, the stem cell is a variety of stem cells known in the art with differentiation function, such as a pluripotent stem cell, a multipotent stem cell or a multipotent stem cell, including but not limited to an embryonic stem cell, various types of mesenchymal stem cells (e.g., umbilical cord mesenchymal stem cell, adipose mesenchymal stem cell, bone marrow mesenchymal stem cell, etc.), a hematopoietic stem cell, and the like.
According to an embodiment of the present invention, the stem cell cryopreservation employs a micro-drop method, for example the application of said stem cell cryopreservation comprises the steps of: adding the cryopreservation liquid into the stem cells, blowing and dispersing to prepare stem cell suspension, placing the stem cell suspension on a freezing slide, and performing cryopreservation by using liquid nitrogen (-196 ℃).
According to an embodiment of the present invention, thawing of the cryopreserved stem cells comprises placing the frozen slides with the stem cells in the a-MEM medium and thawing at 37 ℃.
In the present invention, "cryopreservation" and "cryopreservation" have the same meaning and are used interchangeably, and refer to preservation of a substance, or a cell, a tissue, or an organ at a low temperature so that the substance retains its original physicochemical and/or biological activity, physiological and biochemical functions.
Advantageous effects
The inventor of the invention finds that in the process of controlling the ice crystal growth in an ice-water mixed phase, the material needs to have good interaction with both ice and water, so that the peptide compound which is formed by combining an ice-philic amino acid such as threonine and other hydrophilic amino acids or saccharides is synthesized according to an ice-philic-hydrophilic ice-controlling mechanism, the synthesized peptide compound has good ice-controlling effect, can modify the appearance of the ice crystal, has no thermal hysteresis, and can keep excellent cell survival rate when being used for freezing preservation solution.
Drawings
FIG. 1: electron micrograph of the ice crystal growth inhibition activity of GDL-L-Thr and statistical plot of the size of the ice crystal.
FIG. 2 is a schematic diagram: the GDL-L-Thr modifies the appearance effect of ice crystals in pure water.
FIG. 3: an electron micrograph of the ice crystal growth inhibitory activity of the TR short chain peptide prepared in example 2 and a statistical plot of ice crystal size.
FIG. 4 is a schematic view of: the TR short-chain peptide prepared in the example 2 modifies the morphological effect of ice crystals in pure water.
FIG. 5 is a schematic view of: example 7 peptoids R-COOH, R-CH 3 And R-CH 2 CH 3 Inhibiting the growth activity of ice crystal.
FIG. 6: peptoid (A) R-COOH, (B) R-CH 3 And (C) R-CH 2 CH 3 Modifying the appearance effect of the ice crystals in the pure water.
FIG. 7: photographs of sections of fresh, unfrozen ovarian organs from 3-day-old mice.
FIG. 8: the cryopreservation solution of comparative example 4 is used to cryopreserve a photograph of a thawed section of an intact ovarian organ.
FIG. 9: photographs of thawed sections of intact ovarian organs were cryopreserved using the cryopreservation solution of example 6.
FIG. 10: photographs of sections of fresh unfrozen ovarian tissue from sexually mature mice.
FIG. 11: photograph of frozen ovarian tissue section thawed by the cryopreservation solution of comparative example 5.
FIG. 12: photograph of thawed ovarian tissue section was cryopreserved using the cryopreservation solution of example 7.
Detailed Description
The preparation method of the present invention will be described in further detail with reference to specific examples. It is to be understood that the following examples are only illustrative and explanatory of the present invention and should not be construed as limiting the scope of the present invention. All the techniques realized based on the above-mentioned contents of the present invention are covered in the protection scope of the present invention.
The experimental methods used in the following examples are all conventional methods unless otherwise specified; reagents, materials and the like used in the following examples are commercially available unless otherwise specified.
EXAMPLE 1 Synthesis of Compound of formula (1)
(1) 2-Chlorotrityl Chloride Resin (2-Chlorotrityl Chloride Resin) was placed in a reaction tube, and DCM (20 mL g) was added -1 ) And shaken for 30 minutes. And (3) filtering the sand core to remove the solvent, adding three times of molar excess Fmoc-L-Pro-OH, adding 8 times of molar excess DIEA, finally adding DMF to dissolve, and oscillating for 30 minutes. Methanol was capped for 30 minutes.
(2) The solvent DMF was removed and 20% piperidine/DMF solution (10 mL g) was added -1 ) After 5 minutes, the solvent was removed and a further 20% piperidine/DMF solution (10 mL g) -1 ) After 15 minutes the piperidine solution was removed. Taking a small amount of resin, washing the resin with ethanol for three times, adding ninhydrin reagent, heating the resin for 5 minutes at 105-110 ℃, and taking a positive reaction when the color turns dark blue.
(3) The product obtained from the above reaction was washed sequentially with DMF (15 mL g) -1 Twice, methanol (15 mL g) -1 Twice) and DMF (15 mL g) -1 Twice), adding Fmoc-L-Thr (tBu) -OH dissolved by using DMF as little as possible into a reaction tube; two-fold excess, HBTU two-fold excess. Immediately thereafter, an 8-fold excess of DIEA was added and reacted for 30 minutes.
(4) After the solution is pumped out, a small amount of resin is taken out, washed by ethanol for three times, added with ninhydrin reagent, heated for 5 minutes at 105-110 ℃, and colorless is negative reaction, namely the reaction is complete.
(5) The product obtained from the above reaction was washed sequentially with DMF (15 mL g) -1 Twice, methanol (15 mL g) -1 Twice) and DMF (15 mL g) -1 Twice) washing followed by removal of the solvent and addition of 20% piperidine/DMF solution (10 mL g) -1 ) After 5 minutes, the solvent was removed and a further 20% piperidine/DMF solution (10 mL g) -1 ) And after 15 minutes, removing the piperidine solution, taking a small amount of resin, washing the resin by using ethanol, adding a ninhydrin reagent, heating the resin at 105-110 ℃ for 5 minutes, and turning dark blue to be a positive reaction.
(6) The product obtained from the above reaction was washed sequentially with DMF (15 mL g-1, twice) and methanol (15 mL g) -1 Twice) and DCM (15 mL g) -1 Twice) after washing, the resin was drained.
(7) Using a cutting fluid (15 mL g) -1 And TFA: water: EDT (electro-thermal transfer coating): tis = 95. And drying the cutting fluid by using nitrogen, and freeze-drying to obtain a crude polypeptide product.
(8) Purification of the polypeptide by HPLC and desalting, HPLC: tR =6.1mins (purification column model: kromasil 100-5c18,4.6mm 250mm; gradient eluent: 0.1% tfa acetonitrile solution and 0.1% aqueous tfa solution, 0 mins-1. And (4) freeze-drying the purified solution to obtain a finished product L-Thr-L-Pro (marked as TP). The yield was about 80%. The mass spectrum identification 217.3 is [ M + H ] +.
EXAMPLE 2 Synthesis of Compound of formula (2)
(1) The 2-chlorotrityl chloride resin was placed in a reaction tube and DCM (20 mL g) was added -1 ) And shaken for 30 minutes. And (3) filtering the sand core to remove the solvent by suction, adding three times of molar excess Fmoc-L-Thr (tBu) -OH, adding 8 times of molar excess DIEA, finally adding DMF to dissolve, and oscillating for 30 minutes. Methanol was capped for 30 minutes.
(2) The solvent DMF was removed and 20% piperidine/DMF solution (10 mL g) was added -1 ) After 5 minutes, the solvent was removed and a further 20% piperidine/DMF solution (10 mL g) -1 ) After 15 minutes the piperidine solution was removed. Taking a small amount of resin, washing the resin with ethanol for three times, adding ninhydrin reagent, heating the resin for 5 minutes at 105-110 ℃, and taking a positive reaction when the color turns dark blue.
(3) The product obtained in the above reaction was washed successively with DMF (15 mL g) -1 Twice, methanol (15 mL g) -1 Twice) and DMF (15 mL g) -1 Twice), adding Fmoc-Arg (Pbf) -OH dissolved by using DMF as little as possible into a reaction tube; two-fold excess, HBTU two-fold excess. Immediately thereafter, 8-fold excess of DIEA was added and the reaction was carried out for 30 minutes.
(4) After the solution is pumped out, a small amount of resin is taken out, washed by ethanol for three times, added with ninhydrin reagent, heated for 5 minutes at 105-110 ℃, and colorless is negative reaction, namely the reaction is complete.
(5) The product obtained in the above reaction was washed successively with DMF (15 mL g) -1 Twice, methanol (15 mL g) -1 Twice) and DMF (15 mL g) -1 Two times) washing followed by removal of the solvent and addition of 20% piperidine/DMF solution (10 mL g) -1 ) After 5 minutes, the solvent was removed and a further 20% piperidine/DMF solution (10 mL g) -1 ) After 15 minutes, removing the piperidine solution, taking a small amount of resin, cleaning the resin by using ethanol, adding ninhydrin reagent, heating the resin for 5 minutes at 105-110 ℃, and turning dark blue to be a positive reaction.
(6) The product obtained in the above reaction was washed successively with DMF (15 mL g) -1 Twice, methanol (15 mL g) -1 Twice) and DCM (15 mL g) -1 Twice) after washing, the resin was drained.
(7) Using a cutting fluid (15 mL g) -1 And TFA: water: EDT (electric discharge machining): tis = 95. And drying the cutting fluid by using nitrogen, and freeze-drying to obtain a crude polypeptide product.
(8) Purification of the polypeptide by HPLC and desalting, HPLC: tR =4.8mins (purification column model: kromasil 100-5c18,4.6mm 250mm; gradient eluent: 0.1% tfa acetonitrile solution and 0.1% tfa aqueous solution, 0 mins-1. And (4) freeze-drying the purified solution to obtain a finished product L-Thr-L-Arg (TR). The yield was about 80%. Mass spectrometry identification 276.2 is [ M + H ] +.
EXAMPLE 3 Synthesis of Compound of formula (3)
(1) The 2-chlorotrityl chloride resin was placed in a reaction tube and DCM (20 mL g) was added -1 ) And shaken for 30 minutes. Filtering the sand core to remove the solvent, adding three times of molesFmoc-L-Thr (tBu) -OH in molar excess, DIEA in 8-fold molar excess and finally DMF were added to dissolve and shaken for 30 min. Methanol was capped for 30 minutes.
(2) The solvent DMF was removed and 20% piperidine/DMF solution (10 mL g) was added -1 ) After 5 minutes, the solvent was removed and a further 20% piperidine/DMF solution (10 mL g) -1 ) After 15 minutes the piperidine solution was removed. Taking a small amount of resin, washing the resin with ethanol for three times, adding ninhydrin reagent, heating the resin for 5 minutes at 105-110 ℃, and taking a positive reaction when the color turns dark blue.
(3) The product obtained from the above reaction was washed sequentially with DMF (15 mL g) -1 Twice, methanol (15 mL g) -1 Twice) and DMF (15 mL g) -1 Twice), adding Fmoc-Arg (Pbf) -OH dissolved by using DMF as little as possible into a reaction tube; double excess, HBTU double excess. Immediately thereafter, 8-fold excess of DIEA was added and the reaction was carried out for 30 minutes.
(4) After the solution is pumped out, a small amount of resin is taken out, washed by ethanol for three times, added with ninhydrin reagent, heated for 5 minutes at 105-110 ℃, and colorless is negative reaction, namely the reaction is complete.
(5) The product obtained in the above reaction was washed successively with DMF (15 mL g) -1 Twice, methanol (15 mL g) -1 Twice) and DMF (15 mL g) -1 Twice) washing followed by removal of the solvent and addition of 20% piperidine/DMF solution (10 mL g) -1 ) After 5 minutes, the solvent was removed and a further 20% piperidine/DMF solution (10 mL g) -1 ) And after 15 minutes, removing the piperidine solution, taking a small amount of resin, washing the resin by using ethanol, adding a ninhydrin reagent, heating the resin at 105-110 ℃ for 5 minutes, and turning dark blue to be a positive reaction.
(6) The product obtained in the above reaction was washed successively with DMF (15 mL g) -1 Twice, methanol (15 mL g) -1 Twice) and DMF (15 mL g) -1 Twice), and then the resin is drained after cleaning.
(7) Repeating the operations (3) to (5), and linking the amino acid Fmoc-L-Thr (tBu) -OH. The reaction product was washed sequentially with DMF (15 mL g) -1 Twice, methanol (15 mL g) -1 Twice) and DCM (15 mL g) -1 Twice) after washing, the resin was drained.
(8) Using cutting fluids(15mL g -1 And TFA: water: EDT (electro-thermal transfer coating): tis = 95. And drying the cutting fluid by using nitrogen, and freeze-drying to obtain a crude polypeptide product.
(9) Purification of the polypeptide by HPLC and desalting, HPLC: tR =3.9mins (purification column model: kromasil 100-5c18,4.6mm 250mm; gradient eluent: 0.1% tfa acetonitrile solution and 0.1% tfa aqueous solution, 0 mins-1. And freeze-drying the purified solution to obtain a finished product L-Thr-L-Arg-L-Thr (TRT). The yield was about 75%. Mass spectrometry identification 377.4 is [ M + H ] +.
EXAMPLE 4 Synthesis of Compound of formula (4)
(1) The 2-chlorotrityl chloride resin was placed in a reaction tube and DCM (20 mL g) was added -1 ) And shaken for 30 minutes. And (3) filtering the sand core to remove the solvent by suction, adding three times of molar excess Fmoc-L-Thr (tBu) -OH, adding 8 times of molar excess DIEA, finally adding DMF to dissolve, and oscillating for 30 minutes. Methanol was capped for 30 minutes.
(2) The solvent DMF was removed and 20% piperidine/DMF solution (10 mL g) was added -1 ) After 5 minutes, the solvent was removed and a further 20% piperidine/DMF solution (10 mL g) -1 ) After 15 minutes the piperidine solution was removed. Taking a small amount of resin, washing the resin with ethanol for three times, adding ninhydrin reagent, heating the resin for 5 minutes at 105-110 ℃, and turning dark blue to be a positive reaction.
(3) The product obtained in the above reaction was washed successively with DMF (15 mL g) -1 Twice, methanol (15 mL g) -1 Twice) and DMF (15 mL g) -1 Twice), adding Fmoc-L-Pro-OH dissolved by using DMF as little as possible into a reaction tube; double excess, HBTU double excess. Immediately thereafter, 8-fold excess of DIEA was added and the reaction was carried out for 30 minutes.
(4) After the solution is pumped out, a small amount of resin is taken out, washed by ethanol for three times, added with ninhydrin reagent, heated for 5 minutes at 105-110 ℃, and colorless is negative reaction, namely the reaction is complete.
(5) The product obtained in the above reaction was washed successively with DMF (15 mL g) -1 Twice, methanol (15 mL g) -1 Twice) and DMF (15 mL g) -1 Twice) washing followed by removal of the solvent and addition of 20% piperidine/DMF solution (10 mL g) -1 ) 5 points ofAfter a while, the solvent was removed and 20% piperidine/DMF solution (10 mL g) was added -1 ) After 15 minutes, removing the piperidine solution, taking a small amount of resin, cleaning the resin by using ethanol, adding ninhydrin reagent, heating the resin for 5 minutes at 105-110 ℃, and turning dark blue to be a positive reaction.
(6) The product obtained in the above reaction was washed successively with DMF (15 mL g) -1 Twice, methanol (15 mL g) -1 Twice) and DMF (15 mL g) -1 Twice) after washing, the resin was drained.
(7) Repeating the operations (3) to (5), and linking the amino acid Fmoc-L-Thr (tBu) -OH. The reaction product was washed sequentially with DMF (15 mL g) -1 Twice, methanol (15 mL g) -1 Twice) and DCM (15 mL g) -1 Twice) after washing, the resin was drained.
(8) The product was cleaved for 90 minutes using cleavage solution (15 mL g-1, tfa: water: EDT: tis = 95. And drying the cutting fluid by using nitrogen, and freeze-drying to obtain a crude polypeptide product.
(9) Purification of the polypeptide by HPLC and desalting, HPLC: tR =7.6mins (purification column model: kromasil 100-5c18,4.6mm 250mm; gradient eluent: 0.1% tfa acetonitrile solution and 0.1% aqueous tfa solution, 0 mins-1. And (4) freeze-drying the purified solution to obtain a finished product L-Thr-L-Pro-L-Thr (TPT). The yield was about 70%. Mass spectrum identification 318.3 is [ M + H ] +
EXAMPLE 5 Synthesis of Compound of formula (5)
(1) The 2-chlorotrityl chloride resin was placed in a reaction tube and DCM (20 mL g) was added -1 ) And shaken for 30 minutes. And (3) filtering the sand core to remove the solvent by suction, adding three times of molar excess Fmoc-L-Thr (tBu) -OH, adding 8 times of molar excess DIEA, finally adding DMF to dissolve, and oscillating for 30 minutes. Methanol was capped for 30 minutes.
(2) The solvent DMF was removed and 20% piperidine/DMF solution (10 mL g) was added -1 ) After 5 minutes, the solvent was removed and a further 20% piperidine/DMF solution (10 mL g) -1 ) After 15 minutes the piperidine solution was removed. Taking a small amount of resin, washing the resin with ethanol for three times, adding ninhydrin reagent, heating the resin for 5 minutes at 105-110 ℃, and taking a positive reaction when the color turns dark blue.
(3) The product obtained by the reaction is usedDMF (15 mL g) -1 Twice, methanol (15 mL g) -1 Twice) and DMF (15 mL g) -1 Twice), adding Fmoc-L-Ala-OH dissolved by using DMF as little as possible into a reaction tube; two-fold excess, HBTU two-fold excess. Immediately thereafter, an 8-fold excess of DIEA was added and reacted for 30 minutes.
(4) After the solution is pumped out, a small amount of resin is taken out, washed by ethanol for three times, added with ninhydrin reagent, heated for 5 minutes at 105-110 ℃, and colorless is negative reaction, namely the reaction is complete.
(5) The product obtained from the above reaction was washed sequentially with DMF (15 mL g) -1 Twice, methanol (15 mL g) -1 Twice) and DMF (15 mL g) -1 Twice) washing followed by removal of the solvent and addition of 20% piperidine/DMF solution (10 mL g) -1 ) After 5 minutes, the solvent was removed and a further 20% piperidine/DMF solution (10 mL g) -1 ) After 15 minutes, removing the piperidine solution, taking a small amount of resin, cleaning the resin by using ethanol, adding ninhydrin reagent, heating the resin for 5 minutes at 105-110 ℃, and turning dark blue to be a positive reaction.
(6) The product obtained from the above reaction was washed sequentially with DMF (15 mL g) -1 Twice, methanol (15 mL g) -1 Twice) and DMF (15 mL g) -1 Twice) after washing, the resin was drained.
(7) Repeating the operations (3) to (5) to link the amino acid Fmoc-L-Ala-OH. The reaction product was washed successively with DMF (15 mL g) -1 Twice, methanol (15 mL g) -1 Twice) and DCM (15 mL g) -1 Twice), and then the resin is drained after cleaning.
(8) Using a cutting fluid (15 mL g) -1 And TFA: water: EDT (electro-thermal transfer coating): tis = 95. And drying the cutting fluid by using nitrogen, and freeze-drying to obtain a crude polypeptide product.
(9) Purification of the polypeptide by HPLC and desalting, HPLC: tR =7.9mins (purification column model: kromasil 100-5c18,4.6mm 250mm; gradient eluent: 0.1% tfa acetonitrile solution in acetonitrile and 0.1% tfa aqueous solution in water, 0 mins-1. And (4) freeze-drying the purified solution to obtain a finished product L-Ala-L-Ala-L-Thr (AAT). The yield was about 70%. The mass spectrum identification 260.1 is [ M-8H ] +.
EXAMPLE 6 Synthesis of Compound of formula (6)
(1) 1.0g (0.56 mol) of Gluconolactone (GDL) and 0.56mol of L-Thr were subjected to solid-phase synthesis to prepare GDL-L-Thr.
(2) HPLC purification, HPLC: tR =3.4mins (purification column model: kromasil 100-5C18SHIMADZU Intertsil ODS-SP (4.6 mm 250mm x 5 μ M); gradient eluent: 0.1% TFA acetonitrile solution and 0.1% aqueous TFA solution, 0.01-20mins-1 99, 20-30mins-21, 30-40mins-100, 0,40-50 mins-1.
The GDL-L-Thr prepared by the solid-phase synthesis method has higher purity and is more beneficial to product separation, and experimental results show that the GDL-L-Thr prepared by the solid-phase synthesis method has higher purity and keeps good capability of inhibiting ice crystal growth (figure 1).
EXAMPLE 7 Synthesis of Compound of formula (9)
(1) The DCM solution of dichlodimethylisilane was poured into the polypeptide synthesis tube and after standing for 30 minutes the tube was air dried for use.
(2) 100mg of the resin was taken in a synthesis tube, 2mL of DMF was added, nitrogen was introduced, the resin was swollen for 10 minutes, and suction filtration was carried out.
(3) 1mL of 4-methylpiperidine/DMF solution is added for deprotection, removed after 5 minutes, 1mL of 4-methylpiperidine/DMF solution is added, removed after 15 minutes, bubbled and filtered off.
(4) DMF was washed 5 times, bubbled and filtered with suction.
(5) 2M,0.5mL of bromoacetic acid/DMF solution, N, N-diisopropylcarbodiimide/DMF solution were added in this order, and bubbling was carried out for 20 minutes, followed by suction filtration and 3-time washing with DMF.
(6) 1M,1mL of a primary amine/DMF solution was added, bubbled for 30min, rinsed with DMF, and rinsed with dichloromethane (× 3).
(7) Repeating steps (5) and (6) until the desired molecular weight is reached.
(8) Adding 4mL of cracking solution, fully shaking uniformly, introducing nitrogen to blow dry, finally freeze-drying, and purifying to obtain a final finished product.
(9) R is-CH 3 ,-CH 2 CH 3 and-CH 2 CH 2 Class of COOHA peptide. Mass spectrometric identification 444.6 for R as-CH 3 Of [ M + H] + 528.8 is R is-CH 2 CH 3 Of [ M + H] + 792.1 is that R is-CH 2 CH 2 [ M + H ] of COOH] +
Figure BDA0002409683710000181
Dissolving and dispersing a sample into a DPBS solution by adopting a sputtering freezing method for inhibiting the recrystallization of ice crystals (IRI), dripping 10-30 mu L of the solution onto the surface of a clean silicon wafer precooled at minus 60 ℃ at a height of more than 1.0m, heating to minus 6 ℃ at a speed of 20 ℃/min by using a cold-hot table, annealing for 30mins at the temperature, observing and recording the size of the ice crystals by using a polarizing microscope and a high-speed camera, and sealing the cold-hot table. Each sample was repeated at least three times, and the ice crystal size was counted using a Nano Measurer 1.2 with the error of the statistical result being the standard deviation.
Ice crystal morphology (DIS) observation and Thermal Hysteresis (TH) measurement Using a nanoliter osmometer, a capillary was first melted with an alcohol burner external flame and simultaneously stretched to produce a capillary of very fine pore size, which was connected to a microsyringe. The immersion lens oil with higher viscosity is injected into the micron-aperture wafer, and the water solution dissolved with the material is injected into the micropores by using a microsyringe. The liquid drop is quickly frozen by controlling the temperature, and slowly heated to obtain the single crystal ice, the temperature is slowly reduced with the precision of 0.01 ℃, and the appearance of the ice crystal is observed by utilizing a microscope equipped with a high-speed camera and a TH test is carried out.
IRI activity test was performed on 20. Mu.L of the DPBS solution of TR prepared in the above example 2 by the "sputter freezing method". The measured maximum ice crystal size (%) relative to DPBS is shown in fig. 3. The largest ice crystal size of TR bound by chemical bonds is significantly smaller than the largest ice crystal size of DPBS solutions of arginine and threonine at the same concentration.
The deionized water solution of TR prepared in the above example 2 was taken, and the observation of the morphology of ice crystals by nanoliter revealed that TR had a weak effect of modifying the morphology of ice crystals (supercooling degree-0.1 ℃ C., 0.4-0.01 ℃ C.), as shown in FIG. 4. And no thermal hysteresis was measured.
IRI activity test was performed on 20. Mu.L of the DPBS solution of GDL-L-Thr prepared in the above example 6 by "sputter freezing". The measured maximum ice crystal size (%) relative to DPBS is shown in fig. 1. The maximum ice crystal size of GDL-L-Thr bound by chemical bond is significantly smaller than that of DPBS solution of GDL and DPBS solution of L-Thr at the same concentration and smaller than that of DPBS solution mixed with GDL and L-Thr at the same concentration.
The deionized water solution of GDL-L-Thr prepared in the above example 6 was taken, and the observation of the crystal morphology using nanoliters revealed that GDL-L-Thr had a weak effect of modifying the crystal morphology (supercooling degree of-0.1 ℃ C., 0.4-0.01 ℃ C.), as shown in FIG. 2. And no thermal hysteresis was measured.
IRI activity test was performed using "sputter freezing method" on 20 μ L of DPBS solution of the compound prepared in the above example 7. The maximum ice crystal size (%) relative to DPBS was measured as shown in fig. 5.
Taking deionized water solution of the three peptoids prepared in the above example 7, utilizing nanoliter to perform ice crystal morphology observation to find that R is-CH 3 and-CH 2 CH 3 The peptoid has more obvious effect of modifying the appearance of ice crystals, and R is-CH 2 CH 2 The morphology of COOH peptoids without modification of ice crystal morphology (supercooling degree-0.1 ℃ and-0.4-0.01 ℃) is shown in FIG. 6, and thermal hysteresis is not detected in any of the three peptoids.
The results show that the prepared peptide compound has the effects of inhibiting the growth activity of ice crystals and modifying the appearance of the ice crystals, has no thermal hysteresis, can realize the effect of controlling the growth of the ice crystals, and can be used for freezing storage.
Example 8 oocyte and embryo cryopreservation experiments
(1) The TR synthesized in example 2 and the TPT synthesized in example 4 were used to prepare a cryopreservation solution as follows:
cryopreservation liquid 1: dissolving 28g of TR in 25mL of PBS by ultrasonic treatment in a total volume of 100mL, and adjusting the pH to 7.0 to obtain a solution 1; ultrasonically dissolving 0.05mol of sucrose in 25mL of DPBS, sequentially adding 10mL of glycol and 7.5mL of DMSO to obtain a solution 2 after the sucrose is completely dissolved, uniformly mixing the two solutions after the solution 1 and the solution 2 are returned to room temperature, adjusting the pH value, fixing the volume to 80% of the total volume by adopting the DPBS, and finally adding 20mL of serum before use.
Cryopreservation liquid 2: dissolving 28g of TPT in 25mL of PBS by ultrasonic treatment in a total volume of 100mL, and adjusting the pH to 7.0 to obtain a solution 1; ultrasonically dissolving 0.05mol of sucrose in 25mLDPBS, sequentially adding 10mL of ethylene glycol and 7.5mLDMSO to obtain a solution 2 after the sucrose is completely dissolved, uniformly mixing the two solutions after the solution 1 and the solution 2 are restored to room temperature, adjusting the pH value, fixing the volume to 80% of the total volume by adopting DPBS, and finally adding 20mL of serum before use.
Comparative example 1:
cryopreservation liquid a: each 1mL contained 15% (v/v) DMSO,15% (v/v) ethylene glycol, 20% (v/v) serum, 0.5M sucrose, and the balance DPBS.
(2) Balancing liquid and unfreezing liquid:
freezing equilibrium liquid A: every 1mL of the mixture contains 7.5% (v/v) of DMSO,7.5% (v/v) of glycol, 20% (v/v) of serum and the balance of DPBS;
thawing solution B: thawing solution I (containing 1.0mol L) -1 Sucrose, 20% serum, balance DPBS); thawing solution II (containing 0.5mol L) -1 Sucrose, 20% serum, balance DPBS); thawing solution III (containing 0.25mol L) -1 Sucrose, 20% serum, balance DPBS); thawing solution IV (20% serum, balance DPBS).
(3) The cryopreservation liquid and the formula of the comparative example are used for preserving the oocyte of the mouse
Cryopreservation of mouse oocytes was performed using cryopreservation solutions 1 and 2 prepared in the above steps, and cryopreservation solution and cryoequilibrium solution of comparative example 1. The oocyte cryopreservation method comprises the steps of firstly placing oocytes in a cryopreservation liquid for balancing for 5 minutes, then placing the oocytes in the prepared cryopreservation liquid for 50 seconds, placing the oocytes which are balanced in the cryopreservation liquid on a freezing carrying rod, then quickly putting the oocytes into liquid nitrogen at (-196 ℃) and continuing to preserve the oocytes after the carrying rod is sealed; when unfreezing is carried out, the frozen oocytes are placed in the thawing solution I at 37 ℃ to be balanced for 5 minutes, and then are balanced in the thawing solutions II to IV for 3 minutes respectively in sequence; the number of viable cells was observed after culturing the thawed oocytes for 2 hours. After thawing, the oocytes were cultured for 2 hours to calculate the survival rate of the oocytes (Table 1).
(4) Mouse embryo preservation by using the cryopreservation liquid and the formula of the comparative example
The embryos of the mice are subjected to cryopreservation by using the cryopreservation liquids 1 and 2 prepared in the above steps, and the cryopreservation liquid and the frozen equilibrium liquid in the comparative example 1. The embryo cryopreservation method comprises the steps of firstly placing the embryo in a cryopreservation balance liquid for 5 minutes, then placing the embryo in the prepared cryopreservation liquid for 50 seconds, placing the embryo balanced in the cryopreservation liquid on a freezing carrying rod, then quickly putting the embryo into liquid nitrogen at (-196 ℃) and continuing to preserve after sealing the carrying rod; when unfreezing, placing the frozen embryo in the unfreezing liquid I at 37 ℃ for balancing for 5 minutes, and then balancing in the unfreezing liquids II-IV for 3 minutes respectively in sequence; the thawed embryos were cultured for 2 hours before observing the number of viable cells. After thawing, embryos were cultured for 2 hours to calculate survival (table 2).
In the examples, the survival rate is the average survival rate of 3-12 repeated experiments.
TABLE 1 cryopreservation survival rates of oocytes from mice
Number of Balancing liquid Refrigerating fluid Thawing solution Total number of frozen eggs Survival rate after 2 hours
Comparative example 1 A Cryopreservation liquid a B 146 95%
Application example 1 A Cryopreservation liquid 1 B 93 96.2%
Application example 2 A Cryopreservation liquid 2 B 48 90%
TABLE 2 survival rate of mouse embryos by cryopreservation
Number of Balancing liquid Refrigerating fluid Thawing solution Total number of embryos Survival rate after 2 hours
Comparative example 2 A Cryopreservation liquid a B 38 94.3%
Application example 3 A Cryopreservation liquid 1 B 41 95.9%
The data in tables 1 and 2 show that the polypeptides of the present invention are used for cryopreservation of oocytes and embryos, and that the survival rate of oocytes and embryos of the existing commercial cryopreservation solution (DMSO content 15%) can be achieved by adding only a small amount of DMSO (7.5%), and the data in application example 1 and application example 3 show that TR polypeptides have more excellent effects for cryopreservation of oocytes and embryos.
Example 9: stem cell cryopreservation
Cryopreservation liquid 1: dissolving 28g of TR in 25mL of PBS by ultrasonic treatment in a total volume of 100mL, and adjusting the pH to 7.0 to obtain a solution 1; ultrasonically dissolving 0.05mol of sucrose in 25mL of DPBS, sequentially adding 10mL of ethylene glycol and 7.5mL of DMSO to obtain a solution 2 after the sucrose is completely dissolved, uniformly mixing the two solutions after the solution 1 and the solution 2 are returned to room temperature, adjusting the pH value, fixing the volume to 80% of the total volume by adopting the DPBS, and finally adding 20mL of serum before use.
Cryopreservation liquid 3: dissolving 28g of TR in 25mL of PBS by ultrasonic treatment in a total volume of 100mL, and adjusting the pH to 7.0 to obtain a solution 1; ultrasonically dissolving 0.05mol of sucrose in 25mL of DPBS, adding 10mL of ethylene glycol to obtain a solution 2 after the sucrose is completely dissolved, uniformly mixing the two solutions when the solution 1 and the solution 2 are restored to room temperature, adjusting the pH value, fixing the volume to 80% of the total volume by using the DPBS, and finally adding 20mL of serum before use.
Comparative example 2:
cryopreservation liquid b: each 1mL of the culture medium contained 10% (v/v) DMSO,15% (v/v) serum, and the balance a-MEM (USA, invitrogen, C12571500 BT).
The cryopreservation solution is adopted to perform cryopreservation on of the human umbilical cord mesenchymal stem cells according to the scheme in table 3. The cryopreservation method of the human umbilical cord stem cells is specifically a microdroplet method: digesting the human umbilical cord mesenchymal stem cells on the culture dish for 2 minutes by using 25% pancreatin, putting the same volume of culture solution (10% of FBS + a-MEM culture medium), gently blowing and beating until the stem cells are completely fallen off, adding a 1.5ml centrifuge tube, centrifuging for 5 minutes at 1000rmp, discarding supernatant, separating the cells from the culture medium, adding 10uL of freezing solution to the bottom of the centrifuge tube, gently blowing and beating to disperse stem cell clusters, placing the 10uL of freezing solution with the stem cells on a freezing slide, placing the freezing slide on liquid nitrogen (-196 ℃) and freezing. When thawing, the frozen rods with cells and freezing fluid were placed directly in 37 ℃ a-MEM medium and thawed. After thawing, trypan blue staining was used to check for viability and the number of cells was counted using the instrument JIMBIO-FIL, viability = number of viable cells/total number of cells (see table 3).
TABLE 3 cryopreservation survival rate of human umbilical cord mesenchymal stem cells
Figure BDA0002409683710000221
Figure BDA0002409683710000231
As can be seen from the results shown in table 3, when the cryopreservation solution of the present invention is added with no DMSO or only a small amount of DMSO (7.5%), the cell survival rate equivalent to that of the cryopreservation solution added with 10% DMSO in the prior art can be achieved, the amount of DMSO used is greatly reduced, the damage and toxicity of DMSO to cells are reduced, and the generation stability and cell activity of the stem cells after freezing can be greatly improved.
Example 10: cryopreservation of ovarian organs and ovarian tissues
Cryopreservation liquid 1: dissolving 28g of TR in 25mL of PBS by ultrasonic treatment in a total volume of 100mL, and adjusting the pH to 7.0 to obtain a solution 1; ultrasonically dissolving 0.05mol of sucrose in 25mL of DPBS, sequentially adding 10mL of glycol and 7.5mL of DMSO to obtain a solution 2 after the sucrose is completely dissolved, uniformly mixing the two solutions after the solution 1 and the solution 2 are returned to room temperature, adjusting the pH value, fixing the volume to 80% of the total volume by adopting the DPBS, and finally adding 20mL of serum before use.
Cryopreservation liquid a: each 1mL contained 15% (v/v) DMSO,15% (v/v) ethylene glycol, 20% (v/v) serum, 0.5M sucrose, and the balance DPBS.
Freezing equilibrium liquid A: every 1mL of the reagent contains 7.5% (v/v) DMSO,7.5% (v/v) ethylene glycol, 20% (v/v) serum and the balance of DPBS;
thawing solution B: thawing solution I (containing 1.0mol L) -1 Sucrose, 20% serum, balance DPBS); thawing solution II (containing 0.5mol L) -1 Sucrose, 20% serum, balance DPBS); thawing solution III (containing 0.25mol L) -1 Sucrose, 20% serum, balance DPBS); thawing solution IV (20% serum, balance DPBS).
The cryopreservation solution, the cryopreservation equilibrium solution of the comparative example and the cryopreservation solution are adopted to carry out cryopreservation on the complete ovarian organ of the mouse within 3 days of newborn and the ovarian tissue section of the sexually mature mouse according to the schemes in tables 4 and 5.
And (3) balancing the whole ovarian organ or ovarian tissue section in a freezing balancing solution at room temperature for 25 minutes, then placing the whole ovarian organ or ovarian tissue section in the prepared freezing preservation solution for 15 minutes, then placing the whole ovarian organ or ovarian tissue section on a freezing carrying rod, and putting the whole ovarian organ or ovarian tissue section in liquid nitrogen for preservation. After thawing, the whole ovarian organ or ovarian tissue section is placed in culture medium (10% FBS + a-MEM) and then subjected to 5% CO at 37 ℃ 2 Fixing with 4% paraformaldehyde after resuscitation in incubator for 2 hrFig. 7-12 show the results of morphology observation by paraffin embedding and HE staining, fig. 7 is a photograph of a fresh unfrozen section of an ovarian organ, and fig. 10 is a photograph of a fresh unfrozen section of an ovarian tissue.
TABLE 4 ovarian organ cryopreservation protocol
Number of Balancing liquid Cryopreservation liquid Thawing solution Form of
Application example 6 A Cryopreservation liquid 1 B FIG. 9
Comparative example 4 A Cryopreservation liquid a B FIG. 8
TABLE 5 ovarian tissue cryopreservation protocol
Number of Balancing liquid Cryopreservation liquid Thawing solution Form of
Application example 7 A Cryopreservation liquid 1 B FIG. 12
Comparative example 5 A Cryopreservation liquid a B FIG. 11
As can be seen from fig. 7-9, the thawed section photographs of the ovarian organs preserved by freezing using application example 6 show that the ovarian follicles are relatively intact, the interstitial structures are relatively intact, the cytoplasm of the cells is homogeneous, the light staining is relatively more, and the nuclei of the cells are shriveled and deeply stained relatively less, compared with comparative example 4 using the bionic ice-control material without adding peptides and fresh unfrozen ovarian organs; the vessel wall structure of the blood vessel is complete, the vessel cavity collapse is less, the cytoplasm of endothelial cells is homogeneous, the light staining is relatively more, and the nucleus is shriveled and the deep staining is relatively less. It can be seen that the application example 6 group had better cryopreservation effect on ovarian organs.
As can be seen from FIGS. 10 to 12, the protocol of example 7 was applied to the ovarian tissue of the adult mice which had not been frozen, and the follicles and antrum follicles in the growth phase were relatively intact as compared with the ovarian tissue of the fresh non-frozen adult mice, and it was found that the cryopreservation solution of the present invention was also effective in cryopreservation of the ovarian tissue as compared with the prior art.
Therefore, the cryopreservation liquid prepared by taking the peptide bionic ice control material as the main component can be simultaneously suitable for cryopreservation of oocytes, embryos, stem cells, reproductive organs and tissues, can achieve good cell survival rate and biological activity, and has a good effect of cryopreservation of stem cells.
The embodiments of the present invention have been described above. However, the present invention is not limited to the above embodiment. Any modification, equivalent replacement, or improvement made within the spirit and principle of the present invention should be included in the protection scope of the present invention.

Claims (27)

1. The cryopreservation liquid contains 0.1-50g of peptide compound L-Thr-L-Arg and/or L-Thr-L-Pro-L-Thr in terms of volume of every 100mL of cryopreservation liquid.
2. The use according to claim 1, wherein the cryopreservation solution further comprises one or more of PVA and polyamino acid.
3. The use according to any one of claims 1-2, wherein the cryopreservation solution further comprises serum or human serum albumin or bovine serum albumin.
4. The use according to claim 3, wherein the cryopreservation solution comprises 0.1-50g of said peptidic compounds L-Thr-L-Arg and/or L-Thr-L-Pro-L-Thr,0-6.0 g of PVA,0-9.0g of polyamino acid, 0-15mL of DMSO,5-45mL of polyol, 0.1-1.0mol L per 100mL -1 0-30mL of serum or human serum albumin or bovine serum albumin, and the balance of buffer solution.
5. The use according to claim 4, wherein the cryopreservation solution contains 0.1-15mL of DMSO per 100 mL.
6. The use according to claim 5, wherein the cryopreservation solution contains 0.1-10mL of DMSO.
7. The use according to claim 4, wherein the cryopreservation fluid contains 0.1 to 6.0g of PVA.
8. Use according to claim 7, wherein the cryopreservation solution contains 0.1 to 4.0g of PVA.
9. The use according to claim 4, wherein the cryopreservation solution contains 5.0-30mL of serum or human serum albumin or bovine serum albumin.
10. The use according to claim 9, wherein the cryopreservation solution contains 5.0-20mL of serum or human or bovine serum albumin.
11. Use according to claim 4, the PVA being selected from one or a combination of two or more of isotactic PVA, syndiotactic PVA and atactic PVA.
12. Use according to claim 11, the PVA having a syndiotacticity of from 15% to 60%.
13. Use according to claim 12, the PVA having a syndiotacticity of from 50% to 60%.
14. Use according to claim 13, the PVA having a syndiotacticity of from 50% to 55%.
15. The use according to claim 2, wherein the polyamino acid is selected from at least one homopolymer of lysine, arginine, proline, threonine, histidine, etc., or a copolymer of two or more amino acids.
16. Use according to claim 4, the polyol being a polyol having from 2 to 5 carbon atoms.
17. Use according to claim 16, wherein the polyol is any one of ethylene glycol, propylene glycol and glycerol.
18. The use according to claim 4, wherein the water-soluble saccharide is at least one of a non-reducing disaccharide, a water-soluble polysaccharide, and a sugar anhydride.
19. Use according to claim 18, said water-soluble sugar being selected from sucrose, trehalose, water-soluble cellulose, polysucrose.
20. The use according to claim 19, wherein the water-soluble cellulose is hydroxypropylmethyl cellulose.
21. The use according to claim 4, wherein the buffer is selected from at least one of DPBS or hepes-buffered HTF buffer or other cell culture buffer.
22. The use according to claim 4, wherein the cryopreservation fluid is prepared by: dissolving the peptide compound L-Thr-L-Arg and/or L-Thr-L-Pro-L-Thr in a buffer solution, cooling to room temperature, adjusting the pH, dissolving other components in the buffer solution, cooling, mixing, adjusting the pH, fixing the volume to a preset volume by using the buffer solution, and optionally adding serum or human serum albumin or bovine serum albumin when in use.
23. The use according to claim 22, wherein the method for preparing the cryopreservation solution comprises the following steps:
(1) Dissolving peptide compounds L-Thr-L-Arg and/or L-Thr-L-Pro-L-Thr in a part of buffer solution, cooling to room temperature, and adjusting pH to obtain a solution 1;
(2) Dissolving water-soluble sugar in a part of buffer solution, and adding other components after the water-soluble sugar is completely dissolved to prepare a solution 2;
(3) Optionally, dissolving PVA and/or a polyamino acid in a part of buffer solution, cooling to room temperature, and adjusting the pH value to prepare a solution 3;
(4) And (3) cooling the solution 1, the solution 2 and optionally the solution 3 to room temperature, mixing, adjusting the pH value, and fixing the volume to a preset volume by using a buffer solution to obtain the cryopreservation solution.
24. Use according to any one of claims 1 to 2, said stem cells being selected from the group consisting of pluripotent, multipotent or multipotent stem cells.
25. The use according to claim 24, wherein the stem cells are selected from the group consisting of embryonic stem cells, mesenchymal stem cells of various types, hematopoietic stem cells.
26. The use of claim 25, wherein the mesenchymal stem cells comprise umbilical cord mesenchymal stem cells, adipose mesenchymal stem cells, bone marrow mesenchymal stem cells.
27. The use according to any one of claims 1 to 2, wherein the stem cells are cryopreserved by a micro-drop method comprising the following steps: adding the cryopreservation liquid into the stem cells, blowing and dispersing to prepare stem cell suspension, placing the stem cell suspension on a freezing slide, and performing cryopreservation by using liquid nitrogen (-196 ℃).
CN202010172545.6A 2019-04-09 2020-03-12 Application of cryopreservation liquid containing peptide compounds in stem cell cryopreservation Active CN111793111B (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN2019102819867 2019-04-09
CN201910281986 2019-04-09

Publications (2)

Publication Number Publication Date
CN111793111A CN111793111A (en) 2020-10-20
CN111793111B true CN111793111B (en) 2022-11-29

Family

ID=72806402

Family Applications (3)

Application Number Title Priority Date Filing Date
CN202010171867.9A Active CN111793108B (en) 2019-04-09 2020-03-12 Application of cryopreservation liquid containing peptide compounds in organ and tissue cryopreservation
CN202010171868.3A Active CN111793109B (en) 2019-04-09 2020-03-12 Peptide compound and cryopreservation liquid containing same
CN202010172545.6A Active CN111793111B (en) 2019-04-09 2020-03-12 Application of cryopreservation liquid containing peptide compounds in stem cell cryopreservation

Family Applications Before (2)

Application Number Title Priority Date Filing Date
CN202010171867.9A Active CN111793108B (en) 2019-04-09 2020-03-12 Application of cryopreservation liquid containing peptide compounds in organ and tissue cryopreservation
CN202010171868.3A Active CN111793109B (en) 2019-04-09 2020-03-12 Peptide compound and cryopreservation liquid containing same

Country Status (1)

Country Link
CN (3) CN111793108B (en)

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN113508806B (en) * 2020-04-09 2022-08-16 中国科学院化学研究所 Application of amphiphilic compound in preparation of blood erythrocyte cryoprotectant
CN115119827A (en) * 2021-03-29 2022-09-30 中国科学院大学 Cryoprotectant and preparation method and application thereof
CN115400258B (en) * 2022-09-29 2023-11-21 中国科学院过程工程研究所 Amino acid-based aqueous liquid, preparation method and application thereof

Family Cites Families (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1991013091A1 (en) * 1990-03-01 1991-09-05 Agouron Pharmaceuticals, Inc. Cryoperservation of red blood cells
JP5726525B2 (en) * 2008-06-27 2015-06-03 株式会社バイオベルデ Composition for cryopreservation of cells and tissues
CN101519439B (en) * 2009-04-03 2012-05-02 北京金迪克生物技术研究所 Freezeproof polypeptide, synthesis, composition containing same and application thereof
EP2883951B1 (en) * 2012-06-15 2018-01-17 Kyokuto Pharmaceutical Industrial Co. Ltd Stem cell preservation medium, stem cell preservation method, and stem cell preservation system
CN103689061B (en) * 2013-12-05 2015-10-14 大连博缘科技有限公司 A kind of cell freezing fresh-keeping liquid
KR102347670B1 (en) * 2014-08-25 2022-01-06 삼성전자주식회사 Electronic decvice caes and electronic decvice having the same
CN105230610B (en) * 2015-11-13 2017-10-27 中国科学院化学研究所 A kind of freezen protective liquid and its preparation method and application
CN108207930B (en) * 2016-12-15 2021-06-25 中国科学院理化技术研究所 Cocktail type cryoprotectant and application thereof
CN107156111A (en) * 2017-06-30 2017-09-15 陈印平 A kind of candidate stem cell cell cryopreservation agent
CN109221082B (en) * 2018-09-14 2021-07-09 上海慧存医疗科技有限公司 Cell cryopreservation liquid, cryopreservation recovery method and application thereof

Also Published As

Publication number Publication date
CN111793108A (en) 2020-10-20
CN111793111A (en) 2020-10-20
CN111793109A (en) 2020-10-20
CN111793108B (en) 2022-05-27
CN111793109B (en) 2022-05-27

Similar Documents

Publication Publication Date Title
CN111793111B (en) Application of cryopreservation liquid containing peptide compounds in stem cell cryopreservation
CN111793110B (en) Application of DMSO-free cryopreservation liquid in stem cell cryopreservation
CN111789105B (en) Application of amino acid cryopreservation liquid in stem cell cryopreservation
WO2018064976A1 (en) Compositions and methods for cell cryopreservation
CN111789104B (en) Application of cryopreservation liquid in stem cell cryopreservation
CN111789099B (en) Application of cryopreservation liquid containing peptide compounds in cryopreservation of oocytes or embryos
CN107711823B (en) Cell cryopreservation liquid stored at normal temperature and application thereof
WO2020207152A1 (en) Serum-free cryopreservation solution and preparation method and application thereof
CN113508806B (en) Application of amphiphilic compound in preparation of blood erythrocyte cryoprotectant
CN113383766A (en) Freezing storage liquid for stem cell freezing storage and preparation method thereof
CN112438252A (en) Tissue freezing and thawing method suitable for mouse heart tissue single cell sequencing
JP7389138B2 (en) Bionic ice control material and cryopreservation solution containing this bionic ice control material
CN111789102B (en) Application of thawing solution in thawing frozen and preserved oocyte or embryo
CN111789103B (en) Thawing solution for cryopreservation and thawing method
CN113383767B (en) Application of frozen stock solution in stem cell freezing
JP7459130B2 (en) Melt liquid and its preparation method and application
CN114451400A (en) Application of cryopreservation liquid containing polypeptide in stem cell cryopreservation
CN115119827A (en) Cryoprotectant and preparation method and application thereof

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