CN112831457A - Method for separating and concentrating exosome - Google Patents

Method for separating and concentrating exosome Download PDF

Info

Publication number
CN112831457A
CN112831457A CN202110178197.8A CN202110178197A CN112831457A CN 112831457 A CN112831457 A CN 112831457A CN 202110178197 A CN202110178197 A CN 202110178197A CN 112831457 A CN112831457 A CN 112831457A
Authority
CN
China
Prior art keywords
concentration
buffer solution
sample
exosomes
binding
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.)
Pending
Application number
CN202110178197.8A
Other languages
Chinese (zh)
Inventor
李萍
肖木亮
高秀春
王绍成
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.)
Liaoning Rengen Biosciences Co ltd
Original Assignee
Liaoning Rengen Biosciences 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 Liaoning Rengen Biosciences Co ltd filed Critical Liaoning Rengen Biosciences Co ltd
Priority to CN202110178197.8A priority Critical patent/CN112831457A/en
Publication of CN112831457A publication Critical patent/CN112831457A/en
Pending legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2509/00Methods for the dissociation of cells, e.g. specific use of enzymes

Landscapes

  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Biomedical Technology (AREA)
  • Biotechnology (AREA)
  • Organic Chemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Genetics & Genomics (AREA)
  • Wood Science & Technology (AREA)
  • Zoology (AREA)
  • Microbiology (AREA)
  • Biochemistry (AREA)
  • General Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Cell Biology (AREA)
  • Investigating Or Analysing Biological Materials (AREA)

Abstract

The invention belongs to the technical field of biological detection, and relates to a method for quickly separating and concentrating exosomes. And separating and obtaining the exosome particles from the sample by combining the anion exchange resin with the exosomes in the sample. The method disclosed by the invention is simple to operate, short in time, low in cost and good in repeatability, can be used for quickly concentrating exosomes in a sample into a small volume, can be generally concentrated by 20-40 times, and hardly loses exosome particles. The method is convenient for extracting exosomes in a large-volume low-concentration sample, and is suitable for particle size analysis, electron microscope detection, Western blot, exosome DNA and RNA extraction and other related downstream experiments.

Description

Method for separating and concentrating exosome
Technical Field
The invention belongs to the technical field of biological detection, and relates to a method for quickly separating and concentrating exosomes.
Background
The exosome is a micro vesicle which is secreted by a living cell and contains abundant proteins, nucleic acids and lipid bilayer structures, and the diameter of the exosome is 30-150 nm. Exosomes are found in a wide range of body fluids, such as urine, ascites, blood, cell culture fluid, cerebrospinal fluid, and the like. Exosomes are capable of delivering functional molecules, including proteins, lipids and nucleic acids, to recipient cells, participate in intercellular communication, and affect various physiological and pathological functions of the cells, so that attention of many researchers is paid, and it is important to separate exosomes of a certain concentration before intensive exosome research.
The ultracentrifugation method for separating exosomes in the prior art is the most common method at present, is widely applied to separation of samples such as serum, plasma, cell culture fluid, urine and the like, is also the gold standard in the current separation method, can be used for extraction of exosomes in large-volume and low-concentration samples, but wastes time and labor, is easy to destroy the structure and function of exosomes, and has low recovery rate. The density gradient centrifugation method is a method for obtaining exosomes according to the density of exosomes by utilizing the continuous distribution of media with different densities from low to high to form a density gradient layer under the condition of long-time ultracentrifugal force, but the operation is complicated, the repeatability is poor, and the method needs to be provided with various expensive ultrahigh-speed centrifuges and is not suitable for extracting exosomes in large batch. The ultrafiltration method is to separate exosomes in a sample by using ultrafiltration membranes with different pore diameters to obtain exosomes, and the method is high in cost, suitable for concentrating exosomes in a small-volume sample, and not beneficial to extraction of exosomes in a large-volume low-concentration sample. The polymer precipitation method is characterized in that a high molecular polymer is used for competing for water around an exosome, so that the exosome is aggregated, the exosome concentration of a large-volume and low-concentration sample can be carried out, the operation is simple and convenient, the cost is low, but the impurity-containing protein is more, the polymer which is difficult to remove is generated, and the analysis of a downstream experiment is influenced. The immune affinity method is to realize the enrichment of exosomes by utilizing the specific interaction between corresponding antibodies and exosome membrane proteins, but the magnetic beads and the antibodies in the method are expensive and have harsh storage conditions, and the method is not beneficial to the extraction of exosomes in a large-volume and low-concentration sample. The size exclusion method is to separate the exosomes by using the particle sizes, can remove a part of protein components with small particles, has good separation and purification effects, and is not suitable for extracting the exosomes in a large-volume sample. The ion exchange chromatography uses ion exchange resin as filler to fill a chromatographic column, protein is separated according to the difference of charges carried by the protein, after the sample is added, the protein adsorbed on the column is eluted by adjusting different salt concentrations of a mobile phase, and fractions of the eluent are collected to play a good purification role.
Therefore, there is an urgent need to develop a method for separating and concentrating exosomes with simple operation, low cost, stability and high efficiency aiming at cell culture supernatant or organism liquid sample.
Disclosure of Invention
The invention aims to solve the technical problems of low exosome content, large sample volume, complex sample components and difficult exosome extraction of the existing samples such as cell culture supernatant or biological body fluid, and provides a novel method for quickly separating and concentrating exosomes, namely a method for extracting exosomes by using anion exchange resin without filling chromatographic columns.
In order to achieve the purpose, the technical scheme implemented by the invention is as follows:
a method of isolating and concentrating exosomes:
s1, adding a binding buffer solution into a sample containing an exosome;
s2, adding a binding resin into the system to enable the binding resin to be bound with exosomes in a sample;
s3, washing the bonding resin by using a washing solution to remove impure proteins;
and S4, eluting the combined resin by using an eluent containing a high-concentration salt solution to obtain exosomes in the sample.
The sample is cell culture supernatant or biological fluid; wherein, when the sample is cell culture supernatant, the cell culture supernatant needs 1500g, centrifugates for 15min at 4 ℃, abandons the precipitate, and the supernatant passes through a 0.22 μm filter membrane for standby;
when the sample is biological fluid, the biological fluid is needed to be centrifuged for 15min at 4 ℃ at 3000g, the precipitate is discarded, and the supernatant is filtered through a 0.22 mu m filter membrane for later use.
The volume ratio of the binding buffer solution to the sample is 1/25-1/5; wherein the binding buffer solution can be Tris buffer solution or phosphate buffer solution or MOPES buffer solution with the pH value of 7.0-8.5.
When the binding buffer solution is a Tris buffer solution, the concentration of Tris is 50-500 mM, and the pH value can be adjusted to 7.0-8.5 by HCl;
when the binding buffer solution is phosphate buffer solution, the concentration of NaCl is 20-100 mM, the concentration of KCl is 0.5-6 mM, and Na2HPO4The concentration of KH is 1-10 mM2PO4The concentration is 0.5-20 mM, and the pH is 7.0-8.5;
when the binding buffer solution is an MOPES buffer solution, the MOPES concentration is 10-100 mM, the sodium acetate concentration is 5-100 mM, the EDTA concentration is 5-100 mM, and the pH value is 7.0-8.5.
And the binding buffer solution and the sample are uniformly mixed and then incubated for 5-20 min.
The volume ratio of the binding resin to the sample is 1/50-1/10; wherein the binding resin is a weak anion exchange resin or a strong anion exchange resin, so that the binding resin is combined with the exosome in the sample.
The resin takes polyacrylate or agarose or cellulose or polystyrene as a matrix, and the matrix is prepared by reacting- (CH)2)2N(CH3)2Modifying to obtain weak anion exchange resin; or matrix through- (CH)2)2N+(CH3)3Modifying to obtain the strong anion exchange resin.
And (S2) after the binding resin and the sample system are uniformly mixed, incubating at room temperature for 10-60 min, and centrifuging at 1500-3000 g for 2-5 min.
The washing solution is HEPES buffer solution or phosphate buffer solution or Tris buffer solution with the pH value of 6.8-8.3.
When the washing solution is HEPES buffer solution, the concentration of HEPES is 50-500 mM, and the pH value is 6.8-8.3;
when the washing solution is phosphate buffer solution, the concentration of NaCl is 20-100 mM, the concentration of KCl is 0.5-6 mM, and Na2HPO4The concentration of KH is 1-10 mM2PO4The concentration is 0.5-10 mM, and the pH is 6.8-8.3;
when the washing solution is Tris buffer solution, the concentration of Tris is 50-500 mM, and the pH value can be adjusted to 6.8-8.3 by HCl.
And in the step S3, adding the washing solution into the purification column, incubating for 1-5 min, centrifuging for 2-5 min at 1500-3000 g, and discarding the filtrate.
The eluent is HEPES buffer solution or phosphate buffer solution or Tris buffer solution with the pH value of 6.3-7.4 and containing high-concentration NaCl.
When the eluent is HEPES buffer solution with high-concentration NaCl, the concentration of NaCl is 100-700 mM, the concentration of HEPES is 50-500 mM, and the pH value is 6.3-7.4;
when the eluent is phosphate buffer solution of high-concentration NaCl, the concentration of NaCl is 100-700 mM, the concentration of KCl is 0.5-10 mM, and Na is added2HPO4The concentration of KH is 1-20 mM2PO4The concentration is 0.5-10 mM, and the pH is 6.3-7.4;
when the eluent is Tris buffer solution with high concentration of NaCl, the concentration of NaCl is 100-700 mM, the concentration of Tris is 50-500 mM, and the pH value can be adjusted to 6.3-7.4 by HCl.
And S4, adding the eluent into a purification column, incubating for 3-7 min, centrifuging for 2-5 min at 200-500 g, adding the filtrate into the purification column again, incubating for 3-7 min, and centrifuging for 2-5 min at 1500-3000 g.
The invention can have the following beneficial effects:
the method can concentrate the exosomes in the cell culture supernatant or the biological body fluid with very low exosome content into small volume, has simple operation, low cost, short time consumption and stable result, and the obtained exosome concentrated solution contains salt ions, has no pollution of polymers, and is time-saving and labor-saving.
Drawings
FIG. 1 is a graph showing a comparison of ELISA tests for each component in the isolation and concentration of exosomes from cell culture according to the present invention.
FIG. 2 is a Western Blot identification of exosomes isolated and concentrated from cell culture fluid according to the present invention.
FIG. 3 is a transmission electron microscopy identification of exosomes isolated and concentrated from cell culture fluid according to the present invention; 3-1HEK293T, 3-2 Lncap; FIGS. 3-3 BALL.
FIG. 4 is a NTA identification chart of the isolation and concentration of exosomes from cell culture fluid according to the present invention; FIG. 4-1HEK293T, FIG. 4-2Lncap, FIG. 4-3 BALL.
FIG. 5 is a comparison of ELISA tests for each component in the isolation and concentration of exosomes from urine samples according to the present invention.
FIG. 6 is a Western Blot identification of exosomes isolated and concentrated from urine samples according to the present invention.
FIG. 7 is a transmission electron microscopy identification of exosomes isolated and concentrated from urine samples according to the present invention; urine A in FIG. 7-1, and urine B in FIG. 7-2.
FIG. 8 is a graph of NTA identification of exosomes isolated and concentrated from a urine sample according to the present invention; urine A in FIG. 8-1 and urine B in FIG. 8-2.
Detailed description of the preferred embodiment
The following examples are presented to further illustrate embodiments of the present invention, and it should be understood that the embodiments described herein are for purposes of illustration and explanation only and are not intended to limit the invention. It will be understood by those skilled in the art that various changes in form and details may be made therein without departing from the spirit and scope of the invention, and that such changes and modifications may be made without departing from the spirit and scope of the invention.
The ion exchange resin used in the invention is anion exchange resin, which takes polymer (agarose and cellulose) as a matrix, the matrix is bonded with ion exchange groups after surface hydrophilic modification, and simultaneously has good compatibility to bioactive macromolecules and excellent physical and chemical stability, thereby greatly improving the purification efficiency. The ion exchanger is mainly composed of a high molecular polymer matrix, a charge group and a balance ion. The charged group is covalently bonded to the high molecular weight polymer to form a charged ion-exchangeable group. Counterions are counterions bound to charged groups that can undergo reversible exchange reactions with other ionic groups in solution. The action of anion exchangers is that the ion exchanger which balances the negative charge of the ion is able to exchange with the negatively charged ionic groups.
The invention utilizes the ion exchange function of anion exchange resin, namely, the exosome in the sample is regulated by the binding buffer solution to make the protein on the outer membrane of the exosome carry negative charges and be combined with the anion exchange resin with positive charges, and because the outer membrane of the exosome contains abundant proteins, the combination sites of the exosome and the anion exchange resin are more than that of a single free protein, and the combination is firmer. During washing, the salt concentration and pH of the washing solution are adjusted to remove most of free protein, the free protein is easier to remove because the binding sites of the free protein and the anion exchange resin are few, and then the elution is carried out by the eluent containing high-concentration salt solution, so that a large amount of exosomes are obtained from the sample.
The reagent and equipment adopted by the invention are conventional reagents and equipment in the technical field, and the ELISA detection kit is a human total exosome capture and quantification kit (cell culture supernatant) and a human total exosome capture and quantification kit (urine) purchased from Liaoning Runzi Biotechnology Limited. The reagents, materials and equipment used in the following examples are commercially available.
Example 1
Method for separating and concentrating exosomes from cell culture solution
The culture supernatants of three cell lines HEK293T (DMEM)/Lncap (RPM1640)/BALL (RPM1640) were subjected to exosome extraction and identified.
(1) 10ml of different cell culture solutions (HEK293T/Lncap/BALL) were centrifuged at 1500g for 10min at 4 ℃ to remove suspended cells and cell debris, the supernatant was filtered through a 0.22 μm pore size filter to remove larger microvesicles, and 5ml of the cell culture solutions were pipetted into different 10ml centrifuge tubes (noted as HEK293T, Lncap, BALL, respectively).
(2) Pipette 500. mu.l of binding buffer into the centrifuge tube containing the culture medium in (1), cover tightly, and mix by inversion. Wherein the binding buffer is 200mM Tris-HCl buffer (pH8.0).
(3) Adding 300 μ l of the binding resin into the centrifuge tube in (2), covering the cap tightly, mixing by continuous inversion at room temperature, incubating for 15min, centrifuging at 1500g for 2min, taking out the centrifuge tube gently, taking 500 μ l of supernatant (without discarding) with a pipette, carefully pouring off the remaining supernatant,the resin was gently blown up with liquid (500. mu.l) in a pipette, transferred all to a purification column (collection tube already placed), left to stand for 2min, centrifuged at 2,000g at room temperature for 2min, and the filtrate and collection tube were discarded. Wherein the binding resin is agarose as matrix and has a chemical structure of- (CH)2)2N(CH3)2) The anion exchange resin of (a) may be prepared as described in the prior art or may be obtained from sigma.
(4) Sucking 500 μ l of washing solution, adding into a purification column, standing for 5min, centrifuging at 2000g room temperature for 2min, and discarding the filtrate and collection tube. Wherein the washing solution is 68mM HEPES buffer solution, and the pH value is 7.4.
(5) Transferring the purification column into a 1.5ml centrifuge tube with low protein adsorption, adding 200 mul of eluent, incubating for 5min, centrifuging at 350g for 2min at room temperature, transferring the eluent into the purification column again, incubating for 5min, centrifuging at 2500g at room temperature for 3min, and obtaining the liquid in the centrifuge tube, namely the concentrated exosome solution. Wherein the eluent is 450mM NaCl, 5.6mM KCl, 5.2mM Na2HPO4,6mM KH2PO4Phosphate buffer, pH 6.5.
(6) Exosome ELISA quantitative detection
The presence of exosomes in each sample was verified by ELISA (total exosome capture and quantification kit for human, product number RGEXOC96-1) (reading see table 1, results figure 1) with PBS as negative control.
TABLE 1
HEK293T Lncap BALL
Stock solution (OD)450Value) 0.625 0.745 0.543
Filtrate (OD)450Value) 0.063 0.065 0.059
Cleaning solution (OD)450Value) 0.078 0.065 0.064
Eluent (OD)450Value) 1.542 1.643 1.401
Negative control OD detected by ELISA experiment4500.057, as can be seen from Table 1, the OD of the filtrate of the cell culture fluid sample HEK293T/Lncap/BALL4500.063, 0.065, 0.059, respectively, which is substantially identical to the negative control. That is, adding 300. mu.l of the binding resin to a 5ml sample of the cell culture solution can capture almost all of the exosomes in the sample. Wash OD of sample HEK293T/Lncap/BALL4500.078, 0.065 and 0.064, respectively, indicating that the exosomes were hardly lost during washing. As can be seen from FIG. 1, the values of the eluates for separating and concentrating the three cell culture solutions are significantly higher than those of the stock solutions, indicating that the eluates contain a large amount of exosomes, which can be concentrated about 25-fold with respect to the volume of the stock solutions.
(7) BCA protein quantitative determination
The protein content in each wash and eluate was determined by BCA protein quantification assay (see table 2) with PBS as negative control.
TABLE 2
HEK293T Lncap BALL
Stock solution (mg/ml) 2.145 2.512 2.758
Filtrate (mg/ml) 1.825 2.143 2.300
Washing liquid (mg/ml) 0.212 0.220 0.312
Eluent (mg/ml) 1.848 1.924 2.056
As shown in Table 2, the binding resin was rarely bound to the free protein in each cell culture fluid sample, and 300. mu.l of the binding resin was bound to about 0.3-0.5 mg/ml of the protein in 5ml of the cell culture fluid sample, indicating that the positively charged protein in the sample was not bound to the binding resin, i.e., the free protein in the sample was selectively bound to the binding resin.
As is clear from tables 1 and 2, the protein contained in the outer membrane of the exosome was more strongly bound to the bound resin than the free protein, and the partially free protein on the resin was removed by washing with the washing solution, but the exosome was hardly lost.
(8) Western Blot detection
The exosome solution obtained after elution in step (5) was validated by Western Blot experiment (see fig. 2).
Electrophoresis was run with 10% SDS-PAGE gel:
preparation of 10% SDS-PAGE separation gel:
composition (I) Volume of Unit of
H2O 4 ml
30%Acr/Bise 3.3 ml
1.5M Tris-HCL pH8.8 2.5 ml
10%SDS 100 μl
10%AP 100 μl
TEMED
10 μl
Preparation of 10% SDS-PAGE concentrated gel:
composition (I) Volume of Unit of
H2O 1.53 ml
30%Acr/Bise 0.33 ml
0.5M Tris-HCL pH6.8 0.63 ml
10%SDS 25 μl
10%AP 30 μl
TEMED 3 μl
Sample loading pretreatment:
protein quantification was performed separately on each eluate from step (4) using the BCA protein assay: detecting to obtain the protein concentration of the BALL eluent as 2.4 mg/ml; the protein concentration of the HEK293T eluate was 3.1 mg/ml; the Lncap eluate had a protein concentration of 2.9 mg/ml.
And (3) respectively sucking 20 mu l of each eluent in the step (4) into a 0.5mL centrifuge tube, adding 5 mu l of 5 Xloading buffer solution to a final concentration of 1X, wherein 25 mu l of the buffer solution is used totally, before loading, heating the sample at 99 ℃ for 10min to denature the protein, and after cooling to room temperature, respectively taking 20 mu l of the sample for electrophoresis loading.
The electrophoresis time is about 1.5h generally, the voltage of the concentrated gel is 80V and the constant voltage is 30min, and the voltage of the separation gel is 120V and the constant voltage is 40-50 min. The electrophoresis was terminated as soon as bromophenol blue (approx. 20kDa) came out.
Film transfer: wet transfer was performed using PVDF membrane, 250mM constant current transfer for 1.5 h.
And (3) sealing: PVDF membrane was soaked in 4% skimmed milk powder (TBST lysis) and incubated on a destaining shaker for 1h at room temperature or overnight at 4 ℃.
Washing: the PVDF membrane was washed 3 times with TBST, 8 min/time.
Primary antibody incubation: primary anti-antibody CD9 was solubilized with 4% BSA (TBST) at 1: 500(v/v) dilution; CD63 was diluted 1:1000(v/v) with 4% BSA (TBST lysis); ALIX was diluted 1:400(v/v) with 4% BSA (TBST lysis). Incubation was performed at room temperature for 2h or at 4 ℃ overnight.
Washing: the PVDF membrane was washed 3 times with TBST, 8 min/time.
And (3) secondary antibody incubation: the secondary HRP-labeled goat anti-mouse was diluted 1:3000(v/v) with 4% skim milk powder (TBST solubilized) and incubated at room temperature for 1 h.
Washing: the PVDF membrane was washed 3 times with TBST, 8 min/time.
Color development: ECL luminescence color development, with gel imaging system analysis of target band molecular weight.
As shown in FIG. 2, the exosomes isolated and concentrated from the HEK293T/Lncap/BALL three cell culture solutions were verified by Western Blot, and ALIX/CD9/CD63 bands were clearly visible and deep.
(9) Transmission Electron Microscopy (TEM) detection
Exosomes were removed in 5 μ L and diluted to 10 μ L. Sucking 10 mu L of a sample, dropwise adding the sample on a copper net for incubation for 3min, sucking floating liquid by using filter paper, dropwise adding 10 mu L of uranyl acetate on the copper net for incubation for 1min, sucking the floating liquid by using the filter paper, drying for several minutes at normal temperature, and performing electron microscope detection imaging at 100 kv. Exosomes obtained from the three culture fluids (HEK293T/Lncap/BALL) by the method are detected by an electron microscope (see figure 3), and the exosomes of each sample are complete in structure and are like a saucer as can be seen from figure 3-1HEK293T, figure 3-2Lncap, and figure 3-3 BALL.
(10) Nano particle size detection
Nanoparticle Tracking Analyzer (NTA) detection method: diluting the sample with water to obtain a particle concentration of 1 × 107Perml and 1X 109In the/mL range. The number and size of particles in the sample were then measured using a Zeta View PMX110 instrument at 405nm laser and photographs were taken at 30/sec for 1 minute. Finally the movement of the particles was analyzed using NTA software (zetaview8.02.28).
The exosomes obtained by this method were tested by NTA (see fig. 4), and the number of particles with a particle size of 109.2nm was 98.7% obtained from HEK293T of fig. 4-1, Lncap of fig. 4-2, and BALL of cell culture fluid HEK293T, and the result was 9.6E +10 particles/ml. The number of particles with an exosome particle size of 108.3nm obtained from the cell culture fluid Lncap was 98.4%, and the detection result was 9.4E +10 particles/ml. The number of particles with an exosome particle size of 123nm obtained from the cell culture fluid BALL was 99%, and the detection result was 2.6E +10 particles/ml.
The experiments prove that the invention is feasible and effective for separating and concentrating exosomes in various cell lines cultured by two common culture media of DMEM and RPM1640, and keeps the integrity and biological functions of the exosomes.
Example 2
The method for separating and concentrating the exosome is established, and the exosome is further extracted and identified from a human urine sample.
(1) 15ml of human urine A and urine B are respectively centrifuged at 4 ℃ and 3000g for 10min to remove cell debris and impurities, the supernatant is retained and filtered by a filter membrane with the aperture of 0.22 mu m to remove larger micro vesicles, and 5ml of each of the urine A and the urine B (respectively marked as samples: urine A and urine B) is respectively added into 2 10ml centrifuge tubes.
(2) Pipette 500. mu.l of binding buffer into the centrifuge tube containing the culture medium in (1), cover tightly, and mix by inversion. Wherein, when the binding buffer is MOPES buffer, the MOPES concentration is 15mM, the sodium acetate concentration is 20mM, the EDTA concentration is 15mM, and the pH value is 8.3.
(3) And (3) sucking 200 mu l of binding resin, adding the binding resin into the centrifuge tube in the step (2), covering the cover tightly, continuously reversing and mixing the binding resin evenly at room temperature, incubating for 15min, centrifuging for 2min at 1500g, taking the centrifuge tube out of the centrifuge lightly, taking 500 mu l of supernatant (not to be discarded) by using a pipette, carefully pouring out the residual supernatant, slightly blowing the resin by using liquid (500 mu l) in the pipette, completely transferring the resin into a purification column (placed into a collection tube), standing for 2min, centrifuging for 2min at 2,000g at room temperature, and discarding filtrate and the collection tube. Wherein the bonding resin is prepared from cellulose as matrix and has a chemical structure of- (CH)2)2N+(CH3)3The anion exchange resin of (a) may be prepared as described in the prior art or may be obtained from GE, USA.
(4) The 500. mu.l of washing solution was pipetted into a purification column, allowed to stand for 5min, centrifuged at 2,000g at room temperature for 2min, and the filtrate and collection tube were discarded together. Wherein the washing solution is 45mM Tris-HCl buffer solution, and the pH value is 7.02.
(5) Transferring the purification column into a 1.5ml centrifuge tube with low protein adsorption, adding 150 mu l of eluent, incubating for 5min, centrifuging at 350g for 2min at room temperature, transferring the eluent into the purification column again, incubating for 5min, centrifuging at 2500g at room temperature for 3min, and obtaining the liquid in the centrifuge tube, namely the concentrated exosome solution. Wherein the eluent is 375mM NaCl, 2.1mM KCl, 6.8mM Na2HPO4,3.1mMKH2PO4The pH of the phosphate buffer of (1) was 7.0.
(6) Exosome ELISA quantitative detection
The presence of exosomes in each sample was verified by ELISA (total exosome capture and quantification kit for human, product number RGEXOU96-1) (reading see table 3, results figure 5) with PBS as negative control.
TABLE 3
Urine A Urine B
Stock solution (OD)450Value) 0.645 0.723
Filtrate (OD)450Value) 0.072 0.065
Cleaning solution (OD)450Value) 0.102 0.090
Eluent (OD)450Value) 1.442 1.603
Negative control OD detected by ELISA experiment4500.056, as can be seen from Table 3, the OD of the filtrate in urine samples A and B4500.072 and 0.065, respectively, which is substantially consistent with the negative control, i.e., adding 200. mu.l of binding resin to a 5ml urine sample allowed almost complete capture of exosomes in the sample. Wash OD of urine samples A and B4500.102 and 0.090, respectively, indicating that exosomes were hardly lost upon washing. As can be seen from fig. 5, the elution values of the urine samples a and B are significantly higher than the original solution, which indicates that the elution solution contains a large amount of exosomes, and the exosomes can be concentrated by about 25-30 times relative to the original solution volume.
(7) BCA protein quantitative determination
The protein content in each wash and eluate was determined by BCA protein quantification assay (see table 4) with PBS as negative control.
TABLE 4
Urine A Urine B
Stock solution (protein mg/ml) 9.432 8.789
Filtrate (protein mg/ml) 8.661 8.056
Washing liquid (protein mg/ml) 0.421 0.526
Eluent (protein mg/ml) 2.089 2.125
As shown in Table 4, the binding resin was rarely bound to the free protein in each sample of urine, and 200ul of the binding resin was bound to about 0.5-0.8 mg/ml of the protein in 5ml of the urine, indicating that the positively charged protein in the sample was not bound to the binding resin, i.e., the free protein in the sample was selectively bound to the binding resin.
As can be seen from tables 3 and 4, washing with the wash solution removed some of the free protein on the resin, but almost no exosomes were lost.
(8) Western Blot detection
The exosome solution obtained after elution in step (5) was validated by Western Blot experiment (see fig. 6).
The Western Blot experiment method and the operation steps are the same as above.
As shown in FIG. 6, the ALIX/CD9/CD63 bands of urine A and urine B were clearly visible and the bands were deep as confirmed by Western Blot.
(9) Transmission Electron Microscopy (TEM) detection
The structure of the exosomes obtained from urine a and urine B by this method was examined by Transmission Electron Microscopy (TEM) (see fig. 7), the operation method is the same as above, and the exosomes from fig. 7-1, urine a and urine B from fig. 7-2 were structurally complete and in the form of a saucer.
(10) Nano particle size detection
The exosomes obtained from urine a and urine B by this method were detected by a nanoparticle particle tracking analyzer (NTA) (see fig. 8), the operation was the same as above, and in fig. 8-1, urine a and urine B of fig. 8-2, it was found that the exosomes obtained from urine a had a particle size of 107.4nm, which accounted for 97.1% and a concentration of 1.3E +10particles/ml, respectively. Urine B obtained 96.5% of exosomes having a particle size of 107.5nm at a concentration of 1.3E +10 particles/ml.
The experiments prove that the method is feasible and effective for separating and concentrating exosomes in urine, and keeps the integrity and biological functions of exosomes.
The foregoing are merely exemplary embodiments of the present invention, which enable those skilled in the art to understand or practice the present invention. Various modifications to these embodiments will be readily apparent to those skilled in the art, and the generic principles defined herein may be applied to other embodiments without departing from the spirit or scope of the invention. Thus, the present invention is not intended to be limited to the embodiments shown herein but is to be accorded the widest scope consistent with the principles and novel features disclosed herein.

Claims (10)

1. A method of isolating and concentrating exosomes, characterized by:
s1, adding a binding buffer solution into a sample containing an exosome;
s2, adding a binding resin into the system to enable the binding resin to be bound with exosomes in a sample;
s3, washing the bonding resin by using a washing liquid;
and S4, eluting the combined resin by using an eluent containing a high-concentration salt solution to obtain exosomes in the sample.
2. The method of claim 1, wherein: the sample is cell culture supernatant or biological fluid;
wherein, when the sample is cell culture supernatant, the cell culture supernatant needs 1500g, centrifugates for 15min at 4 ℃, abandons the precipitate, and the supernatant passes through a 0.22 μm filter membrane for standby;
when the sample is biological fluid, the biological fluid is needed to be centrifuged for 15min at 4 ℃ at 3000g, the precipitate is discarded, and the supernatant is filtered through a 0.22 mu m filter membrane for later use.
3. The method of claim 1, wherein: the volume ratio of the binding buffer solution to the sample is 1/25-1/5; wherein the binding buffer solution is Tris buffer solution or phosphate buffer solution or MOPES buffer solution with the pH value of 7.0-8.5; wherein, when the binding buffer solution is a Tris buffer solution, the concentration of Tris is 50-500 mM, and the pH value can be adjusted to 7.0-8.5 by HCl; when the binding buffer solution is phosphate buffer solution, the concentration of NaCl is 20-100 mM, the concentration of KCl is 0.5-6 mM, and Na2HPO4The concentration of KH is 1-10 mM2PO4The concentration is 0.5-20 mM, and the pH is 7.0-8.5; when the binding buffer solution is an MOPES buffer solution, the MOPES concentration is 10-100 mM, the sodium acetate concentration is 5-100 mM, the EDTA concentration is 5-100 mM, and the pH value is 7.0-8.5.
4. A method according to claim 1 or 3, characterized in that: and the binding buffer solution and the sample are uniformly mixed and then incubated for 5-20 min.
5. The method of claim 1, wherein: the volume ratio of the binding resin to the sample is 1/50-1/10; wherein the binding resin is a weak anion exchange resin or a strong anion exchange resin; the resin takes polyacrylate or agarose or cellulose or polystyrene as a matrix, and the matrix is prepared by reacting- (CH)2)2N(CH3)2Or- (CH)2)2N+(CH3)3The modification results in a weak anion exchange resin or a strong anion exchange resin.
6. The method of claim 1 or 5, wherein: and (S2) after the binding resin and the sample system are uniformly mixed, incubating at room temperature for 10-60 min, and centrifuging at 1500-3000 g for 2-5 min.
7. The method of claim 1, wherein: the washing solution is HEPES buffer solution or phosphate buffer solution or Tris buffer solution with the pH value of 6.8-8.3; when the washing solution is an HEPES buffer solution, the concentration of the HEPES is 50-500 mM, and the pH value is 6.8-8.3; when the washing solution is phosphate buffer solution, the concentration of NaCl is 20-100 mM, the concentration of KCl is 0.5-6 mM, and Na2HPO4The concentration of KH is 1-10 mM2PO4The concentration is 0.5-10 mM, and the pH is 6.8-8.3; when the washing solution is Tris buffer solution, the concentration of Tris is 50-500 mM, and the pH value can be adjusted to 6.8-8.3 by HCl.
8. The method of claim 1 or 7, wherein: and in the step S3, adding the washing solution into the purification column, incubating for 1-5 min, centrifuging for 2-5 min at 1500-3000 g, and discarding the filtrate.
9. The method of claim 1, wherein: the eluent is HEPES buffer solution or phosphate buffer solution or Tris buffer solution with the pH value of 6.3-7.4 and containing high-concentration NaCl; when the eluent is HEPES buffer solution with high concentration of NaCl, the concentration of NaCl is 100-700 mM, the concentration of HEPES is 50-500 mM, and the pH value is 6.3-7.4;
when the eluent is phosphate buffer solution of high-concentration NaCl, the concentration of NaCl is 100-700 mM, the concentration of KCl is 0.5-10 mM, and Na is added2HPO4The concentration of KH is 1-20 mM2PO4The concentration is 0.5-10 mM, and the pH is 6.3-7.4; when the eluent is Tris buffer solution with high concentration of NaCl, the concentration of NaCl is 100-700 mM, the concentration of Tris is 50-500 mM, and the pH value can be adjusted to 6.3-7.4 by HCl.
10. The method of claim 1 or 9, wherein: and S4, adding the eluent into a purification column, incubating for 3-7 min, centrifuging for 2-5 min at 200-500 g, adding the filtrate into the purification column again, incubating for 3-7 min, and centrifuging for 2-5 min at 1500-3000 g.
CN202110178197.8A 2021-02-07 2021-02-07 Method for separating and concentrating exosome Pending CN112831457A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202110178197.8A CN112831457A (en) 2021-02-07 2021-02-07 Method for separating and concentrating exosome

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202110178197.8A CN112831457A (en) 2021-02-07 2021-02-07 Method for separating and concentrating exosome

Publications (1)

Publication Number Publication Date
CN112831457A true CN112831457A (en) 2021-05-25

Family

ID=75933090

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202110178197.8A Pending CN112831457A (en) 2021-02-07 2021-02-07 Method for separating and concentrating exosome

Country Status (1)

Country Link
CN (1) CN112831457A (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN113533589A (en) * 2021-09-16 2021-10-22 天九再生医学(天津)科技有限公司 Method for analyzing exosomal charge heterogeneity
CN114323851A (en) * 2021-12-24 2022-04-12 多莱泌生物科技(武汉)有限公司 Method for separating exosomes in serum and plasma based on ultrafiltration and affinity chromatography technology
CN115354031A (en) * 2022-08-26 2022-11-18 多莱泌生物科技(武汉)有限公司 Preparation method for extracting high-purity exosome from biological fluid on cell

Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20150241431A1 (en) * 2014-02-27 2015-08-27 Board Of Regents, The University Of Texas System Methods and Compositions for Isolating Exosomes
CN107858324A (en) * 2017-11-27 2018-03-30 付清玲 A kind of method for the extracellular vesica including excretion body secreted based on anion exchange resin adsorbing separation cell to culture medium
CN108841777A (en) * 2018-06-22 2018-11-20 北京恩泽康泰生物科技有限公司 The extracting method and device of extracellular vesica based on Electrostatic Absorption and its content
CN108866046A (en) * 2018-07-25 2018-11-23 辽宁润基生物科技有限公司 A method of excretion body and/or free nucleic acid in separation, purification of samples
CN108865978A (en) * 2018-07-25 2018-11-23 辽宁润基生物科技有限公司 A method of separation and purifying excretion body
US20190301988A1 (en) * 2016-11-23 2019-10-03 Aman Sharma Method and kit for exosomes and associated biomacromolecules capture
US20200132682A1 (en) * 2017-03-24 2020-04-30 Dapi Meng Lin CHIANG Methods and kits for exosome isolation and quantification

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20150241431A1 (en) * 2014-02-27 2015-08-27 Board Of Regents, The University Of Texas System Methods and Compositions for Isolating Exosomes
US20190301988A1 (en) * 2016-11-23 2019-10-03 Aman Sharma Method and kit for exosomes and associated biomacromolecules capture
US20200132682A1 (en) * 2017-03-24 2020-04-30 Dapi Meng Lin CHIANG Methods and kits for exosome isolation and quantification
CN107858324A (en) * 2017-11-27 2018-03-30 付清玲 A kind of method for the extracellular vesica including excretion body secreted based on anion exchange resin adsorbing separation cell to culture medium
CN108841777A (en) * 2018-06-22 2018-11-20 北京恩泽康泰生物科技有限公司 The extracting method and device of extracellular vesica based on Electrostatic Absorption and its content
CN108866046A (en) * 2018-07-25 2018-11-23 辽宁润基生物科技有限公司 A method of excretion body and/or free nucleic acid in separation, purification of samples
CN108865978A (en) * 2018-07-25 2018-11-23 辽宁润基生物科技有限公司 A method of separation and purifying excretion body

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
CHANG-SOOK HONG, SONJA FUNK, LAURENT MULLER, MICHAEL BOYIADZIS, THERESA L WHITESIDE: "Isolation of biologically active and morphologically intact exosomes from plasma of patients with cancer", J EXTRACELL VESICLES, vol. 5, pages 29289, XP055698874, DOI: 10.3402/jev.v5.29289 *
南阿妮;刁艳君;杨柳;马越云;苏明权;郝晓柯;: "尿液外泌体提取优化及冷冻对外泌体RNA含量的影响", 临床检验杂志, vol. 37, no. 05, pages 325 - 330 *
范维肖;刁艳君;马越云;郝晓柯;: "超速离心法与QIAGEN膜亲和柱法提取前列腺癌细胞培养上清外泌体的方法学比较", 现代检验医学杂志, vol. 34, no. 03, pages 6 - 9 *

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN113533589A (en) * 2021-09-16 2021-10-22 天九再生医学(天津)科技有限公司 Method for analyzing exosomal charge heterogeneity
CN114323851A (en) * 2021-12-24 2022-04-12 多莱泌生物科技(武汉)有限公司 Method for separating exosomes in serum and plasma based on ultrafiltration and affinity chromatography technology
CN115354031A (en) * 2022-08-26 2022-11-18 多莱泌生物科技(武汉)有限公司 Preparation method for extracting high-purity exosome from biological fluid on cell

Similar Documents

Publication Publication Date Title
CN112831457A (en) Method for separating and concentrating exosome
CN106124282B (en) A kind of method of lamination centrifugal filtration separation and Extraction excretion body
JP7364240B2 (en) Method for separating extracellular endoplasmic reticulum using hydrophobic interactions
US20220349901A1 (en) Lectin-macromolecule carrier coupling complex for separating glycosylated exosome in clinical sample
US10125199B2 (en) Manufacture of magnetic particles
JP7383162B2 (en) Lectin-magnetic carrier coupling complex used to isolate glycosylated exosomes from clinical samples
CN108841777A (en) The extracting method and device of extracellular vesica based on Electrostatic Absorption and its content
CN108865978A (en) A method of separation and purifying excretion body
CN113774008A (en) Method for extracting exosome and application thereof
CN109082400A (en) A method of excretion body being separated from biological sample using DEAE magnetic nano particle
CN110231207A (en) A method of separation excretion body
Ströhle et al. Affinity-based isolation of extracellular vesicles and the effects on downstream molecular analysis
CN111474356A (en) Double-immunomagnetic-bead sorting reagent, preparation method thereof and application thereof in enrichment of humoral exosomes
CN114276992A (en) Complete exosome separation and purification kit and detection analysis method
CN113101737B (en) Affinity tangential flow filtration system, construction method thereof, exosome extraction method and application
CN116333987A (en) Application of NK cell exosomes obtained by exosome extraction method in resisting tumor
CN115558629A (en) Exosome extraction affinity magnetic bead and extraction kit thereof
CN112813027B (en) Method for separating exosomes in urine
CN109738630B (en) High-sensitivity magnetic bead sorting combined colloidal gold marked exosome immune electron microscope method
CN112980006A (en) Protein cross-linked nano affinity microsphere, preparation method and application
CN116773500B (en) Method for separating and identifying extracellular vesicles
CN117330481B (en) Flow detection method for exosomes and application thereof
CN113088494B (en) Method for releasing tumor cells captured by erythrocyte biomimetic material
CN112630449B (en) Blood exosome marker for judging buffalo birth time and application thereof
Wehland et al. Combining Immuno-Affinity Chromatography and Filtration to Improve Specificity and Size Distribution of Exosome-Containing EV Populations

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