WO2021031270A1 - 细菌膜囊泡及其分离制备系统和方法 - Google Patents

细菌膜囊泡及其分离制备系统和方法 Download PDF

Info

Publication number
WO2021031270A1
WO2021031270A1 PCT/CN2019/106654 CN2019106654W WO2021031270A1 WO 2021031270 A1 WO2021031270 A1 WO 2021031270A1 CN 2019106654 W CN2019106654 W CN 2019106654W WO 2021031270 A1 WO2021031270 A1 WO 2021031270A1
Authority
WO
WIPO (PCT)
Prior art keywords
bacteria
membrane vesicles
bacterial
supernatant
cells
Prior art date
Application number
PCT/CN2019/106654
Other languages
English (en)
French (fr)
Inventor
王震玲
魏于全
Original Assignee
四川大学
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
Priority claimed from CN201910777473.5A external-priority patent/CN112402601B/zh
Priority claimed from CN201921369450.2U external-priority patent/CN211394490U/zh
Priority claimed from CN201910777595.4A external-priority patent/CN112410212B/zh
Priority claimed from CN201910777606.9A external-priority patent/CN112410240B/zh
Priority claimed from CN201910777479.2A external-priority patent/CN112410239B/zh
Application filed by 四川大学 filed Critical 四川大学
Priority to US17/637,051 priority Critical patent/US20220378902A1/en
Publication of WO2021031270A1 publication Critical patent/WO2021031270A1/zh

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/02Bacterial antigens
    • A61K39/104Pseudomonadales, e.g. Pseudomonas
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K41/00Medicinal preparations obtained by treating materials with wave energy or particle radiation ; Therapies using these preparations
    • A61K41/10Inactivation or decontamination of a medicinal preparation prior to administration to an animal or a person
    • A61K41/17Inactivation or decontamination of a medicinal preparation prior to administration to an animal or a person by ultraviolet [UV] or infrared [IR] light, X-rays or gamma rays
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/69Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit
    • A61K47/6905Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a colloid or an emulsion
    • A61K47/6911Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a colloid or an emulsion the form being a liposome
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/50Microcapsules having a gas, liquid or semi-solid filling; Solid microparticles or pellets surrounded by a distinct coating layer, e.g. coated microspheres, coated drug crystals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/02Drugs for dermatological disorders for treating wounds, ulcers, burns, scars, keloids, or the like
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/195Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria
    • C07K14/21Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria from Pseudomonadaceae (F)
    • 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
    • C12N1/00Microorganisms, e.g. protozoa; Compositions thereof; Processes of propagating, maintaining or preserving microorganisms or compositions thereof; Processes of preparing or isolating a composition containing a microorganism; Culture media therefor
    • C12N1/20Bacteria; 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
    • C12N1/00Microorganisms, e.g. protozoa; Compositions thereof; Processes of propagating, maintaining or preserving microorganisms or compositions thereof; Processes of preparing or isolating a composition containing a microorganism; Culture media therefor
    • C12N1/20Bacteria; Culture media therefor
    • C12N1/205Bacterial isolates
    • 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
    • C12N13/00Treatment of microorganisms or enzymes with electrical or wave energy, e.g. magnetism, sonic waves
    • 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
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/74Vectors or expression systems specially adapted for prokaryotic hosts other than E. coli, e.g. Lactobacillus, Micromonospora
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/52Bacterial cells; Fungal cells; Protozoal cells
    • A61K2039/521Bacterial cells; Fungal cells; Protozoal cells inactivated (killed)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/54Medicinal preparations containing antigens or antibodies characterised by the route of administration
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/545Medicinal preparations containing antigens or antibodies characterised by the dose, timing or administration schedule
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55505Inorganic adjuvants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55511Organic adjuvants
    • A61K2039/55555Liposomes; Vesicles, e.g. nanoparticles; Spheres, e.g. nanospheres; Polymers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55588Adjuvants of undefined constitution
    • A61K2039/55594Adjuvants of undefined constitution from bacteria
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12RINDEXING SCHEME ASSOCIATED WITH SUBCLASSES C12C - C12Q, RELATING TO MICROORGANISMS
    • C12R2001/00Microorganisms ; Processes using microorganisms
    • C12R2001/01Bacteria or Actinomycetales ; using bacteria or Actinomycetales
    • C12R2001/38Pseudomonas
    • C12R2001/385Pseudomonas aeruginosa

Definitions

  • the invention belongs to the field of microbiology, and specifically relates to a bacterial membrane vesicle, a system and method for separating and preparing the membrane vesicle, and the application of the membrane vesicle.
  • Membrane vesicles are vesicle-like structure products produced by the outer membrane of bacterial cells (including gram-positive and gram-negative bacteria). Most of them are spherical with a diameter of about 20-250nm (Structures of gram-negative cell walls and their derived membrane vesicles), Beveridge T, Journal of Bacteriology, 1999vol: 181( 16)pp:4725-33).
  • Bacterial membrane vesicles contain a variety of biologically active macromolecules such as nucleic acids, lipopolysaccharides, outer membrane proteins and other components, as well as metal ions, enzymes, signaling molecules, etc.
  • membrane vesicles occurs at any growth stage of bacteria, which is different from cell lysis and apoptosis. Studies have found that stress stimulation, hypoxia, and antibiotic compression can promote the production of membrane vesicles by bacteria. However, the yield of naturally-occurring bacterial membrane vesicles is low, and it is often necessary to cultivate a large number of bacteria to harvest a certain amount of membrane vesicles, and subsequent purification processes are required to obtain membrane vesicles of a certain quality.
  • the present invention provides a bacterial membrane vesicle and a system and method for isolating and producing such bacterial membrane vesicles, including five sub-inventions (namely, invention A, invention B, invention C, invention D, and invention E) and Based on the respective expansion of these five sub-inventions.
  • the invention first discloses a membrane vesicle isolated from inactivated bacteria.
  • the inactivated bacteria include gram-positive bacteria and gram-negative bacteria, and genetically engineered bacteria.
  • the invention also provides methods for separating and preparing the membrane vesicles, as well as the application as a vaccine.
  • the present invention uses ionizing X-rays to irradiate bacteria for the first time, isolate and purify the MVs produced therefrom, and the prepared MVs can be used as vaccines, vaccine adjuvants, and/or drug carriers.
  • the present invention also discloses a membrane vesicle isolated from inactivated Pseudomonas aeruginosa according to the present invention.
  • the invention also provides methods for separating and preparing the membrane vesicles, as well as the application as a vaccine. And the application of the bacterial vaccine.
  • the present invention uses ionizing X-rays to irradiate Pseudomonas aeruginosa for the first time, isolate and purify the MVs produced therefrom, and the prepared MVs can be used as vaccines, vaccine adjuvants, and/or drug carriers.
  • the present invention also discloses a membrane vesicle isolated from inactivated Staphylococcus aureus.
  • the invention also provides methods for separating and preparing the membrane vesicles, as well as the application as a vaccine. And the application of the bacterial vaccine.
  • the present invention uses ionizing X-rays to irradiate Staphylococcus aureus for the first time to isolate and purify the MVs produced therefrom.
  • the prepared MVs can be used as vaccines, vaccine adjuvants, and/or drug carriers.
  • the present invention also discloses a production system for bacterial membrane vesicles, which is sequentially provided with a fermentation tank, ultraviolet spectrophotometry equipment and irradiation equipment.
  • the bacteria are proliferated by fermentation in the fermentation tank and then passed through ultraviolet
  • the spectrophotometric equipment controls the bacterial content in the bacterial liquid to be within a certain limit, and then uses the irradiation equipment to irradiate the bacterial liquid with radiation to promote the bacteria to produce a large number of membrane vesicles.
  • the present invention also provides a purification system of bacterial membrane vesicles and a separation system of bacterial membrane vesicles.
  • the membrane vesicles prepared by the system have high yield, high purity and easy industrial production.
  • the present invention also discloses a production system and method for bacterial membrane vesicles.
  • the production system is provided with a fermentation tank, ultraviolet spectrophotometry equipment and irradiation equipment in sequence.
  • the bacteria proliferate in the fermentation tank and then The ultraviolet spectrophotometric equipment is used to control the bacterial content in the bacterial liquid within a certain limit, and then the irradiation equipment is used to irradiate the bacterial liquid with radiation to promote the bacteria to produce a large number of membrane vesicles, thereby increasing the content of membrane vesicles.
  • the present invention also provides a bacterial membrane vesicle separation and purification system and purification method.
  • the membrane vesicles prepared by the method have high yield, high purity and easy industrial production.
  • the present invention uses ionizing X-ray irradiation for the first time to isolate and purify the MVs produced by bacteria. No antibiotics and other chemical irritants are added, so the residual irritants and the adverse effects on the membrane vesicles caused by the irritants themselves are avoided.
  • the process flow is simple, suitable for industrial scale-up and standardized production; the vesicle has high yield, high efficiency, good amplification effect and purification effect, and can be used for the large-scale preparation of vesicles. Compared with normal vesicles, the prepared vesicles have better immunogenicity with a yield increase of dozens of times.
  • the bacterial membrane vesicles obtained after optimized production have a broad prospect for further development and application.
  • the bacterial membrane vesicles prepared by the present invention can be directly used as vaccines.
  • the bacterial membrane vesicle production system provided by the present invention can promote membrane vesicles that promote the mass production of bacteria, and is suitable for industrial production; the bacterial membrane vesicle purification and separation system provided by the present invention can remove fermentation waste liquid, bacterial flagella, and bacteria Secretions, etc., the obtained membrane vesicles have high purity.
  • FIG. 1 Transmission electron micrograph of Pseudomonas aeruginosa membrane vesicles (A: control group membrane vesicles; B: irradiation group membrane vesicles; scale bar: 200 nm).
  • Figure 2 Determination of the content of Pseudomonas aeruginosa membrane vesicles.
  • Figure 3 Pseudomonas aeruginosa membrane vesicle particle size distribution diagram.
  • Figure 4 Determination of the content of Staphylococcus aureus membrane vesicles.
  • Irradiated membrane vesicles promoted significant up-regulation of the fine surface molecules of bone marrow-derived dendritic cells CD80, CD86 and MHCII molecules.
  • Figure 6 is a bar graph of the phagocytic ability of DC cells stimulated by irradiated membrane vesicles.
  • Figure 7 Proliferation percentage of CD4+ T cells after interacting with DCs after different treatments.
  • Figure 8 Flow cytometry chart of the proliferation of CD4+ T cells after interacting with DCs after different treatments.
  • Irradiated membrane vesicles enhance the interaction between DC cells and T cells (GC: growth control, dendritic cell growth control group (unstimulated group); Cell+MVs (whole cell + vesicle treatment group); MVs (vesicle processing group)).
  • GC growth control, dendritic cell growth control group (unstimulated group); Cell+MVs (whole cell + vesicle treatment group); MVs (vesicle processing group)).
  • Figure 10 Schematic diagram of bacterial membrane vesicle production system structure.
  • FIG. 11 Schematic diagram of the structure of bacterial membrane vesicle purification system.
  • Figure 12 Schematic diagram of bacterial membrane vesicle separation system structure.
  • Invention A Bacterial membrane vesicle and its preparation method and application
  • the invention A belongs to the field of microbiology, and specifically relates to the preparation, separation and purification of a bacterial membrane vesicle and the application of the membrane vesicle.
  • Invention A provides a method for isolating bacterial MVs and a method for preparing bacterial MVs.
  • the invention does not add chemical stimulating substances, so no residues cause adverse effects, and at the same time, the process flow is simple, the vesicle output is high, the efficiency is high, and the amplification effect is good, and it can be used for mass preparation of vesicles. Compared with normal vesicles, the prepared vesicles have better immunogenicity, and the subsequent further development and application prospects are broad.
  • the purpose of the invention is to provide a bacterial membrane vesicle.
  • Biological particles produced by bacteria which are membrane vesicles isolated from inactivated bacteria.
  • the inactivated bacteria include Gram-positive bacteria, Gram-negative bacteria and genetically engineered bacteria.
  • the gram-positive bacteria include Staphylococcus aureus, Streptococcus, Diphtheria bacillus, Mycobacterium tuberculosis, Tetanus bacillus, Bacillus, Corynebacterium, and genetically engineered bacteria of the above bacteria.
  • the gram-negative bacteria include Pseudomonas aeruginosa, Acinetobacter baumannii, Escherichia coli, Typhoid bacillus, Meningococcus, Proteus, Shigella, and genetically engineered bacteria of the aforementioned bacteria.
  • the inactivated bacteria are anaerobic bacteria.
  • anaerobic bacteria include Helicobacter pylori, denitrifying bacteria or Fusobacterium nuclei.
  • a biological composition characterized by containing the above-mentioned membrane vesicles and the above-mentioned inactivated bacteria.
  • membrane vesicles are used as vaccines and/or vaccine adjuvants and/or drug carriers in combination with the above-mentioned inactivated bacteria.
  • the purpose of the invention is also to provide a method for separating membrane vesicles.
  • a method for separating membrane vesicles from bacteria the bacteria including gram-positive bacteria and gram-negative bacteria, specifically including the following steps:
  • the separation method in step 1) includes centrifugation, column passing or membrane packet concentration.
  • the supernatant 2 collected in the step 2) is concentrated through a membrane pack before entering the step 3).
  • the selected membrane pack can concentrate substances greater than 100KD.
  • the membrane vesicles are resuspended in a buffer solution, and the buffer solution is calculated in a volume of 1 L, including 50 mM Tris, 5 mM NaCl, 1 mM MgSO 4 , and the pH is 7.4.
  • a method for preparing a biological composition collecting the bacterial cells separated in step 1) in the above method for separating membrane vesicles, and mixing the bacterial cells with the membrane vesicles obtained in step 3) to form the biological composition .
  • the supernatant liquid 1 is filtered with a 0.3-0.5 ⁇ M filter to remove impurities.
  • the supernatant 1 is filtered with a 045 ⁇ M filter to remove impurities.
  • the separation method in the step 1) is centrifugation, the centrifugation rate of the centrifugation is 100-10000 g; the centrifugation time is 10-60 min.
  • the speed of centrifugation in step 1) is 400-8000 g; the centrifugation time is 10-30 min.
  • the high-speed centrifugation rate in the step 2) is 5000-25000g; the centrifugation time is 10-100min.
  • the high-speed centrifugation rate in step 2) is 10000-20000g; the centrifugation time is 30-60min.
  • the ultra-high-speed centrifugation rate in the step 3) is 5000-150000g; the centrifugation time is 60-600min.
  • the ultrahigh-speed centrifugation rate in step 3) is 15000-150000g; the centrifugation time is 60-180min.
  • the gram-positive bacteria include Staphylococcus aureus, Streptococcus, Diphtheria bacillus, Mycobacterium tuberculosis, Tetanus bacillus, Bacillus, Corynebacterium, and genetically engineered bacteria of the above bacteria.
  • the gram-negative bacteria include Pseudomonas aeruginosa, Acinetobacter baumannii, Escherichia coli, Typhoid bacillus, Meningococcus, Proteus, Shigella, and genetically engineered bacteria of the aforementioned bacteria.
  • bacteria are inactivated bacteria.
  • the purpose of the present invention is also to provide a preparation method.
  • Cultivate the bacteria to the logarithmic growth phase collect the bacteria, resuspend the bacteria and irradiate the bacteria with ionizing rays to obtain the irradiated bacteria;
  • the ionizing radiation treatment method is X-ray radiation, and the radiation dose is 500-3000 Gy.
  • the radiation dose specifically includes: 500-600Gy, 600-700Gy, 700-800Gy, 800-900Gy, 900-1000Gy, 1000-1100Gy, 1100-1200Gy, 1200-1300Gy, 1300-1400Gy, 1400-1500Gy, 1500- 1600Gy, 1600-1700Gy, 1700-1800Gy, 1800-1900Gy, 1900-2000Gy, 2100-2200Gy, 2200-2300Gy, 2300-2400Gy, 2400-2500Gy, 2500-2600Gy, 2600-2700Gy, 2700-2800Gy, 2800-2900Gy, 2900-3000Gy.
  • the radiation dose is 500-1000 Gy.
  • the radiation dose specifically includes: 500-600 Gy, 600-700 Gy, 700-800 Gy, 800-900 Gy, 900-1000 Gy.
  • the OD 600 value of the bacteria in the logarithmic growth phase in the step 1) is 0.3-0.8.
  • the OD 600 value of the bacteria in the logarithmic growth phase in the step 1) is 0.5-0.7.
  • the bacterial cells are resuspended with phosphate buffer or sterile normal saline.
  • the bacteria are resuspended in phosphate buffer.
  • the bacterial weight is suspended to an OD 600 value of 20-80.
  • the bacterial weight is suspended to an OD 600 value of 40-60.
  • the nucleic acid content and the protein content in the membrane vesicles are increased by 10-20 times compared with bacteria not irradiated with ionizing rays.
  • the purpose of the present invention is also to provide the application of membrane vesicles and inactivated bacteria.
  • membrane vesicles prepared by invention A are used as immunogens.
  • membrane vesicles of membrane vesicles prepared by the invention as an immune response enhancer.
  • the membrane vesicles of the membrane vesicles prepared by the invention A are used as vaccines for the treatment of bacterial infections.
  • the bacterial infection disease includes pneumonia, urinary tract infection, meningitis, sepsis, or skin and soft tissue infections.
  • the vaccine adjuvant is to non-specifically change or enhance the body's specific immune response to the antigen.
  • antigen presenting cells dendritic cells, macrophages, and B cells.
  • Membrane vesicles obtained after irradiation and separation and purification are used as DC cell maturation promoters.
  • membrane vesicles are used as promoters to promote the significant up-regulation of the fine surface molecules of bone marrow-derived dendritic cells CD80, CD86 and MHCII molecules.
  • membrane vesicles prepared by the invention in combination with DC cells in the preparation of CD4+ T cell proliferation agents.
  • the membrane vesicles prepared by irradiation treatment to stimulate and phagocytose the OVA antigen DCs and CFSE-labeled CD4+ T lymphocytes are co-cultured in vitro.
  • Invention A provides a method for isolating and preparing bacterial membrane vesicles, the method comprising the following steps:
  • the OD 600 value of the bacteria in the logarithmic growth phase is 0.3-0.8; it is best to choose an OD 600 value of 0.5-0.8 (the fermentation can also be carried out here to further enrich the bacteria ); Collect the bacteria, resuspend the bacteria with an appropriate amount of phosphate buffer, the ratio of the amount of phosphate buffer added to the total number of bacteria is the amount of bacteria contained per 1ml solution is an OD 600 value of 20- 80; It is best to choose an OD 600 value of 40-60; after resuspension, use ionizing radiation to irradiate the bacteria; it is best to use X-ray radiation, and the radiation dose is 500-3000Gy.
  • the irradiation dose is 1000Gy.
  • step 7 Centrifuge the supernatant of step 6) in a high-speed centrifuge to remove the flagella in the supernatant.
  • step 8 Centrifuge the supernatant of step 7) in an ultra-high-speed centrifuge to precipitate membrane vesicles.
  • ionizing rays can stimulate Pseudomonas aeruginosa PAO1 to produce membrane vesicles.
  • nucleic acid content and protein content of the membrane vesicles prepared in the experimental group were increased 10-20 times compared with the normal control group. See Figure 2. See Table 1 for specific measurement data.
  • the particle size distribution data showed that there was no significant difference in the size of membrane vesicles prepared in the normal control group and the experimental group, with an average of about 150 nm. See Figure 3; see Table 2 for specific measurement data.
  • Ionizing rays irradiate Staphylococcus aureus ATCC 25923 to prepare membrane vesicles:
  • the irradiation dose is 1000Gy.
  • step 7 Centrifuge the supernatant of step 6) in an ultra-high-speed centrifuge, 39,000 ⁇ g, 90 min, and precipitate MVs.
  • Dendritic cells are the body's main antigen presenting cells, whose main function is to swallow, process and process antigen molecules, and present them to T cells. It is known to have the strongest function in the body and the only full-time antigen-presenting cell that can activate resting T cells. It is the central link in initiating, regulating and maintaining the immune response. The maturity of dendritic cells determines the body's immune response or immune tolerance.
  • the main manifestations are changes in the expression of costimulatory molecules CD80 and CD86, the weakened ability of phagocytosis of antigens and the enhanced ability of processing and presenting antigens (increased expression of MHCII molecules), and the interaction with T lymphocytes.
  • BMDC Mouse bone marrow-derived dendritic cells
  • BMDC stimulation Take the 7-day-induced BMDC cells, pipette repeatedly in a 6-well plate to shed adherent cells, collect the cell suspension, centrifuge at 1100 rpm for 5 min, remove the supernatant, add 1ml medium to resuspend the cells and count the viable cells. Adjust the cell concentration to 1 ⁇ 10 6 /ml, and inoculate 2 ml to a new 6-well plate. Add each stimulus and mix, respectively: whole bacteria, whole bacteria + vesicles, vesicles, so that the final concentration is 15 ⁇ g/mL (protein as standard), continue to incubate for 24 hours, and add equal volume of PBS to the growth control group .
  • Flow cytometry mature marker Take out the 6-well plate after 24h, repeatedly pipette the cells to detach, collect the cell suspension into the flow tube, centrifuge at 1500rpm for 3min, remove the supernatant, add 1ml PBS and continue centrifugation at 1500rpm for 3min, then remove the supernatant , Repeated cleaning 3 times. Add CD11c/CD80/CD86/MHCII antibody and incubate at room temperature for 30 minutes in the dark. At the same time, the negative control group should set up an isotype control group to join CD11c/CD80/CD86/MHCII peer control. After the incubation is completed, add PBS to wash twice, then add 200 ⁇ l PBS to resuspend the cells, and check on the flow cytometer.
  • MVs X-ray-treated experimental group
  • DC cells have strong antigen endocytosis and processing capabilities. It is in an immature state when it is not in contact with the antigen material, its phagocytic ability is strong, and it becomes mature after being exposed to the antigen, its phagocytic ability becomes weak, and the antigen presentation ability is enhanced.
  • This experiment detects FITC-labeled dextran fluorescence The intensity is used to determine the amount of phagocytic dextran of the DC to detect whether the phagocytic ability of the DC is enhanced.
  • Stimulation Collect the cells on the 7th day, blow down all the cells, centrifuge and resuspend the counts, then inoculate them into a 6-well plate, inoculate 1 ⁇ 10 6 cells per well, and then add stimuli respectively, and add an equal volume of GC group PBS, Control group and Treatment group were added with the same concentration of membrane vesicles (according to protein level) and cultured at 37°C for 24h.
  • Phagocytosis and detection add dextran (5 ⁇ g/ml), continue culturing for 1h, aspirate the cells to the flow tube, wash 3 times with PBS, add CD11c antibody and incubate at room temperature for 30min in the dark, wash 3 times with PBS, then flow cytometry FITC fluorescence.
  • FITC-dextran as a model antigen for DCs to phagocytose, and detect the FITC average fluorescence intensity value of CD11c+DCs.
  • the experimental results showed that the dendritic cells of the GC group (growth control group) basically did not take up FITC-dextran, but after DCs were stimulated, the average fluorescence intensity of FITC was significantly lower than that of the GC group. This experimental result once again proves that vesicles can promote the maturation of DCs, thereby reducing the ability to uptake antigen. See Figure 6.
  • CFSE-labeled fluorescence can be evenly distributed to the two progeny cells, and its fluorescence intensity is half that of the parent cells. Therefore, we can use flow cytometry to calculate the percentage of cells with weaker CFSE fluorescence to obtain the percentage of proliferating cells.
  • Antigen phagocytosis The DCs cultured for 7 days were cultured in a medium containing 10 ⁇ g/ml OVA for 24 hours as a GC (growth control, growth control). The MVs group was additionally added with vesicles, and then the DCs that had swallowed the antigen were collected by centrifugation Resuspend them in normal culture medium, and plate them in a 96-well plate at a density of 2 ⁇ 10 4 cells/well, with 100 ⁇ l per well, and 3 replicate wells per group.
  • T cell extraction On the second day, CD4+ T lymphocytes specific to OVA in the spleen of OT-II mice were separated and enriched using the negative magnetic bead screening kit of Stem Cell Technologies.
  • DCs stimulated by vesicles and phagocytosed the OVA antigen were co-cultured with CFSE-labeled OT-II mouse CD4+ T lymphocytes in vitro.
  • Flow cytometry analysis of CFSE fluorescence intensity results showed that the proportion of proliferating CD4+ T cells increased.
  • Vesicles (14.05%) can significantly increase the proliferation of specific CD4+ T cells by DCs (6.80%) that phagocytose OVA antigen. See Figure 7 and Figure 8 for details.
  • CFSE Fluorescent dye CFSE
  • CFDA-SE hydroxyfluorescein diacetate succinimidyl ester
  • CFSE-labeled fluorescence can be evenly distributed to the two progeny cells, and its fluorescence intensity is half that of the parent cells. Therefore, we can use flow cytometry to calculate the percentage of cells with weak CFSE fluorescence to obtain the proportion of proliferating cells.
  • Antigen phagocytosis The DCs cultured for 7 days are cultured in the medium for 24 hours as GC (growth control, growth control). The MVs group is additionally added with vesicles, and then the DCs that have swallowed the antigen are collected by centrifugation and resuspended in normal culture The base is plated in a 96-well plate at a density of 4 ⁇ 10 4 cells/well, 100 ⁇ l per well, 3 replicate wells per group.
  • T cell extraction On the second day, the mouse spleen one week after immunization with MVs was used to isolate and enrich the mouse T cells using the negative magnetic bead screening kit of Stem Cell Technologies.
  • DCs stimulated by vesicles and phagocytosed the OVA antigen were co-cultured with CFSE-labeled OT-II mouse CD4+ T lymphocytes in vitro.
  • Flow cytometry analysis of CFSE fluorescence intensity results showed that the proportion of proliferating CD4+ T cells increased.
  • the fluorescence intensity of the bacteria plus vesicle stimulation group was 63.5%, and the vesicle stimulation group was 71%. It shows that DCs after vesicle treatment can significantly stimulate the proliferation of CD4+ T cells. See Figure 9.
  • Invention B Pseudomonas aeruginosa membrane vesicle and its preparation method and application
  • Invention B belongs to the field of microbiology, and specifically relates to the preparation, separation and purification of membrane vesicles of Pseudomonas aeruginosa and the application of the membrane vesicles.
  • Invention B provides a method for separating Pseudomonas aeruginosa MVs and a method for preparing Pseudomonas aeruginosa MVs.
  • the invention adopts the international leading process technology, does not add chemical stimulating substances, and therefore has no adverse effects.
  • the process flow is simple, the vesicle yield is high, the efficiency is high, and the amplification effect is good, and it can be used for mass preparation of vesicles.
  • the prepared vesicles have better immunogenicity, and the subsequent further development and application prospects are broad.
  • the purpose of the B invention is to provide a vaccine against Pseudomonas aeruginosa.
  • Pseudomonas aeruginosa vaccine containing membrane vesicles isolated from inactivated Pseudomonas aeruginosa.
  • a biological composition containing separated membrane vesicles and inactivated Pseudomonas aeruginosa A biological composition containing separated membrane vesicles and inactivated Pseudomonas aeruginosa.
  • the membrane vesicle is used as a vaccine and/or vaccine adjuvant and/or drug carrier in combination with inactivated Pseudomonas aeruginosa.
  • the purpose of the B invention is also to provide a method for isolating Pseudomonas aeruginosa membrane vesicles.
  • a method for separating membrane vesicles from Pseudomonas aeruginosa includes the following steps:
  • step 2 was filtered and sterilized with a 0.45 ⁇ M filter.
  • the centrifugation rate in step 2) is 100-10000 g; the centrifugation time is 10-60 min.
  • the centrifugation rate is 400-8000 g; the centrifugation time is 10-30 min.
  • the high-speed centrifugation rate of step 3 is 5000-25000g; the centrifugation time is 10-100min.
  • the high-speed centrifugation rate is 10000-20000g; the centrifugation time is 30-60min.
  • the ultra-high-speed centrifugation rate in step 4) is 5000-150000g; the centrifugation time is 60-600min.
  • the ultra-high speed centrifugation speed is 15000-150000g; the centrifugation time is 60-180min.
  • Pseudomonas aeruginosa is an inactivated Pseudomonas aeruginosa.
  • Pseudomonas aeruginosa membrane vesicles obtained by the above isolation method.
  • the purpose of the B invention is also to provide a preparation method.
  • the method for preparing the above-mentioned Pseudomonas aeruginosa vaccine includes the following steps:
  • the OD 600 value of the bacteria in the logarithmic growth phase in step 1) is 0.3-0.8.
  • the ratio of the amount of phosphate buffer or sterile physiological saline added in step 2) to the total amount of bacteria is that the amount of bacteria contained in 1 ml of the solution is an OD 600 value of 20-80.
  • the radiation used for the irradiation treatment in step 3) is X-ray rays; the radiation dose is 500-3000 Gy.
  • the radiation dose specifically includes: 500-600Gy, 600-700Gy, 700-800Gy, 800-900Gy, 900-1000Gy, 1000-1100Gy, 1100-1200Gy, 1200-1300Gy, 1300-1400Gy, 1400-1500Gy, 1500- 1600Gy, 1600-1700Gy, 1700-1800Gy, 1800-1900Gy, 1900-2000Gy, 2100-2200Gy, 2200-2300Gy, 2300-2400Gy, 2400-2500Gy, 2500-2600Gy, 2600-2700Gy, 2700-2800Gy, 2800-2900Gy, 2900-3000Gy.
  • the radiation dose is 500-1000 Gy.
  • the radiation dose specifically includes: 500-600 Gy, 600-700 Gy, 700-800 Gy, 800-900 Gy, 900-1000 Gy.
  • the centrifugation rate in step 3 is 100-10000 g; the centrifugation time is 10-60 min.
  • the centrifugation rate is 400-8000 g; the centrifugation time is 10-30 min.
  • step 3 the supernatant was filtered and sterilized with a 0.45 ⁇ M filter.
  • the high-speed centrifugation rate described in step 4) is 5000-25000g; the centrifugation time is 10-100min.
  • the high-speed centrifugation rate is 10000-20000g; the centrifugation time is 30-60min.
  • the ultra-high-speed centrifugation rate described in step 5 is 5000-150000g; the centrifugation time is 60-600min.
  • the ultra-high speed centrifugation speed is 15000-150000g; the centrifugation time is 60-180min.
  • the content of nucleic acid and protein in the membrane vesicles of the above-mentioned Pseudomonas aeruginosa membrane vesicles is increased 10-20 times.
  • a method for increasing the content of Pseudomonas aeruginosa membrane vesicles, the content including nucleic acid and protein uses an irradiation device to process the Pseudomonas aeruginosa bacterial liquid, and the radiation of the irradiation device is X-ray rays, the irradiation dose is 500-1000Gy.
  • the purpose of the B invention is also to provide an application of the Pseudomonas aeruginosa membrane vesicles and inactivated Pseudomonas aeruginosa.
  • the Pseudomonas aeruginosa membrane vesicles prepared by the B invention (hereinafter referred to as the Pseudomonas aeruginosa membrane vesicles prepared by the B invention) in the preparation of anti-bacterial infection vaccines, the Pseudomonas aeruginosa membrane vesicles Foam can be used as an adjuvant for vaccines.
  • the vaccine adjuvant can non-specifically change or enhance the body's specific immune response to the antigen.
  • the bacterial infection disease includes pneumonia, urinary tract infection, meningitis, sepsis, or skin and soft tissue infections.
  • Pseudomonas aeruginosa membrane vesicles can also be used as a vaccine carrier.
  • antigen presenting cells include dendritic cells, macrophages, and B cells.
  • a method for promoting the proliferation of CD4+ T cells is to co-culture DCs prepared by irradiation with Pseudomonas aeruginosa membrane vesicles to stimulate and phagocytose OVA antigens and CD4+ T lymphocytes labeled with CFSE in vitro.
  • Invention B provides a system for separating and preparing Pseudomonas aeruginosa membrane vesicles.
  • the system is sequentially provided with a fermentation tank, an irradiation device, an ultraviolet spectrophotometer device, and a centrifugal device;
  • the radiation generator of the irradiation device is One or more of X-ray generator, ⁇ -ray generator, or Co 60 isotope generator;
  • the centrifugal equipment includes one or more of centrifuges, high-speed centrifuges, and ultra-high-speed centrifuges, see figure 10.
  • Invention B provides a method for preparing Pseudomonas aeruginosa membrane vesicles, the method comprising the following steps:
  • the OD 600 value of the bacteria in the logarithmic growth phase is 0.3-0.8; it is best to choose the OD 600 value of 0.5-0.8 (here, fermentation can also be carried out to further enrich the bacteria set);
  • the radiation dose is 500-3000Gy, specifically including: 500-600Gy, 600-700Gy, 700-800Gy, 800-900Gy, 900-1000Gy, 1000-1100Gy, 1100-1200Gy, 1200-1300Gy, 1300-1400Gy, 1400-1500Gy, 1500-1600Gy, 1600-1700Gy, 1700-1800Gy, 1800-1900Gy, 1900-2000Gy, 2100-2200Gy, 2200-2300Gy, 2300-2400Gy, 2400-2500Gy, 2500- 2600Gy, 2600-2700Gy, 2700-2800Gy, 2800-2900Gy, 2900-3000Gy;
  • Invention B provides a method for separating and purifying membrane vesicles of Pseudomonas aeruginosa
  • the method includes the following steps:
  • the irradiation dose is 1000Gy.
  • step 7 Centrifuge the supernatant of step 6) in a high-speed centrifuge to remove the flagella in the supernatant.
  • step 8 Centrifuge the supernatant of step 7) in an ultra-high-speed centrifuge to precipitate membrane vesicles.
  • ionizing rays can stimulate Pseudomonas aeruginosa PAO1 to produce membrane vesicles.
  • nucleic acid content and protein content of the membrane vesicles prepared in the experimental group were increased 10-20 times compared with the normal control group. See Figure 2. See Table 1 for specific measurement data.
  • BMDC Mouse bone marrow-derived dendritic cells
  • BMDC stimulation Take the 7-day-induced BMDC cells, pipette repeatedly in a 6-well plate to shed adherent cells, collect the cell suspension, centrifuge at 1100 rpm for 5 min, remove the supernatant, add 1ml medium to resuspend the cells and count the viable cells. Adjust the cell concentration to 1 ⁇ 10 6 /ml, and inoculate 2 ml to a new 6-well plate. Add each stimulus and mix, respectively: whole bacteria, whole bacteria + vesicles, vesicles, so that the final concentration is 15 ⁇ g/mL (protein as standard), continue to incubate for 24 hours, and add equal volume of PBS to the growth control group .
  • Flow cytometry mature marker Take out the 6-well plate after 24h, repeatedly pipette the cells to detach, collect the cell suspension into the flow tube, centrifuge at 1500rpm for 3min, remove the supernatant, add 1ml PBS and continue centrifugation at 1500rpm for 3min, then remove the supernatant , Repeated cleaning 3 times. Add CD11c/CD80/CD86/MHCII antibody and incubate at room temperature for 30 minutes in the dark. At the same time, the negative control group should set up an isotype control group to join CD11c/CD80/CD86/MHCII peer control. After the incubation is completed, add PBS to wash twice, then add 200 ⁇ l PBS to resuspend the cells, and check on the flow cytometer.
  • MVs X-ray-treated experimental group
  • Stimulation Collect the cells on the 7th day, blow down all the cells, centrifuge and resuspend the counts, then inoculate them into a 6-well plate, inoculate 1 ⁇ 10 6 cells per well, and then add stimuli respectively, and add an equal volume of GC group PBS, Control group and Treatment group were added with the same concentration of membrane vesicles (according to protein level) and cultured at 37°C for 24h.
  • Phagocytosis and detection add dextran (5 ⁇ g/ml), continue culturing for 1h, aspirate the cells to the flow tube, wash 3 times with PBS, add CD11c antibody and incubate at room temperature for 30min in the dark, wash 3 times with PBS, then flow cytometry FITC fluorescence.
  • FITC-dextran as a model antigen for DCs to phagocytose, and detect the FITC average fluorescence intensity value of CD11c+DCs.
  • the experimental results showed that the dendritic cells of the GC group (growth control group) basically did not take up FITC-dextran, but after DCs were stimulated, the average fluorescence intensity of FITC was significantly lower than that of the GC group. This experimental result once again proves that vesicles can promote the maturation of DCs, thereby reducing the ability to uptake antigen. See Figure 6.
  • Antigen phagocytosis The DCs cultured for 7 days were cultured in a medium containing 10 ⁇ g/ml OVA for 24 hours as a GC (growth control, growth control). The MVs group was additionally added with vesicles, and then the DCs that had swallowed the antigen were collected by centrifugation Resuspend them in normal culture medium, and plate them in a 96-well plate at a density of 2 ⁇ 10 4 cells/well, with 100 ⁇ l per well, and 3 replicate wells per group.
  • T cell extraction On the second day, CD4+ T lymphocytes specific to OVA in the spleen of OT-II mice were separated and enriched using the negative magnetic bead screening kit of Stem Cell Technologies.
  • DCs stimulated by vesicles and phagocytosed the OVA antigen were co-cultured with CFSE-labeled OT-II mouse CD4+ T lymphocytes in vitro.
  • Flow cytometry analysis of CFSE fluorescence intensity results showed that the proportion of proliferating CD4+ T cells increased.
  • Vesicles (14.05%) can significantly increase the proliferation of specific CD4+ T cells by DCs (6.80%) that phagocytose OVA antigen. See Figure 7 and Figure 8 for details.
  • Antigen phagocytosis The DCs cultured for 7 days are cultured in the medium for 24 hours as GC (growth control, growth control). The MVs group is additionally added with vesicles, and then the DCs that have swallowed the antigen are collected by centrifugation and resuspended in normal culture The base is plated in a 96-well plate at a density of 4 ⁇ 10 4 cells/well, 100 ⁇ l per well, 3 replicate wells per group.
  • T cell extraction On the second day, the mouse spleen one week after immunization with MVs was used to isolate and enrich the mouse T cells using the negative magnetic bead screening kit of Stem Cell Technologies.
  • DCs stimulated by vesicles and phagocytosed the OVA antigen were co-cultured with CFSE-labeled OT-II mouse CD4+ T lymphocytes in vitro.
  • Flow cytometry analysis of CFSE fluorescence intensity results showed that the proportion of proliferating CD4+ T cells increased.
  • the fluorescence intensity of the bacteria plus vesicle stimulation group was 63.5%, and the vesicle stimulation group was 71%. It shows that DCs after vesicle treatment can significantly stimulate the proliferation of CD4+ T cells. See Figure 9.
  • Invention C Staphylococcus aureus membrane vesicles and preparation method and application thereof
  • Invention C belongs to the field of microbiology, and specifically relates to the preparation, separation and purification of Staphylococcus aureus membrane vesicles and the application of the membrane vesicles.
  • Invention C provides a method for separating MVs of Staphylococcus aureus and a method for preparing MVs of Staphylococcus aureus.
  • the invention adopts the international leading process technology, does not add chemical stimulating substances, and therefore has no adverse effects.
  • the process flow is simple, the vesicle yield is high, the efficiency is high, and the amplification effect is good, and it can be used for mass preparation of vesicles.
  • the prepared vesicles have better immunogenicity, and the subsequent further development and application prospects are broad.
  • the purpose of the C invention is to provide a Staphylococcus aureus membrane vesicle.
  • Biological particles produced by Staphylococcus aureus which are membrane vesicles isolated from inactivated Staphylococcus aureus.
  • the inactivated Staphylococcus aureus is combined with the biological particles that act as vaccines and/or vaccine adjuvants and/or drug carriers.
  • the object of the C invention is also to provide a method for separating membrane vesicles of Staphylococcus aureus.
  • a method for separating membrane vesicles from Staphylococcus aureus includes the following steps:
  • step 2 was filtered and sterilized with a 0.45 ⁇ M filter.
  • the centrifugation rate in step 2) is 100-10000 g; the centrifugation time is 10-60 min.
  • the centrifugation rate is 400-8000 g; the centrifugation time is 10-30 min.
  • the high-speed centrifugation rate of step 3 is 5000-25000g; the centrifugation time is 10-100min.
  • the high-speed centrifugation rate is 10000-20000g; the centrifugation time is 30-60min.
  • the ultra-high-speed centrifugation rate in step 4) is 5000-150000g; the centrifugation time is 60-600min.
  • the ultra-high speed centrifugation speed is 15000-150000g; the centrifugation time is 60-180min.
  • Staphylococcus aureus is an inactivated Staphylococcus aureus.
  • Staphylococcus aureus membrane vesicles obtained by the above isolation method.
  • the purpose of the C invention is also to provide a preparation method.
  • the method for preparing the above-mentioned biological particles includes the following steps:
  • the OD 600 value of the bacteria in the logarithmic growth phase in step 1) is 0.3-0.8.
  • the ratio of the amount of phosphate buffer or sterile physiological saline added in step 2) to the total amount of bacteria is that the amount of bacteria contained in 1 ml of the solution is an OD 600 value of 20-80.
  • the radiation used for the irradiation treatment in step 2) is X-ray rays; the radiation dose is 500-3000 Gy.
  • the radiation dose specifically includes: 500-600Gy, 600-700Gy, 700-800Gy, 800-900Gy, 900-1000Gy, 1000-1100Gy, 1100-1200Gy, 1200-1300Gy, 1300-1400Gy, 1400-1500Gy, 1500- 1600Gy, 1600-1700Gy, 1700-1800Gy, 1800-1900Gy, 1900-2000Gy, 2100-2200Gy, 2200-2300Gy, 2300-2400Gy, 2400-2500Gy, 2500-2600Gy, 2600-2700Gy, 2700-2800Gy, 2800-2900Gy, 2900-3000Gy.
  • the radiation dose is 500-1000 Gy.
  • the radiation dose specifically includes: 500-600 Gy, 600-700 Gy, 700-800 Gy, 800-900 Gy, 900-1000 Gy.
  • the centrifugation rate in step 3 is 100-10000 g; the centrifugation time is 10-60 min.
  • the centrifugation rate is 400-8000 g; the centrifugation time is 10-30 min.
  • step 3 the supernatant was filtered and sterilized with a 0.45 ⁇ M filter.
  • the high-speed centrifugation rate described in step 4) is 5000-25000g; the centrifugation time is 10-100min.
  • the high-speed centrifugation rate is 10000-20000g; the centrifugation time is 30-60min.
  • the ultra-high-speed centrifugation rate described in step 5 is 5000-150000g; the centrifugation time is 60-600min.
  • the ultra-high speed centrifugation speed is 15000-150000g; the centrifugation time is 60-180min.
  • the membrane vesicles of Staphylococcus aureus are prepared by the above method.
  • a method for increasing the content of Staphylococcus aureus membrane vesicles, the content including nucleic acid and protein uses an irradiation device to process the Staphylococcus aureus bacterium liquid, and the radiation of the irradiation device is X- ray, the radiation dose is 500-1000Gy.
  • the purpose of the C invention is also to provide the application of the Staphylococcus aureus membrane vesicles and inactivated bacteria.
  • the Staphylococcus aureus membrane vesicles prepared by the invention C (hereinafter referred to as the Staphylococcus aureus membrane vesicles prepared by the invention C) in the preparation of anti-bacterial infection vaccines, the Staphylococcus aureus membrane vesicles can be used as Adjuvant use of vaccines.
  • the vaccine adjuvant can non-specifically change or enhance the body's specific immune response to the antigen.
  • the bacterial infection disease includes pneumonia, urinary tract infection, meningitis, sepsis, or skin and soft tissue infections.
  • Staphylococcus aureus membrane vesicles can also be used as vaccine carriers.
  • Staphylococcus aureus membrane vesicles prepared by the invention C as an antigen presenting cell function promoter.
  • antigen presenting cells include dendritic cells, macrophages, and B cells.
  • Staphylococcus aureus membrane vesicles can stimulate the significant up-regulation of the costimulatory molecules CD80, CD86 and MHCII on the surface of DC cells and promote DC Cell maturation and differentiation.
  • Staphylococcus aureus membrane vesicles prepared by the invention of C as a DC cell antigen presentation ability enhancer.
  • composition for promoting the proliferation of CD4 and T cells comprising the above-mentioned Staphylococcus aureus membrane vesicles and DC cells.
  • a method for promoting the proliferation of CD4+ T cells is to co-culture DCs prepared by irradiation with Staphylococcus aureus membrane vesicles to stimulate and phagocytose OVA antigen and CD4+ T lymphocytes labeled with CFSE in vitro.
  • Invention C provides a system for separating and preparing Staphylococcus aureus membrane vesicles.
  • the system is provided with a fermentation tank, an irradiation device, an ultraviolet spectrophotometer device, and a centrifugal device in sequence;
  • the radiation generator of the irradiation device is X One or more of a ray generator, a gamma ray generator, or a Co 60 isotope generator;
  • the centrifugal equipment includes one or more of a centrifuge, a high-speed centrifuge, and an ultra-high-speed centrifuge, see Figure 10 .
  • Invention C provides a method for preparing membrane vesicles of Staphylococcus aureus, the method comprising the following steps:
  • the radiation dose is 500-3000Gy, specifically including: 500-600Gy, 600-700Gy, 700-800Gy, 800-900Gy, 900-1000Gy, 1000-1100Gy, 1100-1200Gy, 1200-1300Gy, 1300-1400Gy, 1400-1500Gy, 1500-1600Gy, 1600-1700Gy, 1700-1800Gy, 1800-1900Gy, 1900-2000Gy, 2100-2200Gy, 2200-2300Gy, 2300-2400Gy, 2400-2500Gy, 2500- 2600Gy, 2600-2700Gy, 2700-2800Gy, 2800-2900Gy, 2900-3000Gy;
  • Invention C provides a method for separating membrane vesicles of Staphylococcus aureus, the method comprising the following steps:
  • Ionizing rays irradiate Staphylococcus aureus ATCC 25923 to prepare membrane vesicles:
  • the irradiation dose is 1000Gy.
  • step 7 Centrifuge the supernatant of step 6) in an ultra-high-speed centrifuge, 39,000 ⁇ g, 90 min, and precipitate MVs.
  • BMDC Mouse bone marrow-derived dendritic cells
  • BMDC stimulation Take the 7-day-induced BMDC cells, pipette repeatedly in a 6-well plate to shed adherent cells, collect the cell suspension, centrifuge at 1100 rpm for 5 min, remove the supernatant, add 1ml medium to resuspend the cells and count the viable cells. Adjust the cell concentration to 1 ⁇ 10 6 /ml, and inoculate 2 ml to a new 6-well plate. Add each stimulus and mix, respectively: whole bacteria, whole bacteria + vesicles, vesicles, so that the final concentration is 15 ⁇ g/mL (protein as standard), continue to incubate for 24 hours, and add equal volume of PBS to the growth control group .
  • Flow cytometry mature marker Take out the 6-well plate after 24h, repeatedly pipette the cells to detach, collect the cell suspension into the flow tube, centrifuge at 1500rpm for 3min, remove the supernatant, add 1ml PBS and continue centrifugation at 1500rpm for 3min, then remove the supernatant , Repeated cleaning 3 times. Add CD11c/CD80/CD86/MHCII antibody and incubate at room temperature for 30 minutes in the dark. At the same time, the negative control group should set up an isotype control group to join CD11c/CD80/CD86/MHCII peer control. After the incubation is completed, add PBS to wash twice, then add 200 ⁇ l PBS to resuspend the cells, and check on the flow cytometer.
  • MVs X-ray-treated experimental group
  • Stimulation Collect the cells on the 7th day, blow down all the cells, centrifuge and resuspend the counts, then inoculate them into a 6-well plate, inoculate 1 ⁇ 10 6 cells per well, and then add stimuli respectively, and add an equal volume of GC group PBS, Control group and Treatment group were added with the same concentration of membrane vesicles (according to protein level) and cultured at 37°C for 24h.
  • Phagocytosis and detection add dextran (5 ⁇ g/ml), continue culturing for 1h, aspirate the cells to the flow tube, wash 3 times with PBS, add CD11c antibody and incubate at room temperature for 30min in the dark, wash 3 times with PBS, then flow cytometry FITC fluorescence.
  • FITC-dextran as a model antigen for DCs to phagocytose, and detect the FITC average fluorescence intensity value of CD11c+DCs.
  • the experimental results showed that the dendritic cells of the GC group (growth control group) basically did not take up FITC-dextran, but after DCs were stimulated, the average fluorescence intensity of FITC was significantly lower than that of the GC group. This experimental result once again proves that vesicles can promote the maturation of DCs, thereby reducing the ability of antigen uptake. See Figure 6.
  • Antigen phagocytosis The DCs cultured for 7 days were cultured in a medium containing 10 ⁇ g/ml OVA for 24 hours as a GC (growth control, growth control). The MVs group was additionally added with vesicles, and then the DCs that had swallowed the antigen were collected by centrifugation Resuspend them in normal culture medium, and plate them in a 96-well plate at a density of 2 ⁇ 10 4 cells/well, with 100 ⁇ l per well, and 3 replicate wells per group.
  • T cell extraction On the second day, CD4+ T lymphocytes specific to OVA in the spleen of OT-II mice were separated and enriched using the negative magnetic bead screening kit of Stem Cell Technologies.
  • DCs stimulated by vesicles and phagocytosed the OVA antigen were co-cultured with CFSE-labeled OT-II mouse CD4+ T lymphocytes in vitro.
  • Flow cytometry analysis of CFSE fluorescence intensity results showed that the proportion of proliferating CD4+ T cells increased.
  • Vesicles (14.05%) can significantly increase the proliferation of specific CD4+ T cells by DCs (6.80%) that phagocytose OVA antigen. See Figure 7 and Figure 8 for details.
  • Antigen phagocytosis The DCs cultured for 7 days are cultured in the medium for 24 hours as GC (growth control, growth control). The MVs group is additionally added with vesicles, and then the DCs that have swallowed the antigen are collected by centrifugation and resuspended in normal culture The base is plated in a 96-well plate at a density of 4 ⁇ 10 4 cells/well, 100 ⁇ l per well, 3 replicate wells per group.
  • T cell extraction On the second day, the mouse spleen one week after immunization with MVs was used to isolate and enrich the mouse T cells using the negative magnetic bead screening kit of Stem Cell Technologies.
  • DCs stimulated by vesicles and phagocytosed the OVA antigen were co-cultured with CFSE-labeled OT-II mouse CD4+ T lymphocytes in vitro.
  • Flow cytometry analysis of CFSE fluorescence intensity results showed that the proportion of proliferating CD4+ T cells increased.
  • the fluorescence intensity of the bacteria plus vesicle stimulation group was 63.5%, and the vesicle stimulation group was 71%. It shows that DCs after vesicle treatment can significantly stimulate the proliferation of CD4+ T cells. See Figure 9.
  • Invention D A production system and separation and purification system for bacterial membrane vesicles
  • Invention D relates to the field of biotechnology, and has a production system and a separation and purification system involving a bacterial membrane vesicle.
  • Invention D provides a bacterial membrane vesicle production system and purification system that is easy to industrially use.
  • a production system for bacterial membrane vesicles is sequentially provided with a fermentation unit 1 and an irradiation unit 2.
  • the fermentation unit 1 is composed of a bacterial fermentation tank 3 and an ultraviolet spectrophotometer 4, and the irradiation unit 2 Consists of irradiation equipment 5.
  • the fermentation tank provided by D invention is suitable for the fermentation of various bacteria, including gram-positive bacteria and gram-negative bacteria.
  • the ultraviolet spectrophotometric equipment provided by D invention is used to accurately measure the bacterial content of the bacterial liquid.
  • the fermentation tank 3 is a ventilated fermentation tank, a bubbling fermentation tank, an air-lift fermentation tank, or a nozzle circulation type fermentation tank.
  • the ray emitter in the irradiation device 5 is an X-ray generator, a ⁇ -ray generator, or a Co 60 isotope generator.
  • Another purpose of the D invention is to provide a purification system for bacterial membrane vesicles.
  • a purification system for bacterial membrane vesicles is sequentially provided with a fermentation unit 1, an irradiation unit 2 and a separation device 6.
  • the fermentation unit 1 is composed of a bacterial fermentation tank 3 and an ultraviolet spectrophotometer 4,
  • the irradiation unit 2 is composed of an irradiation device 5.
  • the separation device 6 includes a centrifugal unit 7, a membrane package unit and/or a column passing unit.
  • the fermentation tank 3 is a ventilated fermentation tank, a bubbling fermentation tank, an air-lift fermentation tank, or a nozzle circulation type fermentation tank.
  • the ray emitter in the irradiation device 5 is an X-ray generator, a ⁇ -ray generator, or a Co 60 isotope generator.
  • the separation device 6 includes a centrifugal unit 7, which is a normal centrifuge, a high-speed centrifuge, or an ultra-high-speed centrifuge.
  • Another object of the D invention is to provide a bacterial membrane vesicle separation system.
  • a separation system for bacterial membrane vesicles The separation system is provided with a fermentation unit 1 and a separation device 6 in sequence.
  • the fermentation unit 1 is composed of a bacterial fermentation tank 3 and an ultraviolet spectrophotometer 4.
  • the separation device 6 includes a centrifugal unit 7, a membrane package unit and/or a column passing unit.
  • the fermentation tank 3 is a ventilated fermentation tank, a bubbling fermentation tank, an air-lift fermentation tank, or a nozzle circulation type fermentation tank.
  • the separation device 6 includes a centrifugal unit 7, which is a normal centrifuge, a high-speed centrifuge, or an ultra-high-speed centrifuge.
  • the production system of the D embodiment is provided with a fermentation unit 1 and an irradiation unit 2 in sequence.
  • the fermentation unit 1 is composed of a bacterial fermentation tank 3 and an ultraviolet spectrophotometer 4, and the irradiation unit 2 is an irradiation device 5.
  • the fermentation tank 3 is a ventilated fermentation tank, a bubbling fermentation tank, an airlift fermentation tank, or a nozzle circulation type fermentation tank.
  • the ray emitter in the irradiation device 5 is an X-ray generator, a ⁇ -ray generator, or a Co 60 isotope generator.
  • Example D the gram-positive bacteria and gram-negative bacteria were first fermented in the fermentor 3, and then the violet spectrophotometer 4 was used to precisely control the bacterial content of the fermentation broth within a certain limit, and then use the radiation
  • the irradiating device 5 irradiates the fermented bacterial liquid with radiation, thereby promoting the bacteria to produce MVs in large quantities and increasing the MVs content.
  • the purification system of the D embodiment is sequentially provided with a fermentation unit 1, an irradiation unit 2 and a separation device 6.
  • the fermentation unit 1 consists of a bacterial fermentation tank 3 and an ultraviolet spectrophotometer 4, and the irradiation unit 2 consists of
  • the irradiation equipment 5 is constituted, and the separation equipment 6 is constituted by a centrifugal unit 7, a membrane package unit and/or a column passing unit.
  • Example D the gram-positive bacteria and gram-negative bacteria are first fermented in the fermentor 3, and then the bacterial content of the fermentation broth is accurately controlled within a certain limit by the ultraviolet spectrophotometer 4, and then the radiation
  • the irradiation equipment 5 irradiates the fermented bacterial liquid with radiation, and finally uses the centrifugal unit 7 to further centrifuge the irradiated bacterial liquid to remove fermentation waste liquid, bacterial flagella, bacterial secretions, etc., and finally settle
  • the membrane vesicles obtained are the purified membrane vesicles.
  • the radiation generator of the radiation irradiation equipment is an X-ray generator, a ⁇ -ray generator, or a Co 60 isotope generator.
  • the fermentation tank 3 is a ventilated fermentation tank, a bubbling fermentation tank, an air-lift fermentation tank, or a nozzle circulation type fermentation tank.
  • the centrifugal unit 7 includes a centrifuge, a high-speed centrifuge, or an ultra-high-speed centrifuge.
  • the separation system of the D embodiment is provided with a fermentation unit 1 and a centrifugal unit 6 in sequence.
  • the fermentation unit 1 is composed of a bacterial fermentation tank 3 and an ultraviolet spectrophotometer 4, and the separation device 6 is composed of a centrifugal unit 7.
  • Example D Gram-positive bacteria and Gram-negative bacteria are fermented in the fermentor 3 first, and then the fermentation waste liquid, bacterial flagella, bacterial secretions and other wastes are sequentially removed by the centrifuge 7.
  • the fermentation tank 3 is a ventilated fermentation tank, a bubbling fermentation tank, an air-lift fermentation tank, or a nozzle circulation type fermentation tank.
  • the centrifugal unit 7 includes a centrifuge, a high-speed centrifuge, or an ultra-high-speed centrifuge.
  • E Invention A production system, separation and purification system and method for bacterial membrane vesicles
  • the E invention relates to the field of biotechnology, and has a system and method for producing bacterial membrane vesicles, and a system and method for separating and purifying bacterial membrane vesicles.
  • the present invention Based on the problems existing in the production, preparation and purification of bacterial membrane vesicles by modern technology, the present invention provides a set of systems and methods for increasing the production of bacterial MVs, and a set of systems and methods for purifying bacterial MVs.
  • the process technology adopted by the present invention does not add chemical stimulating substances, so there is no adverse effect.
  • the process flow is simple, the vesicle yield is high, the efficiency is high, and the amplification effect is good, and it can be used for mass preparation of vesicles.
  • the purpose of the present invention is to provide a production system for bacterial membrane vesicles.
  • a production system for bacterial membrane vesicles is sequentially provided with a fermentation unit 1 and an irradiation unit 2.
  • the fermentation unit 1 is composed of a bacterial fermentation tank 3 and an ultraviolet spectrophotometer 4, and the irradiation unit 2 Consists of irradiation equipment 5.
  • the fermentation tank 3 is a ventilated fermentation tank, a bubbling fermentation tank, an air-lift fermentation tank, or a nozzle circulation type fermentation tank.
  • the ray emitter in the irradiation device 5 is an X-ray generator, a ⁇ -ray generator, or a Co 60 isotope generator.
  • the purpose of the E invention is also to provide a method for producing bacterial membrane vesicles.
  • the method for producing bacterial membrane vesicles using the above-mentioned production system includes the following steps:
  • the bacteria of the present invention include Gram-positive bacteria and Gram-negative bacteria.
  • the bacteria specifically include the following types:
  • the OD 600 value of the bacteria in the logarithmic growth phase described in step 1) is 0.3-0.8; the ratio of the amount of phosphate buffer or sterile saline added in step 2) to the total amount of bacteria is per 1 ml The amount of bacteria contained in the solution is OD 600 value 20-80.
  • the OD 600 value of the bacteria in the logarithmic growth phase in step 1) is 0.5-0.8; in step 2) the ratio of the amount of phosphate buffer or sterile saline to the total amount of bacteria is The OD 600 value of bacteria in 1ml solution is 40-60.
  • the radiation used for the irradiation treatment in step 3) is X-ray rays; the radiation dose range is 500-3000 Gy.
  • the radiation dose specifically includes: 500-600Gy, 600-700Gy, 700-800Gy, 800-900Gy, 900-1000Gy, 1000-1100Gy, 1100-1200Gy, 1200-1300Gy, 1300-1400Gy, 1400-1500Gy, 1500- 1600Gy, 1600-1700Gy, 1700-1800Gy, 1800-1900Gy, 1900-2000Gy, 2100-2200Gy, 2200-2300Gy, 2300-2400Gy, 2400-2500Gy, 2500-2600Gy, 2600-2700Gy, 2700-2800Gy, 2800-2900Gy, 2900-3000Gy.
  • the purpose of the E invention is also to provide a purification system for bacterial membrane vesicles.
  • a purification system for bacterial membrane vesicles is sequentially provided with a fermentation unit 1, an irradiation unit 2 and a centrifugal unit 6.
  • the fermentation unit 1 is composed of a bacterial fermentation tank 3 and an ultraviolet spectrophotometer 4,
  • the irradiation unit 2 is constituted by an irradiation device 5, and the centrifugal device 6 is constituted by a centrifugal unit 7.
  • the fermentation tank 3 is a ventilated fermentation tank, a bubbling fermentation tank, an air-lift fermentation tank, or a nozzle circulation type fermentation tank.
  • the ray emitter in the irradiation device 5 is an X-ray generator, a ⁇ -ray generator, or a Co 60 isotope generator.
  • the centrifugal unit 7 is an ordinary centrifuge, a high-speed centrifuge, or an ultra-high-speed centrifuge.
  • the purpose of the E invention is also to provide a method for purifying bacterial membrane vesicles.
  • the OD 600 value of the bacteria in the logarithmic growth phase described in step 1) is 0.3-0.8; the ratio of the amount of phosphate buffer or sterile saline added in step 2) to the total amount of bacteria is per 1 ml The amount of bacteria contained in the solution is OD 600 value 20-80.
  • the OD 600 value of the bacteria in the logarithmic growth phase in step 1) is 0.5-0.8; in step 2) the ratio of the amount of phosphate buffer or sterile saline to the total amount of bacteria is The OD 600 value of bacteria in 1ml solution is 40-60.
  • the radiation used for the irradiation treatment in step 3) is X-ray rays; the radiation dose range is 500-3000 Gy.
  • the radiation dose specifically includes: 500-600Gy, 600-700Gy, 700-800Gy, 800-900Gy, 900-1000Gy, 1000-1100Gy, 1100-1200Gy, 1200-1300Gy, 1300-1400Gy, 1400-1500Gy, 1500- 1600Gy, 1600-1700Gy, 1700-1800Gy, 1800-1900Gy, 1900-2000Gy, 2100-2200Gy, 2200-2300Gy, 2300-2400Gy, 2400-2500Gy, 2500-2600Gy, 2600-2700Gy, 2700-2800Gy, 2800-2900Gy, 2900-3000Gy.
  • step 3 the rate of centrifugation of the bacterial liquid after irradiation is 100-10000 g; the centrifugation time is 10-60 min.
  • the speed of centrifugation in step 3) is 400-8000 g; the centrifugation time is 10-30 min.
  • step 4 the speed of high-speed centrifugation is 5000-25000g; the centrifugation time is 10-100min.
  • the high-speed centrifugation rate in step 4) is 10000-20000 g; the centrifugation time is 30-60 min.
  • the ultra-high-speed centrifugation rate is 5000-150000g; the centrifugation time is 60-600min.
  • the ultra-high-speed centrifugation rate in step 5 is 15000-150000g; the centrifugation time is 60-180min.
  • the production system of the E embodiment is provided with a fermentation unit 1 and an irradiation unit 2 in sequence.
  • the fermentation unit 1 is composed of a bacterial fermentation tank 3 and an ultraviolet spectrophotometer 4, and the irradiation unit 2 is an irradiation device 5.
  • the fermentation tank 3 is a ventilated fermentation tank, a bubbling fermentation tank, an airlift fermentation tank, or a nozzle circulation type fermentation tank.
  • the ray emitter in the irradiation device 5 is an X-ray generator, a ⁇ -ray generator, or a Co 60 isotope generator.
  • Example E the gram-positive bacteria and gram-negative bacteria are first fermented in the fermentor 3, and then the violet spectrophotometer 4 is used to precisely control the bacterial content of the fermentation broth within a certain limit, and then use the radiation
  • the irradiating device 5 irradiates the fermented bacterial liquid with radiation to promote the bacteria to produce MVs in large quantities.
  • the method for producing bacterial membrane vesicles using the above production system includes the following steps:
  • the irradiation dose range is 500-3000Gy
  • the specific selection limits include 500-600Gy, 600-700Gy, 700-800Gy, 800-900Gy, 900-1000Gy, 1000-1100Gy , 1100-1200Gy, 1200-1300Gy, 1300-1400Gy, 1400-1500Gy, 1500-1600Gy, 1600-1700Gy, 1700-1800Gy, 1800-1900Gy, 1900-2000Gy, 2100-2200Gy, 2200-2300Gy, 2300-2400Gy, 2400 -2500Gy, 2500-2600Gy, 2600-2700Gy, 2700-2800Gy, 2800-2900Gy, 2900-3000Gy.
  • the purification system of the E embodiment is sequentially provided with a fermentation unit 1, an irradiation unit 2 and a centrifugal unit 6,
  • the fermentation unit 1 is composed of a bacterial fermentation tank 3 and an ultraviolet spectrophotometer 4, and the irradiation unit 2 consists of
  • the irradiation device 5 is constituted, and the centrifugal unit 6 is constituted by a centrifuge 7.
  • Example E the gram-positive bacteria and gram-negative bacteria are first fermented in the fermentor 3, and then the bacteria content of the fermentation broth is accurately controlled within a certain limit by the ultraviolet spectrophotometer 4, and then the radiation
  • the irradiation equipment 5 performs radiation treatment on the fermented bacterial liquid, and finally uses the centrifugal equipment 6 to further centrifuge the irradiated bacterial liquid to sequentially remove the fermentation waste liquid, bacterial flagella, bacterial secretions, etc., and finally settle
  • the membrane vesicles obtained are the purified membrane vesicles.
  • the radiation generator of the radiation irradiation equipment is an X-ray generator, a ⁇ -ray generator, or a Co 60 isotope generator.
  • the fermentation tank 3 is a ventilated fermentation tank, a bubbling fermentation tank, an air-lift type fermentation tank, or a nozzle circulation type fermentation tank.
  • the centrifugal unit 7 includes a centrifuge, a high-speed centrifuge, or an ultra-high-speed centrifuge.
  • the method for purifying bacterial membrane vesicles using the above purification system includes the following steps:

Abstract

属于微生物学领域,具体涉及一种从细菌中分离的膜囊泡(MVs)以及所述膜囊泡的分离和制备系统及方法,以及所述膜囊泡的应用。所述的细菌包括革兰氏阳性菌和革兰氏阴性菌。采用电离射线辐照细菌,分离并纯化其产生的膜囊泡,制得的膜囊泡可作为疫苗、疫苗佐剂、和/或药物载体。还提供一种生物组合物,包括所述膜囊泡和灭活细菌。还提供一种细菌膜囊泡的制备及分离纯化系统以及相应的方法,利用该系统及方法制得的膜囊泡产量大、纯度高、易于工业化生产。

Description

细菌膜囊泡及其分离制备系统和方法
本申请要求2019年8月22日提交的中国发明专利1)201910777479.2“细菌膜囊泡及其制备方法与应用”;2)201910777473.5“金黄色葡萄球菌膜囊泡及其制备方法与应用”;3)201910777606.9“铜绿假单胞菌膜囊泡及其制备方法与应用”;4)201921369450.2“一种细菌膜囊泡的生产系统和分离纯化系统”;5)201910777595.4“一种细菌膜囊泡的生产系统和分离纯化系统及方法”的优先权,这五件优先权专利申请以引用方式全文并入。
技术领域
本发明属于微生物学领域,具体涉及一种细菌膜囊泡以及这种膜囊泡的分离和制备系统及方法,以及这种膜囊泡的应用。
背景技术
膜囊泡(MVs,membrane vesicles)是细菌细胞(包括革兰氏阳性菌和阴性菌)外膜产生的囊泡状结构产物。多数为球形,直径大约20–250nm(Structures of gram-negative cell walls and their derived membrane vesicles(革兰氏阴性菌细胞壁结构及其产生的膜囊泡),Beveridge T,Journal of Bacteriology,1999vol:181(16)pp:4725-33)。细菌膜囊泡含有多种生物活性大分子如核酸、脂多糖、外膜蛋白等成分,以及金属离子、酶、信号分子等(Biological functions and biogenesis of secreted bacterial outer membrane vesicles(经分泌产生的细菌外膜囊泡的生物功能与起源),Kulp A,Kuehn M,Annual Review of Microbiology,2010vol:64(1)pp:163-184)。它在细菌的多种生命活动中扮演着重要角色,如产生毒力因子、压力应激反应、营养摄取,及作为细菌间、细菌与宿主细胞间进行信息交流的载体等。
膜囊泡的产生发生在细菌的任何生长阶段,不同于细胞裂解及凋亡,研究发现在压力刺激、缺氧、抗生素压迫等情况能够促进细菌产生膜囊泡。然而,自然生成的细菌膜囊泡产量低,往往需要培养大量的菌体才能收获一定量的膜囊泡,并且后续还需要额外的纯化工艺才能得到一定质量的膜囊泡。
现代工艺技术在生产制备及纯化细菌膜囊泡时存在以下问题:1)利用抗生素、去污剂、氧化剂等手段虽然能促进膜囊泡产生,但也伴随着有毒残留问题,给其应用带来了不确定性。2)上述干预因 素刺激细菌产生膜囊泡的效率也相对较低,且该制备工艺不能实现标准化生产。3)上述方法会使菌体本身的外膜抗原性、构象等可能发生变化,进而影响囊泡,限制其后续应用。
发明内容
针对以上技术问题,本发明提供一种细菌膜囊泡以及分离生产这种细菌膜囊泡的系统和方法,包含五个子发明(即A发明、B发明、C发明、D发明和E发明)以及基于这五个子发明的各自拓展。
本发明首先公开了一种从灭活细菌中分离的膜囊泡。所述灭活细菌包括革兰氏阳性菌和革兰氏阴性菌,以及基因工程菌。本发明还提供了这种膜囊泡的分离和制备方法,以及作为疫苗的应用。本发明首次采用电离射线X射线辐照细菌,分离并纯化其产生的MVs,制得的MVs可作为疫苗、疫苗佐剂、和/或药物载体。
在另一方面,本发明还公开了一种本发明提供了一种从灭活铜绿假单胞菌中分离的膜囊泡。本发明还提供了这种膜囊泡的分离和制备方法,以及作为疫苗的应用。以及该细菌疫苗的应用。本发明首次采用电离射线X射线辐照铜绿假单胞菌,分离并纯化其产生的MVs,制得的MVs可作为疫苗、疫苗佐剂、和/或药物载体。
在另一方面,本发明还公开了一种从灭活金黄色葡萄球菌中分离的膜囊泡。本发明还提供了这种膜囊泡的分离和制备方法,以及作为疫苗的应用。以及该细菌疫苗的应用。本发明首次采用电离射线X射线辐照金黄色葡萄球菌,分离并纯化其产生的MVs,制得的MVs可作为疫苗、疫苗佐剂、和/或药物载体。
在另一方面,本发明还公开了一种用于细菌膜囊泡的生产系统,其依次设有发酵罐、紫外分光光度设备和辐照设备,细菌通过在发酵罐中发酵增殖,然后通过紫外分光光度设备控制菌液中的含菌量在一定限度内,再采用辐照设备对菌液进行射线辐照,促进细菌大量产生膜囊泡。相应地,本发明还提供了一种细菌膜囊泡的纯化系统和一种细菌膜囊泡的分离系统,利用该系统制得的膜囊泡产量大、纯度高、易于工业化生产。
在另一方面,本发明还公开了一种细菌膜囊泡的生产系统及方法,该生产系统依次设有发酵罐、紫外分光光度设备和辐照设备,细菌通过在发酵罐中发酵增殖,然后通过紫外分光光度设备控制菌液中的含菌量在一定限度内,再采用辐照设备对菌液进行射线辐照,促进细菌大量产生膜囊泡,从而提高膜囊泡的含量。相应地,本发明还提供了一种细菌膜囊泡的分离纯化系统及纯化方法,利用该方法制得的膜囊泡产量大、纯度高、易于工业化生产。
本发明有益效果
本发明首次采用电离射线X射线辐照细菌分离并纯化其产生的MVs。不添加抗生素及其他化学刺激物质,因此避免了刺激物残留及刺激物本身对膜囊泡导致的不良作用。同时工艺流程简单、适合工业放大化、标准化生产;囊泡产量高,高效、放大效果及纯化效果好,可以用于囊泡的大量制备。制备的囊泡与正常囊泡相比,产量提高数十倍具有较好的免疫原性。优化生产后得到的细菌膜囊泡后续进一步的开发应用前景广阔。此外,本发明制备的细菌膜囊泡可直接作为疫苗使用。
本发明提供的细菌膜囊泡的生产系统能促进促进细菌大量产生的膜囊泡,适用于工业化生产;本发明提供的细菌膜囊泡的纯化及分离系统能除去发酵废液、细菌鞭毛、细菌分泌物等,得到的膜囊泡纯度高。
附图说明
为了更清楚地说明本发明实施例或现有技术中的技术方案,下面将对实施例或现有技术描述中所需要使用的附图作一简单地介绍。显而易见地,下面描述中的附图是本发明的一些实施例,对于本领域普通技术人员来讲,在不付出创造性劳动性的前提下,还可以根据这些附图获得其它的附图。
图1铜绿假单胞菌膜囊泡透射电镜图(A:对照组膜囊泡;B:辐照组膜囊泡;比例尺:200nm)。
图2铜绿假单胞菌膜囊泡内容物含量测定图。
图3铜绿假单胞菌膜囊泡粒径分布图。
图4金黄色葡萄球菌膜囊泡内容物含量测定图。
图5辐照处理的膜囊泡促进骨髓来源树突状细胞细表面分子CD80、CD86及MHCII分子显著上调。
图6辐照处理的膜囊泡刺激后的DC细胞对抗原吞噬能力的柱状图。
图7 CD4+T细胞在与不同处理方式后的DC发生作用后的增殖百分比。
.图8 CD4+T细胞在与不同处理方式后的DC发生作用后的增殖流式图。
图9辐照处理的膜囊泡增强DC细胞与T细胞相互作用(GC:growth control,树突状细胞生长对照组(未刺激组);Cell+MVs(全菌体+囊泡处理组);MVs(囊泡处理组))。
图10细菌膜囊泡生产系统结构示意图。
图11细菌膜囊泡纯化系统结构示意图。
图12细菌膜囊泡分离系统结构示意图。
具体实施方式
为使本发明实施例的目的、技术方案和优点更加清楚,下面将结合本发明实施例中的附图,对本发明实施例中的技术方案进行清楚、完整地描述。显然,所描述的实施例是本发明一部分实施例,而不是全部的实施例。基于本发明中的实施例,本领域普通技术人员在没有作出创造性劳动前提下所获得的所有其他实施例,都属于本发明保护的范围。
所举实施例是为了更好地对本发明进行说明,但并不是本发明的内容仅局限于所举实施例。所以熟悉本领域的技术人员根据上述发明内容对实施方案进行非本质的改进和调整,仍属于本发明的保护范围。
需要注意的是,本发明中的各个子发明(A发明、B发明、C发明、D发明和E发明)是可以组合在一起衍生出其它的技术方案、解决更多的技术问题、产生更多的技术效果的,因此本文中针对各个子发明的描述是可以互相印证、解释、说明、结合、组合、融合的。
但是,针对本文中各个子发明的描述可能会涉及到一些相同或相似的用词,如果这些相同或相似的用词在理解和含义上产生冲突或者矛盾,那么以这些用词在各自所属于的子发明的描述部分的理解和含义为准。
A发明:细菌膜囊泡及其制备方法与应用
A发明属于微生物学领域,具体涉及一种细菌膜囊泡的制备分离纯化以及该膜囊泡的应用。
现代工艺技术在生产制备及纯化细菌膜囊泡时存在以下问题:1)利用抗生素、去污剂、氧化剂等手段虽然能促进膜囊泡产生,但也伴随着有毒残留问题,给其应用带来了不确定性。2)上述干预因素刺激细菌产生膜囊泡的效率也相对较低,且该制备工艺不能实现标准化生产。3)上述方法会使菌体本身的外膜抗原性、构象等可能发生变化,进而影响囊泡,限制其后续应用。
鉴于此,A发明提供了一种分离细菌MVs的方法,和一种制备细菌MVs的方法。本发明不添加化学刺激物质,因此无残留物引起不良作用,同时工艺流程简单、囊泡产量高,高效、放大效果好,可以用于囊泡的大量制备。制备的囊泡与正常囊泡相比具有较好的免疫原性,后续进一步的开发应用前景广阔。
A发明的目的在于提供一种细菌膜囊泡。
细菌产生的生物颗粒,所述生物颗粒是从灭活细菌中分离的膜 囊泡。
进一步,所述灭活细菌包括革兰氏阳性菌、革兰氏阴性菌和基因工程改造菌。
进一步,所述革兰氏阳性菌包括金黄色葡萄球菌、链球菌、白喉杆菌、结核杆菌、破伤风杆菌、芽孢杆菌、棒状杆菌,以及上述菌的基因工程改造菌。
进一步,所述革兰氏阴性菌包括铜绿假单胞菌、鲍曼不动杆菌、大肠杆菌、伤寒杆菌、脑膜炎双球菌、变形杆菌、志贺菌,以及上述菌的基因工程改造菌。
进一步,所述灭活菌为厌氧菌。
进一步,所述厌氧菌包括幽门螺杆菌、反硝化细菌或核梭杆菌。
一种生物组合物,其特征在于,含有上述膜囊泡和上述灭活细菌。
进一步,上述膜囊泡作为疫苗和/或疫苗佐剂和/或药物载体与上述灭活细菌组合。
A发明的目的还在于提供一种膜囊泡的分离方法。
一种从细菌中分离膜囊泡的方法,所述细菌包括革兰氏阳性菌和革兰氏阴性菌,具体包括以下步骤:
1)将培养细菌的菌液中的菌体和培养液进行分离,收集获得上清液1,
2)将所述上清液1用高速离心机离心,收集上清液2;
3)将所述上清液2用超高速离心机离心,沉淀为膜囊泡。
进一步,所述步骤1)中分离方法包括离心、过柱或膜包浓缩。
133.进一步,在所述步骤2)中收集获得的所述上清液2在进入所述步骤3)之前,先经膜包浓缩。在一些实施例中,所选择的膜包可以浓缩大于100KD的物质。
进一步,将所述膜囊泡用缓冲液重悬,所述缓冲液按1L体积计算,包括50mM Tris,5mM NaCl,1mM MgSO 4,并且pH值为7.4。
一种制备生物组合物的方法,收集上述分离膜囊泡方法中步骤1)分离的所述菌体,将所述菌体与步骤3)所得的所述膜囊泡混合形成所述生物组合物。
进一步,所述步骤1)中,将所述上清液1用0.3-0.5μM滤器过滤去杂质。
优选的,将所述上清液1用045μM滤器过滤去杂质。
进一步,所述步骤1)中的分离方法为离心,所述离心的离心速率为100-10000g;离心时间为10-60min。
优选的,步骤1)的离心的速率为400-8000g;离心时间为10-30min。
进一步,所述步骤2)中的高速离心速率为5000-25000g;离心时间为10-100min。
优选的,步骤2)中的高速离心速率为10000-20000g;离心时间为30-60min。
进一步,所述步骤3)中的所述超高速离心速率为5000-150000g;离心时间为60-600min。
优选的,步骤3)中的超高速离心速率为15000-150000g;离心时间为60-180min。
进一步,所述革兰氏阳性菌包括金黄色葡萄球菌、链球菌、白喉杆菌、结核杆菌、破伤风杆菌、芽孢杆菌、棒状杆菌,以及上述菌的基因工程改造菌。
进一步,所述革兰氏阴性菌包括铜绿假单胞菌、鲍曼不动杆菌、大肠杆菌、伤寒杆菌、脑膜炎双球菌、变形杆菌、志贺菌,以及上述菌的基因工程改造菌。
进一步,所述细菌为灭活细菌。
采用上述方法所得的膜囊泡。
本发明的目的还在于提供一种制备方法。
一种制备生物颗粒的方法,其特征在于,具体包括以下步骤:
1)膜囊泡增量
培养细菌至对数生长期;收集菌体,将所述菌体重悬后用电离射线辐照处理,得辐照细菌;
2)分离纯化
用上述分离膜囊泡的方法进行分离,得膜囊泡。
进一步,所述电离射线辐照处理的方式为X-ray射线,辐照剂量为500-3000Gy。所述辐照剂量具体包括:500-600Gy,600-700Gy,700-800Gy,800-900Gy,900-1000Gy,1000-1100Gy,1100-1200Gy,1200-1300Gy,1300-1400Gy,1400-1500Gy,1500-1600Gy,1600-1700Gy,1700-1800Gy,1800-1900Gy,1900-2000Gy,2100-2200Gy,2200-2300Gy,2300-2400Gy,2400-2500Gy,2500-2600Gy,2600-2700Gy,2700-2800Gy,2800-2900Gy,2900-3000Gy。
优选的,所述辐照剂量为500-1000Gy。所述辐照剂量具体包括:500-600Gy,600-700Gy,700-800Gy,800-900Gy,900-1000Gy。
进一步,所述步骤1)中所述对数生长期的细菌的OD 600值为0.3-0.8。
优选的,所述步骤1)中所述对数生长期的细菌的OD 600值为0.5-0.7。
进一步,所述步骤1)中用磷酸盐缓冲液或无菌生理盐水重悬所述菌体。
优选的,所述步骤1)中用磷酸盐缓冲液重悬所述菌体。
进一步,所述步骤1)中将所述菌体重悬至OD 600值为20-80。
优选的,所述步骤1)中将所述菌体重悬至OD 600值为40-60。
采用上述方法所得的膜囊泡。
上述方法制得的膜囊泡,同未经电离射线辐照的细菌相比,所述膜囊泡中核酸的含量和蛋白质的含量提升了10-20倍。
本发明的目的还在于提供膜囊泡及灭活菌的应用。
上述膜囊泡(以下简称A发明制得的膜囊泡)作为免疫原中的应用。
A发明制得的膜囊泡的膜囊泡作为免疫反应促进剂中的应用。
A发明制得的膜囊泡的膜囊泡作为治疗细菌感染疾病的疫苗中的应用。
进一步,所述细菌感染疾病包括肺炎、泌尿道感染、脑膜炎、败血症,或皮肤、软组织感染。
A发明制得的膜囊泡作为疫苗佐剂中的应用。
进一步,所述疫苗佐剂为非特异性地改变或增强机体对抗原的特异性免疫应答。
A发明制得的膜囊泡作为抗原递呈细胞功能促进剂中的应用。
进一步,抗原递呈细胞树突状细胞,巨噬细胞,及B细胞。
经过辐射再分离纯化得到的膜囊泡作为DC细胞成熟的促进剂中的应用。
进一步,所述的膜囊泡作为促进骨髓来源树突状细胞细表面分子CD80、CD86及MHCII分子显著上调的促进剂中的应用。
A发明制得的膜囊泡作为DC细胞抗原递呈能力促进剂中的应用。
A发明制得的膜囊泡联合DC细胞在制备CD4+T细胞的增殖剂中的应用。
促进CD4+T细胞增殖的方法,将辐照处理制备的膜囊泡刺激并吞噬OVA抗原的DCs与CFSE标记的CD4+T淋巴细胞在体外共培养。
A发明制得的膜囊泡在制备兽药中的应用。
A发明所述的灭活细菌作为细菌疫苗的应用。
A发明的实施例一
A发明提供了一种分离制备细菌膜囊泡的方法,所述方法包括如下步骤:
1.培养细菌至对数生长期,对数生长期的细菌的OD 600值为0.3-0.8;最好是选择OD 600值为0.5-0.8(此处还可以进行发酵,使菌体进一步富集);收集菌体,将所述菌体用适量的磷酸盐缓冲液重悬, 所加磷酸盐缓冲液的量与菌体总量的比例为每1ml溶液含有的菌量为OD 600值20-80;最好是选择OD 600值为40-60;重悬后用电离射线辐照处理,得辐照细菌;最好用X-ray射线辐照处理,辐照剂量为500-3000Gy,具体包括:500-600Gy,600-700Gy,700-800Gy,800-900Gy,900-1000Gy,1000-1100Gy,1100-1200Gy,1200-1300Gy,1300-1400Gy,1400-1500Gy,1500-1600Gy,1600-1700Gy,1700-1800Gy,1800-1900Gy,1900-2000Gy,2100-2200Gy,2200-2300Gy,2300-2400Gy,2400-2500Gy,2500-2600Gy,2600-2700Gy,2700-2800Gy,2800-2900Gy,2900-3000Gy。
2.收集菌液,菌液离心并收集上清液,上清液用03-0.5μM滤器过滤除菌;离心速率为400-8000g;离心时间为10-30min。
3.将过滤后的上清液用高速离心机离心,收集上清液,去除鞭毛;高速离心速率为10000-20000g;离心时间为30-60min。
4.将去除鞭毛后的上清液用超高速离心机离心,沉淀膜囊泡;超高速离心速率为15000-150000g;离心时间为60-180min。
5.收集膜囊泡,得纯化后的膜囊泡。
A发明的实施例二
电离射线辐照铜绿假单胞杆菌PAO1制备、分离纯化膜囊泡:
1.从-80℃复苏铜绿假单胞菌PAO1划线至LB平板,37℃培养箱培养16-18h。
2.从LB平板上挑取单克隆菌落接种于20mL LB液体培养基中,37℃,250rpm恒温培养16-18h。
3.取过夜菌液接种至1L LB培养基中,至起始浓度为0.05OD 600/mL,37℃,250rpm培养至对数生长期,测OD 600值。
4.将上述3)的菌液转移至离心桶中,5,000g离心20min,收集菌体用生理盐水重悬,调整菌体浓度为50OD左右。
5.取上述菌液置于辐照仪中,辐照剂量为1000Gy。
6.将辐照后的菌液8,000×g,20min,离心两次,收集上清液;上清液用0.45μM滤器过滤除菌再次收集得到的上清,同时取少量上清液涂在LB平板上,37℃培养24-72h,以确认无活菌存在。
7.将步骤6)上清液高速离心机离心,除去上清液中的鞭毛。
8.将步骤7)上清液超高速离心机离心,沉淀膜囊泡。
9.弃去上清,沉淀用MV buffer重悬,-80℃保存。
10.将提取的正常组膜囊泡和本发明实验组膜囊泡进行透射电镜观察。同时测定提取的正常组膜囊泡和本发明实验组膜囊泡的内容物含量,包括测定其DNA含量、RNA含量和蛋白质含量。最后再测量提取的正常组膜囊泡和本发明实验组膜囊泡粒径大小。
实验结果:
由电镜结果可知,电离射线能够刺激绿脓杆菌PAO1产生膜囊泡,实验组膜囊泡数量更多;囊泡形态大小与正常对照组无明显差异。见图1。
内容物含量测定结果:实验组制备的膜囊泡的核酸含量和蛋白质含量与正常对照组相比提高10-20倍。见图2。具体测量数据见表1。
表1 膜囊泡内容物含量测定
辐照剂量Gy DNA ng/μL RNA ng/μL 蛋白μg/mL 内毒素(EU/ml)
- 24.8 19.7 262.7 1.28×10 5
980 469.0 364.0 4551.0 1.07×10 6
粒径分布图数据显示,正常对照组和实验组制备的膜囊泡大小上无明显差别,平均在150nm左右。见图3;具体测量数据见表2。
表2 粒径
辐照剂量Gy 平均粒径(nm) Peak(nm)
- 152.2 192.6
980 146.5 222.9
A发明的实施例三
电离射线辐照金黄色葡萄球菌ATCC 25923制备膜囊泡:
1.从-80℃复苏金黄色葡萄球菌ATCC 25923划线至LB平板,37℃培养箱培养16-18h。
2.从LB平板上挑取单克隆菌落接种于100mL LB液体培养基中,37℃,250rpm恒温培养16-18h。
3.取过夜菌液接种至2L LB培养基中,至起始浓度为0.05OD 600/mL,37℃,250rpm培养至对数生长期,测OD 600值。
4.将上述3)菌液转移至2L离心桶中,5,000g离心20min,收集菌体用生理盐水重悬,调整菌体浓度为50OD左右,取100μL稀释10 7涂板计算活菌数。
5.取上述菌液置于辐照仪中,辐照剂量为1000Gy。
6.将辐照后的菌液8,000×g,20min,离心两次,收集上清液;上清液用0.45μM滤器过滤除菌再次收集得到的上清,同时取少量上清液涂在LB平板上,37℃培养24-72h,以确认无活菌存在。
7.将步骤6)上清液超高速离心机离心,39,000×g,90min,沉淀MVs。
8.弃去上清,沉淀用2mL MV buffer重悬,-80℃保存。
9.测定提取的正常组膜囊泡和本发明实验组膜囊泡的内 容物含量,包括测定其DNA含量、RNA含量和蛋白质含量。
实验结果
经测定结果表明,对于金黄色葡萄球菌ATCC27853,实验组制备的膜囊泡的核酸含量和蛋白质含量明显提高,表明,电离射线辐照革兰氏阳性菌也能刺激膜囊泡的产生。见图4。
A发明实施例四
辐照处理的细菌膜囊泡的免疫调节作用-促进树突状细胞成熟:
树突状细胞(Dentitic Cells,DCs)是机体的主要的抗原递呈细胞,其主要功能是吞噬、处理加工抗原分子,并递呈给T细胞。是已知体内功能最强、唯一能活化静息T细胞的专职抗原提呈细胞,是启动、调控和维持免疫应答的中心环节。树突状细胞的成熟状况决定机体产生免疫应答或免疫耐受。DCs表面的共刺激分子B7(B7-1=CD80和B7-2=CD86)可以和T细胞表面的CD28或CD152分子结合,加强或者减弱DCs与T细胞之间MHC-TCR的信号传导。在成熟的DCs上主要表现为表明共刺激分子CD80、CD86表达变化,吞噬抗原能力减弱而处理递呈抗原能力增强(MHCII分子表达增高),以及与T淋巴细胞的相互作用等。
1.小鼠骨髓来源树突状细胞(BMDC)细胞诱导培养:取6-8周C57雌性小鼠,无菌分离小鼠股骨,将股骨上的肌肉剔干净后剪开股骨两端,PBS冲洗骨管腔直至发白,将PBS悬液过滤后1200rpm离5min,去上清,加入5ml红细胞裂解液重悬细胞。静置15min后1200rpm离心5min,去上清,加入50ml 1640完全培养基(20ng/ml GM-CSF、10%FBS、50mM 2-巯基乙醇)重悬细胞。混匀后分至5个培养皿中置培养箱中培养,每2天换液,第7天收集细胞。
2.BMDC刺激:取诱导7天的BMDC细胞,6孔板中反复吹打使贴壁细胞脱落,收集细胞悬液,1100rpm离心5min,去上清,加入1ml培养基重悬细胞、活细胞计数,调整细胞浓度至1×10 6/ml,接种2ml至新的6孔板。加入各刺激物并混匀,分别为:全菌,全菌+囊泡,囊泡,使得终浓度为15μg/mL(以蛋白质为标准),继续培养24小时,生长对照组加入等体积的PBS。
3.流式检测成熟marker:24h后取出6孔板,反复吹打细胞使其脱落,收集细胞悬液至流式管,1500rpm离心3min,去上清,加入1ml PBS继续1500rpm离心3min后去上清,反复清洗3遍。加入CD11c/CD80/CD86/MHCII抗体室温避光孵育30min,同时阴性对照组应设立一个同型对照组加入CD11c/CD80/CD86/MHCII的同行对照。孵育完成后加入PBS清洗2遍后加入200μl PBS重悬细胞,上流式细胞仪检测。
4.结果处理:流式细胞仪软件分析CD11c细胞中CD80/CD86/MHCII比例。
实验结果:
与全菌体相比,X射线处理的实验组(MVs)囊泡能使刺激后DCs表面共刺激分子CD80、CD86和MHCII等显著上调,而这些表面分子是树突状细胞成熟的标志。综上证明囊泡能显著促进DCs的分化和成熟。见图5。
A发明实施例五
通过检测FITC标记的葡聚糖荧光强度来检测DC细胞的吞噬能力:
DC细胞具有极强的抗原内吞和加工处理能力。在未接触到抗原物质时处于非成熟状态,其吞噬能力强,在接触到抗原激活后变为成熟,其吞噬能力变弱,抗原提呈能力增强,本实验通过检测FITC标记的葡聚糖荧光强度来确定DC的吞噬葡聚糖的多少从而检测DC的吞噬能力是否增强。
1.BMDC细胞诱导培养(同上一实施例)。
2.刺激:第7天收集细胞,将细胞全部吹下后离心重悬计数,然后接种到6孔板,每孔接种1×10 6个细胞,再分别加入刺激物,GC组加入等体积的PBS,Control组及Treatment组加入相同浓度的膜囊泡(按蛋白质水平)后37℃培养24h。
3.吞噬与检测:加入葡聚糖(5μg/ml),继续培养1h后,吸出细胞到流式管,PBS洗涤3次后加入CD11c抗体室温避光孵育30min,PBS清洗3次后流式检测FITC荧光。
4.结果处理:流式细胞仪软件分析CD11c细胞中FITC比例。
实验数据:
为了检测DCs的吞噬功能,我们采用FITC-葡聚糖作为模式抗原供DCs吞噬,检测CD11c+DCs的FITC平均荧光强度值。实验结果显示,GC组树突状细胞(生长对照组)基本没有FITC-葡聚糖摄入,但是无论是DCs接受刺激后,FITC平均荧光强度值相比于GC组明显降低。这个实验结果再次证明囊泡能够促进DCs成熟,从而降低对抗原的摄取能力。见图6。
A发明实施例六
X射线处理组细菌膜囊泡刺激后的成熟DC与T细胞相互作用:
A.成熟DC与CD4+T细胞相互作用:
DCs对胞外蛋白有效的交叉抗原递呈在诱导特异性细胞免疫反应中具有重要作用。因此检测囊泡刺激后DCs对OVA抗原的交 叉递呈作用。在DCs-T细胞共培养后的72h,我们通过CFSE流式细胞术来检测OT-II CD4+T淋巴细胞的增殖情况。荧光染料CFSE(CFDA-SE),即羟基荧光素二醋酸盐琥珀酰亚胺脂,是一种可对活细胞进行荧光标记的细胞染色试剂。它进入细胞后可以不可逆地与细胞内的氨基结合偶联到细胞蛋白质上。在细胞分裂增殖过程中,CFSE标记荧光可平均分配至两个子代细胞中,其荧光强度是亲代细胞的一半。因此我们可以利用流式细胞术对CFSE荧光较弱的细胞百分比进行统计,从而得到增殖细胞的比例。
1.BMDC细胞诱导培养(同上实施例)。
2.抗原吞噬:将培养7天的DCs在含10μg/ml OVA培养基中培养24h作为GC(growth control,生长对照组),MVs组别另外加入囊泡,接下来离心收集吞噬过抗原的DCs并重悬在正常培养基中,以2×10 4细胞/孔的密度铺板于96孔板中,每孔100μl,每组3个复孔。
3.T细胞提取:在第二天,OT-II小鼠脾脏OVA特异性的CD4+T淋巴细胞使用Stem Cell Technologies公司的阴性磁珠筛选试剂盒分离富集。
4.DC与T细胞共培养:对分选出的CD4+T细胞按照试剂盒说明书使用1μM CFSE进行标记。标记后用PBS洗涤3次,以10 5细胞/孔的密度加入到96孔板中,使其培养终体积为200μl(CD4:DC=5:1)。
5.在共培养后的第3天,利用流式细胞术通过CFSE递减来检测CD4+T细胞群的增殖情况。
实验结果:
将囊泡刺激并吞噬OVA抗原的DCs与CFSE标记的OT-II小鼠CD4+T淋巴细胞在体外共培养。流式分析CFSE荧光强度结果表明,增殖CD4+T细胞的比例增加。囊泡(14.05%)能显著增加吞噬OVA抗原的DCs(6.80%)对于特异性CD4+T细胞的促进增殖作用。详见图7、图8。
B.囊泡处理后的DC促进T细胞增殖:
DCs对胞外蛋白有效的交叉抗原递呈在诱导特异性细胞免疫反应中具有重要作用。因此检测囊泡刺激后DCs对OVA抗原的交叉递呈作用。在DCs-T细胞共培养后的72h,我们通过CFSE流式细胞术来检测T淋巴细胞的增殖情况。荧光染料CFSE(CFDA-SE),即羟基荧光素二醋酸盐琥珀酰亚胺脂,是一种可对活细胞进行荧光标记的细胞染色试剂。它进入细胞后可以不可逆地与细胞内的氨基结合偶联到细胞蛋白质上。在细胞分裂增殖过程中,CFSE标记荧光可平均分配至两个子代细胞中,其荧光强度是亲代细胞的一半。因此我们可 以利用流式细胞术对CFSE荧光较弱的细胞百分比进行统计,从而得到增殖细胞的比例。
1.BMDC细胞诱导培养(同上实施例)。
2.抗原吞噬:将培养7天的DCs在培养基中培养24h作为GC(growth control,生长对照组),MVs组别另外加入囊泡,接下来离心收集吞噬过抗原的DCs并重悬在正常培养基中,以4×10 4细胞/孔的密度铺板于96孔板中,每孔100μl,每组3个复孔。
3.T细胞提取:在第二天,使用MVs免疫一次后一周的小鼠脾脏Stem Cell Technologies公司的阴性磁珠筛选试剂盒分离富集小鼠的T细胞。
4.DC与T细胞共培养:对分选出的T细胞按照试剂盒说明书使用1μM CFSE进行标记。标记后用PBS洗涤3次,以4×10 5细胞/孔的密度加入到96孔板中,使其培养终体积为200μl(CD3:DC=10:1)。
5.在共培养后的第3天,利用流式细胞术通过CFSE递减来检测CD3+,CD8+,CD4+T细胞群的增殖情况。
实验结果:
将囊泡刺激并吞噬OVA抗原的DCs与CFSE标记的OT-II小鼠CD4+T淋巴细胞在体外共培养。流式分析CFSE荧光强度结果表明,增殖CD4+T细胞的比例增加。如图所示,菌体加囊泡刺激组荧光强度为63.5%,囊泡刺激组为71%。说明,囊泡处理后DCs,能显著刺激CD4+T细胞的增殖。见图9。
B发明:铜绿假单胞菌膜囊泡及其制备方法与应用
B发明属于微生物学领域,具体涉及一种铜绿假单胞菌膜囊泡的制备分离纯化以及该膜囊泡的应用。
现代工艺技术在生产制备及纯化细菌膜囊泡时存在以下问题:1)利用抗生素、去污剂、氧化剂等手段虽然能促进膜囊泡产生,但也伴随着有毒残留问题,给其应用带来了不确定性。2)上述干预因素刺激细菌产生膜囊泡的效率也相对较低,且该制备工艺不能实现标准化生产。3)上述方法会使菌体本身的外膜抗原性、构象等可能发生变化,进而影响囊泡,限制其后续应用。
B发明提供了一种分离铜绿假单胞菌MVs的方法,和一种制备铜绿假单胞菌MVs的方法。本发明采用国际领先的工艺技术,不添加化学刺激物质,因此无不良作用,同时工艺流程简单、囊泡产量高,高效、放大效果好,可以用于囊泡的大量制备。制备的囊泡与正常囊泡相比具有较好的免疫原性,后续进一步的开发应用前景广阔。
B发明的目的在于提供一种铜绿假单胞菌疫苗。
铜绿假单胞菌疫苗,所述铜绿假单胞菌疫苗含有从灭活铜绿假单胞菌中分离的膜囊泡。
一种生物组合物,含有被分离的膜囊泡和灭活铜绿假单胞菌。
进一步,所述膜囊泡作为疫苗和/或疫苗佐剂和/或药物载体与灭活铜绿假单胞菌组合。
B发明的目的还在于提供一种分离铜绿假单胞菌膜囊泡的方法。
一种从铜绿假单胞菌中分离膜囊泡的方法,包括如下步骤:
1)培养细菌至对数生长期然后发酵;
2)收集菌液,菌液离心并取上清液1,所述上清液1用0.3-0.5μM滤器过滤除菌;
3)将过滤后的所述上清液1用高速离心机离心,收集上清液2,去除鞭毛;
4)将去除鞭毛后的所述上清液2用超高速离心机离心,沉淀即得到膜囊泡。
进一步,步骤2)的上清液用0.45μM滤器过滤除菌。
进一步,步骤2)的离心速率为100-10000g;离心时间为10-60min。
优选的,所述离心速率为400-8000g;离心时间为10-30min。
进一步,步骤3)的高速离心速率为5000-25000g;离心时间为10-100min。
优选的,所述的高速离心速率为10000-20000g;离心时间为30-60min。
进一步,步骤4)的超高速离心速率为5000-150000g;离心时间为60-600min。
优选的,所述的超高速离心速率为15000-150000g;离心时间为60-180min。
进一步,所述铜绿假单胞菌为灭活的铜绿假单胞菌。
采用上述分离方法得到的铜绿假单胞菌膜囊泡。
B发明的目的还在于提供一种制备方法。
制备上述铜绿假单胞菌疫苗的方法,所述方法包括如下步骤:
1)培养细菌至对数生长期(此处也可以进行发酵,使菌体进一步富集);
2)收集菌体,将菌体用适量的磷酸盐缓冲液或无菌生理盐水重悬并用电离射线辐照处理;
3)收集辐照处理后的菌液,离心并取上清液1,所述上清液1用0.3-0.5μM滤器过滤除菌;
4)将过滤后的所述上清液1用高速离心机离心,收集上清液2,去除鞭毛;
5)将去除鞭毛后的所述上清液2超高速离心,沉淀即得膜囊泡。
进一步,步骤1)中所述对数生长期的细菌的OD 600值为0.3-0.8。
进一步,其特征在于,步骤2)中所加磷酸盐缓冲液或无菌生理盐水的量与菌体总量的比例为每1ml溶液含有的菌量为OD 600值20-80。
进一步,步骤3)中辐照处理用的射线为X-ray射线;辐照剂量为500-3000Gy。所述辐照剂量具体包括:500-600Gy,600-700Gy,700-800Gy,800-900Gy,900-1000Gy,1000-1100Gy,1100-1200Gy,1200-1300Gy,1300-1400Gy,1400-1500Gy,1500-1600Gy,1600-1700Gy,1700-1800Gy,1800-1900Gy,1900-2000Gy,2100-2200Gy,2200-2300Gy,2300-2400Gy,2400-2500Gy,2500-2600Gy,2600-2700Gy,2700-2800Gy,2800-2900Gy,2900-3000Gy。
优选的,辐照剂量为500-1000Gy。所述辐照剂量具体包括:500-600Gy,600-700Gy,700-800Gy,800-900Gy,900-1000Gy。
进一步,步骤3)中的离心速率为100-10000g;离心时间为10-60min。
优选的,所述离心速率为400-8000g;离心时间为10-30min。
进一步,步骤3)中上清液用0.45μM滤器过滤除菌。
进一步,步骤4)中所述的高速离心速率为5000-25000g;离心时间为10-100min。
优选的,所述高速离心速率为10000-20000g;离心时间为30-60min。
进一步,步骤5)中所述的超高速离心速率为5000-150000g;离心时间为60-600min。
优选的,所述的超高速离心速率为15000-150000g;离心时间为60-180min。
采用上述方法制得得到的铜绿假单胞菌膜囊泡和疫苗。
进一步,上述铜绿假单胞菌膜囊泡同未经电离射线辐照的细菌膜囊泡相比,所述膜囊泡中核酸的含量和蛋白质的含量提升了10-20倍。
一种提高铜绿假单胞菌膜囊泡内容物含量的方法,所述内容物包括核酸和蛋白质,所述方法采用辐照设备处理铜绿假单胞菌菌液,所述辐照设备的射线为X-ray射线,辐照剂量为500-1000Gy。
B发明的目的还在于提供一种所述铜绿假单胞菌膜囊泡和灭活铜绿假单胞菌的应用。
B发明制得的铜绿假单胞菌膜囊泡(以下均简称B发明制得的铜绿假单胞菌膜囊泡)在制备抗细菌感染疫苗中的应用,所述铜绿假单胞菌膜囊泡可作为疫苗的佐剂使用。
进一步,所述疫苗佐剂可以非特异性地改变或增强机体对抗原的特异性免疫应答。
进一步,所述细菌感染疾病包括肺炎、泌尿道感染、脑膜炎、败血症,或皮肤、软组织感染。
进一步,所述铜绿假单胞菌膜囊泡还可作为疫苗的载体应用。
B发明制得的铜绿假单胞菌膜囊泡作为免疫原的应用。
B发明制得的铜绿假单胞菌膜囊泡作为抗原递呈细胞功能促进剂中的应用。
进一步,抗原递呈细胞包括树突状细胞,巨噬细胞,及B细胞。
经辐照制备的铜绿假单胞菌膜囊泡在制备DC细胞生长促进剂中的应用,所述铜绿假单胞菌膜囊泡能刺激DC细胞表面共刺激分子CD80、CD86和MHCII显著上调,促进DC细胞成熟和分化。
B发明制得的铜绿假单胞菌膜囊泡作为DC细胞抗原递呈能力促进剂中的应用。
一种促进CD4+T细胞增值的方法,将经辐照制备的铜绿假单胞菌膜囊泡刺激并吞噬OVA抗原的DCs与CFSE标记的CD4+T淋巴细胞在体外共培养。
B发明制得的铜绿假单胞菌膜囊泡在制备兽药中的应用。
B发明所述的灭活铜绿假单胞菌作为细菌疫苗的应用。
B发明的实施例一
B发明提供了一种分离制备铜绿假单胞菌膜囊泡的系统,所述系统依次设有发酵罐、辐照设备、紫外分光光度设备和离心设备;所述辐照设备的射线发生器为X射线发生器、γ射线发生器,或Co 60同位素发生器中的一种或几种;所述离心设备包括离心机、高速离心机、超高速离心机中的一种或多种,见图10。
B发明的实施例二
B发明提供了一种制备铜绿假单胞菌膜囊泡的方法,所述方法包括如下步骤:
1)培养细菌至对数生长期,对数生长期的细菌的OD 600值为0.3-0.8;最好是选择OD 600值为0.5-0.8(此处,还可以进行发酵,使菌体进一 步富集);
2)收集菌体,将菌体用适量的磷酸盐缓冲液重悬并用电离射线辐照处理;所加磷酸盐缓冲液的量与菌体总量的比例为每1ml溶液含有的菌量为OD 600值20-80;最好是选择OD 600值为40-60;
3)用X-ray射线辐照处理,辐照剂量为500-3000Gy,具体包括:500-600Gy,600-700Gy,700-800Gy,800-900Gy,900-1000Gy,1000-1100Gy,1100-1200Gy,1200-1300Gy,1300-1400Gy,1400-1500Gy,1500-1600Gy,1600-1700Gy,1700-1800Gy,1800-1900Gy,1900-2000Gy,2100-2200Gy,2200-2300Gy,2300-2400Gy,2400-2500Gy,2500-2600Gy,2600-2700Gy,2700-2800Gy,2800-2900Gy,2900-3000Gy;
4)将辐照后菌液离心,离心速率为100-10,000g;离心时间为10-60min。上清液用高速离心机再离心,收集上清液,去除鞭毛;高速离心速率为5000-25000g;离心时间为10-100min;
5)将去除鞭毛后的上清超高速离心,沉淀膜囊泡;超高速离心速率为5000-150000g;离心时间为60-600min;
7)收集膜囊泡。
B发明的实施例三
B发明提供了一种分离纯化铜绿假单胞菌膜囊泡的方法,
所述方法包括如下步骤:
1)培养细菌至对数生长期;
2)收集菌液,菌液离心并弃上清液,上清液用0.45μM滤器过滤除菌;离心率为400-8000g;离心时间为10-30min;
3)将过滤后的上清液用高速离心机离心,收集上清液,去除鞭毛;高速离心速率为10000-20000g;离心时间为30-60min;
4)将去除鞭毛后的上清液用超高速离心机离心,沉淀膜囊泡;超高速离心速率为15000-150000g;离心时间为60-180min;
5)收集膜囊泡
B发明实施例四
电离射线辐照铜绿假单胞杆菌PAO1制备、分离纯化膜囊泡:
1.从-80℃复苏铜绿假单胞菌PAO1划线至LB平板,37℃培养箱培养16-18h。
2.从LB平板上挑取单克隆菌落接种于20mL LB液体培养基中,37℃,250rpm恒温培养16-18h。
3.取过夜菌液接种至1L LB培养基中,至起始浓度为0.05OD 600/mL,37℃,250rpm培养至对数生长期,测OD 600值。
4.将上述3)的菌液转移至离心桶中,5,000g离心20min, 收集菌体用生理盐水重悬,调整菌体浓度为50OD左右。
5.取上述菌液置于辐照仪中,辐照剂量为1000Gy。
6.将辐照后的菌液8,000×g,20min,离心两次,收集上清液;上清液用0.45μM滤器过滤除菌再次收集得到的上清,同时取少量上清液涂在LB平板上,37℃培养24-72h,以确认无活菌存在。
7.将步骤6)上清液高速离心机离心,除去上清液中的鞭毛。
8.将步骤7)上清液超高速离心机离心,沉淀膜囊泡。
9.弃去上清,沉淀用MV buffer重悬,-80℃保存。
10.将提取的正常组膜囊泡和本发明实验组膜囊泡进行透射电镜观察。同时测定提取的正常组膜囊泡和本发明实验组膜囊泡的内容物含量,包括测定其DNA含量、RNA含量和蛋白质含量。最后再测量提取的正常组膜囊泡和本发明实验组膜囊泡粒径大小。
实验结果:
由电镜结果可知,电离射线能够刺激绿脓杆菌PAO1产生膜囊泡,实验组膜囊泡数量更多;囊泡形态大小与正常对照组无明显差异。见图1。
内容物含量测定结果:实验组制备的膜囊泡的核酸含量和蛋白质含量与正常对照组相比提高10-20倍。见图2。具体测量数据见表1。
B发明实施例五
辐照处理的细菌膜囊泡的免疫调节作用-促进树突状细胞成熟:
1.小鼠骨髓来源树突状细胞(BMDC)细胞诱导培养:取6-8周C57雌性小鼠,无菌分离小鼠股骨,将股骨上的肌肉剔干净后剪开股骨两端,PBS冲洗骨管腔直至发白,将PBS悬液过滤后1200rpm离5min,去上清,加入5ml红细胞裂解液重悬细胞。静置15min后1200rpm离心5min,去上清,加入50ml 1640完全培养基(20ng/ml GM-CSF、10%FBS、50mM 2-巯基乙醇)重悬细胞。混匀后分至5个培养皿中置培养箱中培养,每2天换液,第7天收集细胞。
2.BMDC刺激:取诱导7天的BMDC细胞,6孔板中反复吹打使贴壁细胞脱落,收集细胞悬液,1100rpm离心5min,去上清,加入1ml培养基重悬细胞、活细胞计数,调整细胞浓度至1×10 6/ml,接种2ml至新的6孔板。加入各刺激物并混匀,分别为:全菌,全菌+囊泡,囊泡,使得终浓度为15μg/mL(以蛋白质为标准),继续培养24小时,生长对照组加入等体积的PBS。
3.流式检测成熟marker:24h后取出6孔板,反复吹打细胞使其脱落,收集细胞悬液至流式管,1500rpm离心3min,去上清, 加入1ml PBS继续1500rpm离心3min后去上清,反复清洗3遍。加入CD11c/CD80/CD86/MHCII抗体室温避光孵育30min,同时阴性对照组应设立一个同型对照组加入CD11c/CD80/CD86/MHCII的同行对照。孵育完成后加入PBS清洗2遍后加入200μl PBS重悬细胞,上流式细胞仪检测。
4.结果处理:流式细胞仪软件分析CD11c细胞中CD80/CD86/MHCII比例。
实验结果:
与全菌体相比,X射线处理的实验组(MVs)囊泡能使刺激后DCs表面共刺激分子CD80、CD86和MHCII等显著上调,而这些表面分子是树突状细胞成熟的标志。综上证明囊泡能显著促进DCs的分化和成熟。见图5。
B发明实施例六
通过检测FITC标记的葡聚糖荧光强度来检测DC细胞的吞噬能力:
1.BMDC细胞诱导培养。
2.刺激:第7天收集细胞,将细胞全部吹下后离心重悬计数,然后接种到6孔板,每孔接种1×10 6个细胞,再分别加入刺激物,GC组加入等体积的PBS,Control组及Treatment组加入相同浓度的膜囊泡(按蛋白质水平)后37℃培养24h。
3.吞噬与检测:加入葡聚糖(5μg/ml),继续培养1h后,吸出细胞到流式管,PBS洗涤3次后加入CD11c抗体室温避光孵育30min,PBS清洗3次后流式检测FITC荧光。
4.结果处理:流式细胞仪软件分析CD11c细胞中FITC比例。
实验结果:
为了检测DCs的吞噬功能,我们采用FITC-葡聚糖作为模式抗原供DCs吞噬,检测CD11c+DCs的FITC平均荧光强度值。实验结果显示,GC组树突状细胞(生长对照组)基本没有FITC-葡聚糖摄入,但是无论是DCs接受刺激后,FITC平均荧光强度值相比于GC组明显降低。这个实验结果再次证明囊泡能够促进DCs成熟,从而降低对抗原的摄取能力。见图6。
B发明实施例七
X射线处理组细菌膜囊泡刺激后的成熟DC与T细胞相互作用:
A、成熟DC与CD4+T细胞相互作用:
1.BMDC细胞诱导培养。
2.抗原吞噬:将培养7天的DCs在含10μg/ml OVA培养基 中培养24h作为GC(growth control,生长对照组),MVs组别另外加入囊泡,接下来离心收集吞噬过抗原的DCs并重悬在正常培养基中,以2×10 4细胞/孔的密度铺板于96孔板中,每孔100μl,每组3个复孔。
3.T细胞提取:在第二天,OT-II小鼠脾脏OVA特异性的CD4+T淋巴细胞使用Stem Cell Technologies公司的阴性磁珠筛选试剂盒分离富集。
4.DC与T细胞共培养:对分选出的CD4+T细胞按照试剂盒说明书使用1μM CFSE进行标记。标记后用PBS洗涤3次,以10 5细胞/孔的密度加入到96孔板中,使其培养终体积为200μl(CD4:DC=5:1)。
5.在共培养后的第3天,利用流式细胞术通过CFSE递减来检测CD4+T细胞群的增殖情况。
实验结果:
将囊泡刺激并吞噬OVA抗原的DCs与CFSE标记的OT-II小鼠CD4+T淋巴细胞在体外共培养。流式分析CFSE荧光强度结果表明,增殖CD4+T细胞的比例增加。囊泡(14.05%)能显著增加吞噬OVA抗原的DCs(6.80%)对于特异性CD4+T细胞的促进增殖作用。详见图7、图8。
B、囊泡处理后的DC促进T细胞增殖:
1.BMDC细胞诱导培养。
2.抗原吞噬:将培养7天的DCs在培养基中培养24h作为GC(growth control,生长对照组),MVs组别另外加入囊泡,接下来离心收集吞噬过抗原的DCs并重悬在正常培养基中,以4×10 4细胞/孔的密度铺板于96孔板中,每孔100μl,每组3个复孔。
3.T细胞提取:在第二天,使用MVs免疫一次后一周的小鼠脾脏Stem Cell Technologies公司的阴性磁珠筛选试剂盒分离富集小鼠的T细胞。
4.DC与T细胞共培养:对分选出的T细胞按照试剂盒说明书使用1μM CFSE进行标记。标记后用PBS洗涤3次,以4×10 5细胞/孔的密度加入到96孔板中,使其培养终体积为200μl(CD3:DC=10:1)。
5.在共培养后的第3天,利用流式细胞术通过CFSE递减来检测CD3+,CD8+,CD4+T细胞群的增殖情况。
实验结果:
将囊泡刺激并吞噬OVA抗原的DCs与CFSE标记的OT-II小鼠CD4+T淋巴细胞在体外共培养。流式分析CFSE荧光强度结果表明,增殖CD4+T细胞的比例增加。如图所示,菌体加囊泡刺激组 荧光强度为63.5%,囊泡刺激组为71%。说明,囊泡处理后DCs,能显著刺激CD4+T细胞的增殖。见图9。
C发明:金黄色葡萄球菌膜囊泡及其制备方法与应用
C发明属于微生物学领域,具体涉及一种金黄色葡萄球菌膜囊泡的制备分离纯化以及该膜囊泡的应用。
现代工艺技术在生产制备及纯化细菌膜囊泡时存在以下问题:1)利用抗生素、去污剂、氧化剂等手段虽然能促进膜囊泡产生,但也伴随着有毒残留问题,给其应用带来了不确定性。2)上述干预因素刺激细菌产生膜囊泡的效率也相对较低,且该制备工艺不能实现标准化生产。3)上述方法会使菌体本身的外膜抗原性、构象等可能发生变化,进而影响囊泡,限制其后续应用。
C发明提供了一种分离金黄色葡萄球菌MVs的方法,和一种制备金黄色葡萄球菌MVs的方法。本发明采用国际领先的工艺技术,不添加化学刺激物质,因此无不良作用,同时工艺流程简单、囊泡产量高,高效、放大效果好,可以用于囊泡的大量制备。制备的囊泡与正常囊泡相比具有较好的免疫原性,后续进一步的开发应用前景广阔。
C发明的目的在于提供一种金黄色葡萄球菌模膜囊泡。
金黄色葡萄球菌产生的生物颗粒,所述生物颗粒是从灭活金黄色葡萄球菌中分离的膜囊泡。
含有上述生物颗粒的灭活金黄色葡萄球菌。
进一步,所述灭活金黄色葡萄球菌与所述作用疫苗和/或疫苗佐剂和/或药物载体的生物颗粒组合。
C发明的目的还在于提供一种分离金黄色葡萄球菌膜囊泡的方法。
一种从金黄色葡萄球菌中分离膜囊泡的方法,包括如下步骤:
1)培养细菌至对数生长期;
2)收集菌液,菌液离心并取上清液1,上清液1用0.3-0.5μM滤器过滤除菌;
3)将过滤后的上清液1用高速离心机离心,收集上清液2;
4)将去除鞭毛后的上清液2用超高速离心机离心,沉淀即得到膜囊泡。
进一步,步骤2)的上清液用0.45μM滤器过滤除菌。
进一步,步骤2)的离心速率为100-10000g;离心时间为10-60min。
优选的,所述离心速率为400-8000g;离心时间为10-30min。
进一步,步骤3)的高速离心速率为5000-25000g;离心时间为10-100min。
优选的,所述的高速离心速率为10000-20000g;离心时间为30-60min。
进一步,步骤4)的超高速离心速率为5000-150000g;离心时间为60-600min。
优选的,所述的超高速离心速率为15000-150000g;离心时间为60-180min。
进一步,所述金黄色葡萄球菌为灭活的金黄色葡萄球菌。
采用上述分离方法得到的金黄色葡萄球菌膜囊泡。
C发明的目的还在于提供一种制备方法。
制备上述生物颗粒的方法,所述方法包括如下步骤:
1)培养细菌至对数生长期;
2)收集菌体,将菌体用适量的磷酸盐缓冲液或无菌生理盐水重悬并用电离射线辐照处理;
3)收集辐照处理后的菌液,离心并取上清液1,上清液1用03-0.5μM滤器过滤除菌;
4)将过滤后的上清液1用高速离心机离心,收集上清液2;
5)将上清2超高速离心,沉淀即得膜囊泡。
进一步,步骤1)中所述对数生长期的细菌的OD 600值为0.3-0.8。
进一步,其特征在于,步骤2)中所加磷酸盐缓冲液或无菌生理盐水的量与菌体总量的比例为每1ml溶液含有的菌量为OD 600值20-80。
进一步,步骤2)中辐照处理用的射线为X-ray射线;辐照剂量为500-3000Gy。所述辐照剂量具体包括:500-600Gy,600-700Gy,700-800Gy,800-900Gy,900-1000Gy,1000-1100Gy,1100-1200Gy,1200-1300Gy,1300-1400Gy,1400-1500Gy,1500-1600Gy,1600-1700Gy,1700-1800Gy,1800-1900Gy,1900-2000Gy,2100-2200Gy,2200-2300Gy,2300-2400Gy,2400-2500Gy,2500-2600Gy,2600-2700Gy,2700-2800Gy,2800-2900Gy,2900-3000Gy。
优选的,辐照剂量为500-1000Gy。所述辐照剂量具体包括:500-600Gy,600-700Gy,700-800Gy,800-900Gy,900-1000Gy。
进一步,步骤3)中的离心速率为100-10000g;离心时间为10-60min。
优选的,所述离心速率为400-8000g;离心时间为10-30min。
进一步,步骤3)中上清液用0.45μM滤器过滤除菌。
进一步,步骤4)中所述的高速离心速率为5000-25000g; 离心时间为10-100min。
优选的,所述高速离心速率为10000-20000g;离心时间为30-60min。
进一步,步骤5)中所述的超高速离心速率为5000-150000g;离心时间为60-600min。
优选的,所述的超高速离心速率为15000-150000g;离心时间为60-180min。
采用上述方法制得得到的金黄色葡萄球菌膜囊泡。
一种提高金黄色葡萄球菌膜囊泡内容物含量的方法,所述内容物包括核酸和蛋白质,所述方法采用辐照设备处理金黄色葡萄球菌菌液,所述辐照设备的射线为X-ray射线,辐照剂量为500-1000Gy。
C发明的目的还在于提供所述金黄色葡萄球菌膜囊泡及灭活菌的应用。
C发明制得的金黄色葡萄球菌膜囊泡(以下均简称C发明制得的金黄色葡萄球菌膜囊泡)在制备抗细菌感染疫苗中的应用,所述金黄色葡萄球菌膜囊泡可作为疫苗的佐剂使用。
进一步,所述疫苗佐剂可以非特异性地改变或增强机体对抗原的特异性免疫应答。
进一步,所述细菌感染疾病包括肺炎、泌尿道感染、脑膜炎、败血症,或皮肤、软组织感染。
进一步,所述金黄色葡萄球菌膜囊泡还可作为疫苗的载体应用。
C发明制得的金黄色葡萄球菌膜囊泡作为免疫原的应用。
C发明制得的金黄色葡萄球菌膜囊泡作为抗原递呈细胞功能促进剂中的应用。
进一步,抗原递呈细胞包括树突状细胞,巨噬细胞,及B细胞。
经辐照制备的金黄色葡萄球菌膜囊泡在制备DC细胞生长促进剂中的应用,所述金黄色葡萄球菌膜囊泡能刺激DC细胞表面共刺激分子CD80、CD86和MHCII显著上调,促进DC细胞成熟和分化。
C发明制得的金黄色葡萄球菌膜囊泡作为DC细胞抗原递呈能力促进剂中的应用。
一种促进CD4和T细胞增殖的组合物,所述组合物包含上述的金黄色葡萄球菌膜囊泡和DC细胞。
一种促进CD4+T细胞增值的方法,将经辐照制备的金黄色葡萄球菌膜囊泡刺激并吞噬OVA抗原的DCs与CFSE标记的CD4+T淋巴细胞在体外共培养。
C发明制得的金黄色葡萄球菌膜囊泡在制备兽药中的应用。
C发明所述的灭活金黄色葡萄球菌作为细菌疫苗的应用。
C发明实施例一
C发明提供了一种分离制备金黄色葡萄球菌膜囊泡的系统,所述系统依次设有发酵罐、辐照设备、紫外分光光度设备和离心设备;所述辐照设备的射线发生器为X射线发生器、γ射线发生器,或Co 60同位素发生器中的一种或几种;所述离心设备包括离心机、高速离心机、超高速离心机中的一种或多种,见图10。
C发明实施例二
C发明提供了一种制备金黄色葡萄球菌膜囊泡的方法,所述方法包括如下步骤:
1)培养细菌至对数生长期,所述对数生长期的细菌的OD 600值为0.3-0.8(此处还可以对细菌进行发酵,使菌体进一步富集);
2)收集菌体,将菌体用适量的磷酸盐缓冲液或无菌生理盐水重悬,优先选用无菌生理盐水,且所加生理盐水的量与菌体总量的比例为每1ml溶液含有的菌量为OD 600值20-80;
3)用X-ray射线辐照处理,辐照剂量为500-3000Gy,具体包括:500-600Gy,600-700Gy,700-800Gy,800-900Gy,900-1000Gy,1000-1100Gy,1100-1200Gy,1200-1300Gy,1300-1400Gy,1400-1500Gy,1500-1600Gy,1600-1700Gy,1700-1800Gy,1800-1900Gy,1900-2000Gy,2100-2200Gy,2200-2300Gy,2300-2400Gy,2400-2500Gy,2500-2600Gy,2600-2700Gy,2700-2800Gy,2800-2900Gy,2900-3000Gy;
4)将辐照后菌液离心,离心速率为100-10000g;离心时间为10-60min。上清液用高速离心机再离心,收集上清液;高速离心速率为5000-25000g;离心时间为10-100min;
5)将上清超高速离心,沉淀膜囊泡;超高速离心速率为5000-150000g;离心时间为60-600min。
7)收集膜囊泡。
C发明实施例三
C发明提供了一种分离金黄色葡萄球菌膜囊泡的方法,所述方法包括如下步骤:
1)培养细菌至对数生长期然后发酵;
2)收集菌液,菌液离心并弃上清液,上清液用03-0.5μM滤器过滤除菌;离心速率为400-8000g;离心时间为10-30min;
3)将过滤后的上清液用高速离心机离心,收集上清液;高速离心速率为10000-20000g;离心时间为30-60min;
4)将上清液用超高速离心机离心,沉淀膜囊泡;超高速离心速率为15000-150000g;离心时间为60-180min;
5)收集膜囊泡。
C发明实施例四
电离射线辐照金黄色葡萄球菌ATCC 25923制备膜囊泡:
1.从-80℃复苏金黄色葡萄球菌ATCC 25923划线至LB平板,37℃培养箱培养16-18h。
2.从LB平板上挑取单克隆菌落接种于100mL LB液体培养基中,37℃,250rpm恒温培养16-18h。
3.取过夜菌液接种至2L LB培养基中,至起始浓度为0.05OD 600/mL,37℃,250rpm培养至对数生长期,测OD 600值。
4.将上述3)菌液转移至2L离心桶中,5,000g离心20min,收集菌体用生理盐水重悬,调整菌体浓度为50OD左右,取100μL稀释10 7涂板计算活菌数。
5.取上述菌液置于辐照仪中,辐照剂量为1000Gy。
6.将辐照后的菌液8,000×g,20min,离心两次,收集上清液;上清液用0.45μM滤器过滤除菌再次收集得到的上清,同时取少量上清液涂在LB平板上,37℃培养24-72h,以确认无活菌存在。
7.将步骤6)上清液超高速离心机离心,39,000×g,90min,沉淀MVs。
8.弃去上清,沉淀用2mL MV buffer重悬,-80℃保存。
9.测定提取的正常组膜囊泡和本发明实验组膜囊泡的内容物含量,包括测定其DNA含量、RNA含量和蛋白质含量。
实验结果:
经测定结果表明,对于金黄色葡萄球菌ATCC27853,实验组制备的膜囊泡的核酸含量和蛋白质含量明显提高,表明,电离射线辐照革兰氏阳性菌也能刺激膜囊泡的产生。见图4。
C发明实施例五
辐照处理的细菌膜囊泡的免疫调节作用-促进树突状细胞成熟:
1.小鼠骨髓来源树突状细胞(BMDC)细胞诱导培养:取6-8周C57雌性小鼠,无菌分离小鼠股骨,将股骨上的肌肉剔干净后剪开股骨两端,PBS冲洗骨管腔直至发白,将PBS悬液过滤后1200rpm离5min,去上清,加入5ml红细胞裂解液重悬细胞。静置15min后1200rpm离心5min,去上清,加入50ml 1640完全培养基(20ng/ml GM-CSF、10%FBS、50mM 2-巯基乙醇)重悬细胞。混匀后分至5个培养皿中置培养箱中培养,每2天换液,第7天收集细胞。
2.BMDC刺激:取诱导7天的BMDC细胞,6孔板中反复 吹打使贴壁细胞脱落,收集细胞悬液,1100rpm离心5min,去上清,加入1ml培养基重悬细胞、活细胞计数,调整细胞浓度至1×10 6/ml,接种2ml至新的6孔板。加入各刺激物并混匀,分别为:全菌,全菌+囊泡,囊泡,使得终浓度为15μg/mL(以蛋白质为标准),继续培养24小时,生长对照组加入等体积的PBS。
3.流式检测成熟marker:24h后取出6孔板,反复吹打细胞使其脱落,收集细胞悬液至流式管,1500rpm离心3min,去上清,加入1ml PBS继续1500rpm离心3min后去上清,反复清洗3遍。加入CD11c/CD80/CD86/MHCII抗体室温避光孵育30min,同时阴性对照组应设立一个同型对照组加入CD11c/CD80/CD86/MHCII的同行对照。孵育完成后加入PBS清洗2遍后加入200μl PBS重悬细胞,上流式细胞仪检测。
4.结果处理:流式细胞仪软件分析CD11c细胞中CD80/CD86/MHCII比例。
实验结果:
与全菌体相比,X射线处理的实验组(MVs)囊泡能使刺激后DCs表面共刺激分子CD80、CD86和MHCII等显著上调,而这些表面分子是树突状细胞成熟的标志。综上证明囊泡能显著促进DCs的分化和成熟。见图5。
C发明实施例六
通过检测FITC标记的葡聚糖荧光强度来检测DC细胞的吞噬能力:
1.BMDC细胞诱导培养。
2.刺激:第7天收集细胞,将细胞全部吹下后离心重悬计数,然后接种到6孔板,每孔接种1×10 6个细胞,再分别加入刺激物,GC组加入等体积的PBS,Control组及Treatment组加入相同浓度的膜囊泡(按蛋白质水平)后37℃培养24h。
3.吞噬与检测:加入葡聚糖(5μg/ml),继续培养1h后,吸出细胞到流式管,PBS洗涤3次后加入CD11c抗体室温避光孵育30min,PBS清洗3次后流式检测FITC荧光。
4.结果处理:流式细胞仪软件分析CD11c细胞中FITC比例。
实验数据:
为了检测DCs的吞噬功能,我们采用FITC-葡聚糖作为模式抗原供DCs吞噬,检测CD11c+DCs的FITC平均荧光强度值。实验结果显示,GC组树突状细胞(生长对照组)基本没有FITC-葡聚糖摄入,但是无论是DCs接受刺激后,FITC平均荧光强度值相比于GC组明显降低。这个实验结果再次证明囊泡能够促进DCs成熟,从 而降低对抗原的摄取能力。见图6。
C发明实施例七
X射线处理组细菌膜囊泡刺激后的成熟DC与T细胞相互作用:
A、成熟DC与CD4+T细胞相互作用:
1.BMDC细胞诱导培养。
2.抗原吞噬:将培养7天的DCs在含10μg/ml OVA培养基中培养24h作为GC(growth control,生长对照组),MVs组别另外加入囊泡,接下来离心收集吞噬过抗原的DCs并重悬在正常培养基中,以2×10 4细胞/孔的密度铺板于96孔板中,每孔100μl,每组3个复孔。
3.T细胞提取:在第二天,OT-II小鼠脾脏OVA特异性的CD4+T淋巴细胞使用Stem Cell Technologies公司的阴性磁珠筛选试剂盒分离富集。
4.DC与T细胞共培养:对分选出的CD4+T细胞按照试剂盒说明书使用1μM CFSE进行标记。标记后用PBS洗涤3次,以10 5细胞/孔的密度加入到96孔板中,使其培养终体积为200μl(CD4:DC=5:1)。
5.在共培养后的第3天,利用流式细胞术通过CFSE递减来检测CD4+T细胞群的增殖情况。
实验结果:
将囊泡刺激并吞噬OVA抗原的DCs与CFSE标记的OT-II小鼠CD4+T淋巴细胞在体外共培养。流式分析CFSE荧光强度结果表明,增殖CD4+T细胞的比例增加。囊泡(14.05%)能显著增加吞噬OVA抗原的DCs(6.80%)对于特异性CD4+T细胞的促进增殖作用。详见图7、图8。
B、囊泡处理后的DC促进T细胞增殖:
1.BMDC细胞诱导培养。
2.抗原吞噬:将培养7天的DCs在培养基中培养24h作为GC(growth control,生长对照组),MVs组别另外加入囊泡,接下来离心收集吞噬过抗原的DCs并重悬在正常培养基中,以4×10 4细胞/孔的密度铺板于96孔板中,每孔100μl,每组3个复孔。
3.T细胞提取:在第二天,使用MVs免疫一次后一周的小鼠脾脏Stem Cell Technologies公司的阴性磁珠筛选试剂盒分离富集小鼠的T细胞。
4.DC与T细胞共培养:对分选出的T细胞按照试剂盒说明书使用1μM CFSE进行标记。标记后用PBS洗涤3次,以4×10 5细胞/孔的密度加入到96孔板中,使其培养终体积为200μl (CD3:DC=10:1)。
5.在共培养后的第3天,利用流式细胞术通过CFSE递减来检测CD3+,CD8+,CD4+T细胞群的增殖情况。
实验结果:
将囊泡刺激并吞噬OVA抗原的DCs与CFSE标记的OT-II小鼠CD4+T淋巴细胞在体外共培养。流式分析CFSE荧光强度结果表明,增殖CD4+T细胞的比例增加。如图所示,菌体加囊泡刺激组荧光强度为63.5%,囊泡刺激组为71%。说明,囊泡处理后DCs,能显著刺激CD4+T细胞的增殖。见图9。
D发明:一种细菌膜囊泡的生产系统和分离纯化系统
D发明涉及生物技术领域,具有涉及一种细菌膜囊泡的生产系统和分离纯化系统。
针对现代工艺技术在制备及纯化细菌膜囊泡时存在的问题,D发明提供一种易于工业化使用的细菌膜囊泡生产系统和纯化系统。
一种细菌膜囊泡的生产系统,所述生产系统依次设有发酵单元1和辐照单元2,所述发酵单元1由细菌发酵罐3和紫外分光光度计4构成,所述辐照单元2由辐照设备5构成。
D发明提供的发酵罐适合用于各种细菌的发酵,包括革兰氏阳性菌和革兰氏阴性菌。
D发明提供的紫外分光光度设备用于精确测量菌液的含菌量。
进一步,所述发酵罐3为通风发酵罐、鼓泡发酵罐、气升式发酵罐,或喷嘴环流式发酵罐。
进一步,所述辐照设备5中的射线发射器为X射线发生器、γ射线发生器,或Co 60同位素发生器。
D发明的另一目的在于提供一种细菌膜囊泡的纯化系统。
一种用于细菌膜囊泡的纯化系统,所述纯化系统依次设有发酵单元1、辐照单元2和分离设备6,所述发酵单元1由细菌发酵罐3和紫外分光光度计4构成,所述辐照单元2由辐照设备5构成。
进一步,所述分离设备6包括离心单元7、膜包单元和/或过柱单元。
进一步,所述发酵罐3为通风发酵罐、鼓泡发酵罐、气升式发酵罐,或喷嘴环流式发酵罐。
进一步,所述辐照设备5中的射线发射器为X射线发生器、γ射线发生器,或Co 60同位素发生器。
进一步,所述分离设备6包括离心单元7,离心单元7为 普通离心机、高速离心机,或超高速离心机。
D发明的另一目的在于提供一种细菌膜囊泡的分离系统。
一种细菌膜囊泡的分离系统,所述分离系统依次设有发酵单元1和分离设备6,所述发酵单元1由细菌发酵罐3和紫外分光光度计4构成。
进一步,所述分离设备6包括离心单元7、膜包单元和/或过柱单元。
进一步,所述发酵罐3为通风发酵罐、鼓泡发酵罐、气升式发酵罐,或喷嘴环流式发酵罐。
进一步,所述分离设备6包括离心单元7,离心单元7为普通离心机、高速离心机,或超高速离心机。
D发明实施例一
参见图10。具体地,D实施例的生产系统依次设有发酵单元1和辐照单元2,所述发酵单元1由细菌发酵罐3和紫外分光光度计4构成,所述辐照单元2由辐照设备5构成。所述发酵罐3为通风发酵罐、鼓泡发酵罐、气升式发酵罐,或喷嘴环流式发酵罐。所述辐照设备5中的射线发射器为X射线发生器、γ射线发生器,或Co 60同位素发生器。
D实施例中,革兰氏阳性菌和革兰氏阴性菌首先在发酵罐3中进行发酵,然后通过该紫光分光光度计4精确控制发酵菌液的含菌量在一定限度内,再利用辐照设备5对发酵后的菌液进行射线辐照处理,从而促进细菌大量产生MVs,以提高MVs含量。
D发明实施例二
参见图11。具体的,D实施例的纯化系统依次设有发酵单元1、辐照单元2和分离设备6,所述发酵单元1由细菌发酵罐3和紫外分光光度计4构成,所述辐照单元2由辐照设备5构成,所述分离设备6由离心单元7、膜包单元和/或过柱单元构成。
D实施例中,革兰氏阳性菌和革兰氏阴性菌首先在该发酵罐3中进行发酵,然后通过紫外分光光度计4精确控制发酵菌液的含菌量在一定限度内,再利用辐照设备5对发酵后的菌液进行射线辐照处理,最后利用离心单元7对辐照处理后的菌液进行进一步的离心处理,依次除去发酵废液、细菌鞭毛、细菌分泌物等,最后沉淀获得的膜囊泡即为纯化后的膜囊泡。
D实施例中,该射线辐照设备的射线发生器为X射线发生器、γ射线发生器,或Co 60同位素发生器。
D实施例中,发酵罐3为通风发酵罐、鼓泡发酵罐、气升式发酵罐,或喷嘴环流式发酵罐。
D实施例中,离心单元7包括离心机、高速离心机或超高 速离心机。
D发明实施例三
参见图12。具体的,D实施例的分离系统依次设有发酵单元1和离心单元6,所述发酵单元1由细菌发酵罐3和紫外分光光度计4构成,所述分离设备6由离心单元7构成。
D实施例中,革兰氏阳性菌和革兰氏阴性菌首先在该发酵罐3中进行发酵,然后再通过离心机7依次除去发酵废液、细菌鞭毛、细菌分泌物等废物。
D实施例中,发酵罐3为通风发酵罐、鼓泡发酵罐、气升式发酵罐,或喷嘴环流式发酵罐。
D实施例中,离心单元7包括离心机、高速离心机或超高速离心机。
E发明:一种细菌膜囊泡的生产系统和分离纯化系统及方法
E发明涉及生物技术领域,具有涉及一种细菌膜囊泡的生产系统及方法,以及一种细菌膜囊泡的分离纯化系统及方法。
基于现代工艺技术在生产制备及纯化细菌膜囊泡时存在的问题,E本发明提供了一套提高细菌MVs产量的系统及方法,和一套纯化细菌MVs的系统及方法。本发明所采用工艺技术,不添加化学刺激物质,因此无不良作用,同时工艺流程简单、囊泡产量高,高效、放大效果好,可以用于囊泡的大量制备。
E本发明的目的在于提供一种细菌膜囊泡的生产系统。
一种细菌膜囊泡的生产系统,所述生产系统依次设有发酵单元1和辐照单元2,所述发酵单元1由细菌发酵罐3和紫外分光光度计4构成,所述辐照单元2由辐照设备5构成。
进一步,所述发酵罐3为通风发酵罐、鼓泡发酵罐、气升式发酵罐,或喷嘴环流式发酵罐。
进一步,所述辐照设备5中的射线发射器为X射线发生器、γ射线发生器,或Co 60同位素发生器。
E发明的目的还在于提供一种细菌膜囊泡的生产方法。
采用上述的生产系统生产细菌膜囊泡的方法,包括如下步骤:
1)培养细菌至对数生长期,发酵,使菌体进一步富集;
2)收集菌体,将菌体用适量的磷酸盐缓冲液或无菌生理盐水重悬;
3)用电离射线辐照处理。
本发明所述细菌包括革兰氏阳性菌和革兰氏阴性菌。所述细菌具体包括如下种类:
Figure PCTCN2019106654-appb-000001
进一步,步骤1)中所述对数生长期的细菌的OD 600值为0.3-0.8;步骤2)中所加磷酸盐缓冲液或无菌生理盐水的量与菌体总量的比例为每1ml溶液含有的菌量为OD 600值20-80。
优选的,步骤1)中所述对数生长期的细菌的OD 600值为0.5-0.8;步骤2)中所加磷酸盐缓冲液或无菌生理盐水的量与菌体总量的比例为每1ml溶液含有的菌量OD 600值为40-60。
进一步,步骤3)中辐照处理用的射线为X-ray射线;辐照剂量范围为500-3000Gy。所述辐照剂量具体包括:500-600Gy,600-700Gy,700-800Gy,800-900Gy,900-1000Gy,1000-1100Gy,1100-1200Gy,1200-1300Gy,1300-1400Gy,1400-1500Gy,1500-1600Gy,1600-1700Gy,1700-1800Gy,1800-1900Gy,1900-2000Gy,2100-2200Gy,2200-2300Gy,2300-2400Gy,2400-2500Gy,2500-2600Gy,2600-2700Gy,2700-2800Gy,2800-2900Gy,2900-3000Gy。
E发明的目的还在于提供一种细菌膜囊泡的纯化系统。
一种用于细菌膜囊泡的纯化系统,所述纯化系统依次设有发酵单元1、辐照单元2和离心单元6,所述发酵单元1由细菌发酵罐3和紫外分光光度计4构成,所述辐照单元2由辐照设备5构成,所述离心设备6由离心单元7构成。
进一步,所述发酵罐3为通风发酵罐、鼓泡发酵罐、气升式发酵罐,或喷嘴环流式发酵罐。
进一步,所述辐照设备5中的射线发射器为X射线发生器、γ射线发生器,或Co 60同位素发生器。
进一步,所述离心单元7为普通离心机、高速离心机,或超高速离心机。
E发明的目的还在于提供一种细菌膜囊泡的纯化方法。
采用上述的纯化系统纯化细菌膜囊泡的方法,其特征在于,包括如下步骤:
1)培养细菌至对数生长期,发酵,使菌体进一步富集;
2)收集菌体,将菌体用适量的磷酸盐缓冲液或无菌生理盐水重悬并 用电离射线辐照处理;
3)收集辐照处理后的菌液,离心并弃上清液,上清液用0.3-0.5μM滤器过滤除菌;
4)将过滤后的上清液用高速离心机离心,收集上清液,去除鞭毛;
5)将去除鞭毛后的上清超高速离心,沉淀膜囊泡;
6)收集纯化后的膜囊泡。
进一步,步骤1)中所述对数生长期的细菌的OD 600值为0.3-0.8;步骤2)中所加磷酸盐缓冲液或无菌生理盐水的量与菌体总量的比例为每1ml溶液含有的菌量为OD 600值20-80。
优选的,步骤1)中所述对数生长期的细菌的OD 600值为0.5-0.8;步骤2)中所加磷酸盐缓冲液或无菌生理盐水的量与菌体总量的比例为每1ml溶液含有的菌量OD 600值为40-60。
进一步,步骤3)中辐照处理用的射线为X-ray射线;辐照剂量范围为500-3000Gy。所述辐照剂量具体包括:500-600Gy,600-700Gy,700-800Gy,800-900Gy,900-1000Gy,1000-1100Gy,1100-1200Gy,1200-1300Gy,1300-1400Gy,1400-1500Gy,1500-1600Gy,1600-1700Gy,1700-1800Gy,1800-1900Gy,1900-2000Gy,2100-2200Gy,2200-2300Gy,2300-2400Gy,2400-2500Gy,2500-2600Gy,2600-2700Gy,2700-2800Gy,2800-2900Gy,2900-3000Gy。
进一步,步骤3)中辐照后菌液离心的速率为100-10000g;离心时间为10-60min。
优选的,步骤3)的离心的速率为400-8000g;离心时间为10-30min。
进一步,步骤4)中高速离心的速率为5000-25000g;离心时间为10-100min。
优选的,步骤4)的高速离心速率为10000-20000g;离心时间为30-60min。
进一步,步骤5)中超高速离心的速率为5000-150000g;离心时间为60-600min。
优选的,步骤5)的超高速离心速率为15000-150000g;离心时间为60-180min。
E发明实施例一
参见图10。具体地,E实施例的生产系统依次设有发酵单元1和辐照单元2,所述发酵单元1由细菌发酵罐3和紫外分光光度计4构成,所述辐照单元2由辐照设备5构成。所述发酵罐3为通风发酵罐、鼓泡发酵罐、气升式发酵罐,或喷嘴环流式发酵罐。所述辐照设备5中的射线发射器为X射线发生器、γ射线发生器,或Co 60同位素发生器。
E实施例中,革兰氏阳性菌和革兰氏阴性菌首先在发酵罐3中进行发酵,然后通过该紫光分光光度计4精确控制发酵菌液的含菌量在一定限度内,再利用辐照设备5对发酵后的菌液进行射线辐照处理,从而促进细菌大量产生MVs。
采用上述生产系统生产细菌膜囊泡的方法,包括如下步骤:
1)培养细菌至对数生长期,对数生长期的细菌的OD 600值为0.3-0.8;然后发酵,使菌体进一步富集;
2)收集菌体,将菌体用适量的磷酸盐缓冲液或无菌生理盐水重悬,所加磷酸盐缓冲液或无菌生理盐水的量与菌体总量的比例为每1ml溶液含有的菌量OD 600值为20-80;
3)用X-ray射线辐照菌液,辐照剂量范围为500-3000Gy,可具体选择的限度包括500-600Gy,600-700Gy,700-800Gy,800-900Gy,900-1000Gy,1000-1100Gy,1100-1200Gy,1200-1300Gy,1300-1400Gy,1400-1500Gy,1500-1600Gy,1600-1700Gy,1700-1800Gy,1800-1900Gy,1900-2000Gy,2100-2200Gy,2200-2300Gy,2300-2400Gy,2400-2500Gy,2500-2600Gy,2600-2700Gy,2700-2800Gy,2800-2900Gy,2900-3000Gy。
E发明实施例二
参见图11。具体的,E实施例的纯化系统依次设有发酵单元1、辐照单元2和离心单元6,所述发酵单元1由细菌发酵罐3和紫外分光光度计4构成,所述辐照单元2由辐照设备5构成,所述离心单元6由离心机7构成。
E实施例中,革兰氏阳性菌和革兰氏阴性菌首先在该发酵罐3中进行发酵,然后通过紫外分光光度计4精确控制发酵菌液的含菌量在一定限度内,再利用辐照设备5对发酵后的菌液进行射线辐照处理,最后利用离心设备6对辐照处理后的菌液进行进一步的离心处理,依次除去发酵废液、细菌鞭毛、细菌分泌物等,最后沉淀获得的膜囊泡即为纯化后的膜囊泡。
E实施例中,该射线辐照设备的射线发生器为X射线发生器、γ射线发生器,或Co 60同位素发生器。
E实施例中,发酵罐3为通风发酵罐、鼓泡发酵罐、气升式发酵罐,或喷嘴环流式发酵罐。
E实施例中,离心单元7包括离心机、高速离心机或超高速离心机。
采用上述纯化系统纯化细菌膜囊泡的方法,包括如下步骤:
1)培养细菌至对数生长期,对数生长期的细菌的OD 600值为0.3-0.8;发酵,使菌体进一步富集;
2)收集菌体,将菌体用适量的磷酸盐缓冲液或无菌生理盐水重悬, 所加磷酸盐缓冲液或无菌生理盐水的量与菌体总量的比例为每1ml溶液含有的菌量为OD 600值20-80;重悬后的菌液用X-ray射线辐照处理,辐照剂量范围为500-3000Gy;
3)收集辐照处理后的菌液离心,离心的速率为100-10000g,离心时间为10-60min;离心完毕后弃上清液,上清液用0.45μM滤器过滤除菌;
4)将除菌后的上清液用高速离心机离心,高速离心的速率为5000-25000g,离心时间为10-100min;收集上清液,去除鞭毛;
5)将去除鞭毛后的上清超高速离心,超高速离心的速率为5000-150000g,离心时间为60-600min;沉淀膜囊泡;
6)收集优化生产后的膜囊泡。
上面结合附图对本发明的实施例进行了描述,但是本发明并不局限于上述的具体实施方式,上述的具体实施方式仅仅是示意性的,而不是限制性的,本领域的普通技术人员在本发明的启示下,在不脱离本发明宗旨和权利要求所保护的范围情况下,还可做出很多形式,这些均属于本发明的保护之内。
参考文献
Beveridge T J.Structures of gram-negative cell walls and their derived membrane vesicles[J].Journal of bacteriology,1999,181(16):4725-4733.
Kulp A,Kuehn M J.Biological functions and biogenesis of secreted bacterial outer membrane vesicles[J].Annual review of microbiology,2010,64(163-184).
Devos S,Van Oudenhove L,Stremersch S,Van Putte W,De Rycke R,et.al.The effect of imipenem and diffusible signaling factors on the secretion of outer membrane vesicles and associated Ax21proteins in Stenotrophomonas maltophilia[J].Frontiers in Microbiology,2015vol:6pp:298.

Claims (132)

  1. 细菌产生的生物颗粒,其特征在于,所述生物颗粒是从灭活细菌中分离的膜囊泡。
  2. 根据权利要求1所述的生物颗粒,其特征在于,所述灭活细菌包括革兰氏阳性菌、革兰氏阴性菌和/或基因工程改造菌。
  3. 根据权利要求2所述的生物颗粒,其特征在于,所述革兰氏阳性菌包括金黄色葡萄球菌、链球菌、白喉杆菌、结核杆菌、破伤风杆菌、芽孢杆菌、棒状杆菌,以及上述菌的基因工程改造菌中的一种或多种。
  4. 根据权利要求2所述的生物颗粒,其特征在于,所述革兰氏阴性菌包括铜绿假单胞菌、鲍曼不动杆菌、大肠杆菌、伤寒杆菌、脑膜炎双球菌、变形杆菌、志贺菌,以及上述菌的基因工程改造菌中的一种或多种。
  5. 根据权利要求1所述的生物颗粒,其特征在于,所述灭活细菌为厌氧菌。
  6. 一种生物组合物,其特征在于,含有权利要求1中所述膜囊泡和所述灭活细菌。
  7. 根据权利要求6所述的生物组合物,其特征在于,所述膜囊泡作为疫苗和/或疫苗佐剂和/或药物载体与所述灭活细菌组合。
  8. 从细菌中分离膜囊泡的方法,所述细菌包括革兰氏阳性菌和革兰氏阴性菌,其特征在于,具体包括以下步骤:
    1)将培养细菌的菌液中的菌体和培养液进行分离,收集获得上清液1;
    2)将所述上清液1用高速离心机离心,收集上清液2;
    3)将所述上清液2用超高速离心机离心,沉淀为膜囊泡。
  9. 根据权利要求8所述的方法,其特征在于,所述步骤1)中的分离方法包括离心、过柱或膜包浓缩。
  10. 根据权利要求8所述的方法,其特征在于,在所述步骤2)中收集获得的所述上清液2在进入所述步骤3)之前,先经膜包浓缩。
  11. 根据权利要求8所述的方法,其特征在于,将所述膜囊泡用缓冲液重悬,所述缓冲液按1L体积计算,包括50mM Tris,5mM NaCl,1mM MgSO 4,并且pH值为7.4。
  12. 一种制备生物组合物的方法,其特征在于,收集经权利要求8所述步骤1)分离出的所述菌体,将所述菌体与经权利要求8所述步骤3)所得的所述膜囊泡混合形成所述生物组合物。
  13. 根据权利要求8所述的方法,其特征在于,所述步骤1)中,将所述上清液1用0.3-0.5μM滤器过滤去杂。
  14. 根据权利要求8所述的方法,其特征在于,所述步骤1)中的分离方法为离心,所述离心的离心速率为100-10000g;离心时间 为10-60min。
  15. 根据权利要求8所述的方法,其特征在于,所述步骤2)中的所述高速离心速率为5000-25000g;离心时间为10-100min。
  16. 根据权利要求15所述的方法,其特征在于,所述步骤2)中高速离心速率为10000-20000g;离心时间为30-60min。
  17. 根据权利要求8所述的方法,其特征在于,所述步骤3)中的所述超高速离心速率为5000-150000g;离心时间为60-600min。
  18. 根据权利要求17所述的方法,其特征在于,所述步骤3)中所述的超高速离心速率为15000-150000g;离心时间为60-180min。
  19. 根据权利要求8所述的方法,其特征在于,所述革兰氏阳性菌包括金黄色葡萄球菌、链球菌、白喉杆菌、结核杆菌、破伤风杆菌、芽孢杆菌、棒状杆菌,以及上述菌的基因工程改造菌。
  20. 根据权利要求8所述的方法,其特征在于,所述革兰氏阴性菌包括铜绿假单胞菌、鲍曼不动杆菌、大肠杆菌、伤寒杆菌、脑膜炎双球菌、变形杆菌、志贺菌,以及上述菌的基因工程改造菌。
  21. 根据权利要求8所述的方法,其特征在于,所述细菌为灭活细菌。
  22. 权利要求8-21任一方法所得的膜囊泡。
  23. 制备权利要求1所述的生物颗粒的方法,其特征在于,具体包括以下步骤:
    1)培养细菌至对数生长期;
    2)收集菌体,将所述菌体重悬后用电离射线辐照处理,得辐照细菌;
    3)用权利要求8-21所述的方法进行分离,获得膜囊泡。
  24. 根据权利要求23所述的方法,其特征在于,所述电离射线辐照处理的方式为X-ray射线,辐照剂量为500-3000Gy.
  25. 根据权利要求24所述的方法,其特征在于,所述辐照剂量为500-1000Gy.
  26. 根据权利要求23所述的方法,其特征在于,所述步骤1)中所述对数生长期的细菌的OD 600值为0.3-0.8。
  27. 根据权利要求26所述的方法,其特征在于,所述步骤1)中所述对数生长期的细菌的OD 600值为0.5-0.7。
  28. 根据权利要求23所述的方法,其特征在于,所述步骤1)中用磷酸盐缓冲液或无菌生理盐水重悬所述菌体。
  29. 根据权利要求23所述的方法,其特征在于,所述步骤1)中将所述菌体重悬至OD 600值为20-80。
  30. 根据权利要求29所述的方法,其特征在于,所述步骤1)中将所述菌体重悬至OD 600值为40-60。
  31. 权利要求23-30任一所述的方法所得的膜囊泡。
  32. 根据权利要求31所述的膜囊泡,其特征在于,同未经电离射线辐照的细菌相比,所述膜囊泡中核酸的含量和蛋白质的含量提升了10-20倍。
  33. 权利要求22、31或32所述的膜囊泡作为免疫反应促进剂中的应用。
  34. 权利要求22、31或32所述的膜囊泡作为治疗细菌感染疾病的疫苗中的应用。
  35. 根据权利要求34所述的应用,其特征在于,所述细菌感染疾病包括肺炎、泌尿道感染、脑膜炎、败血症,或皮肤、软组织感染。
  36. 权利要求22、31或32所述的膜囊泡作为疫苗佐剂中的应用。
  37. 根据权利要求34所述的应用,其特征在于,所述疫苗佐剂为非特异性地改变或增强机体对抗原的特异性免疫应答。
  38. 权利要求22、31或32所述的膜囊泡作为抗原递呈细胞功能促进剂中的应用。
  39. 根据权利要求38所述的应用,其特征在于,抗原递呈细胞包括树突状细胞、巨噬细胞及B细胞。
  40. 权利要求31所述的膜囊泡作为DC细胞成熟的促进剂中的应用。
  41. 根据权利要求40所述的应用,其特征在于,所述的膜囊泡作为促进骨髓来源树突状细胞细表面分子CD80、CD86及MHCII分子显著上调的促进剂中的应用。
  42. 权利要求22、31或32所述的膜囊泡作为DC细胞抗原递呈能力促进剂中的应用。
  43. 权利要求22、31或32所述的膜囊泡联合DC细胞在制备CD4+T细胞的增殖剂中的应用。
  44. 促进CD4+T细胞增殖的方法,其特征在于,将权利要求31所述的膜囊泡刺激并吞噬OVA抗原的DCs与CFSE标记的CD4+T淋巴细胞在体外共培养。
  45. 权利要求22、31或32所述的膜囊泡在制备兽药中的应用。
  46. 权利要求6或7所述的生物组合物作为细菌疫苗的应用。
  47. 铜绿假单胞菌疫苗,其特征在于,所述铜绿假单胞菌疫苗含有从灭活铜绿假单胞菌中分离的膜囊泡。
  48. 一种生物组合物,其特征在于,含有如权利要求47中的所述膜囊泡与所述灭活铜绿假单胞菌。
  49. 根据权利要求48所述的生物组合物,其特征在于,所述膜囊 泡作为疫苗和/或疫苗佐剂和/或药物载体与所述灭活铜绿假单胞菌组合。
  50. 从铜绿假单胞菌中分离膜囊泡的方法,其特征在于,包括如下步骤:
    1)培养细菌至对数生长期;
    2)收集菌液,菌液离心并取上清液1,所述上清液1用0.3-0.5μM滤器过滤除菌;
    3)将过滤后的所述上清液1用高速离心机离心,收集上清液2,去除鞭毛;
    4)将去除鞭毛后的所述上清液2用超高速离心机离心,沉淀即得到膜囊泡。
  51. 根据权利要求50所述的方法,其特征在于,步骤2)的上清液用0.45μM滤器过滤除菌。
  52. 根据权利要求50所述的方法,其特征在于,步骤2)的离心速率为100-10000g;离心时间为10-60min.
  53. 根据权利要求50所述的方法,其特征在于,步骤3)的高速离心速率为5000-25000g;离心时间为10-100min。
  54. 根据权利要求50所述的方法,其特征在于,步骤4)的超高速离心速率为5000-150000g;离心时间为60-600min。
  55. 根据权利要求50所述的方法,其特征在于,所述铜绿假单胞菌为灭活的铜绿假单胞菌。
  56. 权利要求50-55所述任一方法得到的铜绿假单胞菌膜囊泡。
  57. 制备权利要求47所述的铜绿假单胞菌疫苗的方法,其特征在于,所述方法包括如下步骤:
    1)培养细菌至对数生长期;
    2)收集菌体,将菌体用适量的磷酸盐缓冲液或无菌生理盐水重悬并用电离射线辐照处理;
    3)收集辐照处理后的菌液,离心并取上清液1,所述上清液1用0.3-0.5μM滤器过滤除菌;
    4)将过滤后的所述上清液1用高速离心机离心,收集上清液2,去除鞭毛;
    5)将去除鞭毛后的所述上清液进行超高速离心,沉淀即得膜囊泡。
  58. 根据权利要求57所述的方法,其特征在于,步骤1)中所述对数生长期的细菌的OD 600值为0.3-0.8。
  59. 根据权利要求57所述的方法,其特征在于,步骤2)中所加磷酸盐缓冲液或无菌生理盐水的量与菌体总量的比例为每1ml溶液含有的菌量为OD 600值20-80。
  60. 根据权利要求57所述的方法,其特征在于,步骤2)中辐照 处理用的射线为X-ray射线;辐照剂量为500-3000Gy。
  61. 根据权利要求57所述的方法,其特征在于,步骤3)中的离心速率为100-10000g;离心时间为10-60min。
  62. 根据权利要求57所述的方法,其特征在于,步骤3)中上清液用0.45μM滤器过滤除菌。
  63. 根据权利要求57所述的方法,其特征在于,步骤4)中所述的高速离心速率为5000-25000g;离心时间为10-100min。
  64. 根据权利要求57所述的方法,其特征在于,步骤5)中所述的超高速离心速率为5000-150000g;离心时间为60-600min。
  65. 权利要求57-64所述任一方法制得的铜绿假单胞菌膜囊泡和疫苗。
  66. 根据权利要求65所述的铜绿假单胞菌膜囊泡,其特征在于,同未经电离射线辐照的细菌膜囊泡相比,所述膜囊泡中核酸的含量和蛋白质的含量提升了10-20倍。
  67. 一种提高铜绿假单胞菌膜囊泡内容物含量的方法,所述内容物包括核酸和蛋白质,其特征在于,所述方法采用辐照设备处理铜绿假单胞菌菌液,所述辐照设备的射线为X-ray射线,辐照剂量为500-1000Gy。
  68. 权利要求56、65或66所述的铜绿假单胞菌膜囊泡在制备抗细菌感染疫苗中的应用,其特征在于,所述铜绿假单胞菌膜囊泡可作为疫苗的佐剂使用。
  69. 根据权利要求68所述的应用,其特征在于,所述疫苗佐剂可以非特异性地改变或增强机体对抗原的特异性免疫应答。
  70. 根据权利要求68所述的应用,其特征在于,所述细菌感染疾病包括肺炎、泌尿道感染、脑膜炎、败血症,或皮肤、软组织感染。
  71. 根据权利要求68所述的应用,其特征在于,所述铜绿假单胞菌膜囊泡还可作为疫苗的载体应用。
  72. 权利要求56、65或66所述的铜绿假单胞菌膜囊泡作为免疫反应促进剂中的应用。
  73. 权利要求56、65或66所述的铜绿假单胞菌膜囊泡作为抗原递呈细胞功能促进剂中的应用。
  74. 根据权利要求73所述的应用,其特征在于,抗原递呈细胞包括树突状细胞、噬细胞及B细胞。
  75. 权利要求65所述的铜绿假单胞菌膜囊泡在制备DC细胞生长促进剂中的应用,其特征在于,所述铜绿假单胞菌膜囊泡能刺激DC细胞表面共刺激分子CD80、CD86和MHCII显著上调,促进 DC细胞成熟和分化。
  76. 权利要求56、65或66所述的铜绿假单胞菌膜囊泡作为DC细胞抗原递呈能力促进剂中的应用。
  77. 一种促进CD4+T细胞增值的方法,其特征在于,将权利要求65所述的铜绿假单胞菌膜囊泡刺激并吞噬OVA抗原的DCs与CFSE标记的CD4+T淋巴细胞在体外共培养。
  78. 权利要求56、65或66所述的铜绿假单胞菌膜囊泡在制备兽药中的应用。
  79. 权利要求48或49所述的生物组合物作为细菌疫苗的应用。
  80. 金黄色葡萄球菌产生的生物颗粒,其特征在于,所述生物颗粒是从灭活金黄色葡萄球菌中分离的膜囊泡。
  81. 一种生物组合物,其特征在于,含有如权利要求80所述的膜囊泡和所述灭活金黄色葡萄球菌。
  82. 根据权利要求81所述的生物组合物,其特征在于,所述膜囊泡作为疫苗和/或疫苗佐剂和/或药物载体与所述灭活金黄色葡萄球菌组合。
  83. 从金黄色葡萄球菌中分离膜囊泡的方法,其特征在于,包括如下步骤:
    1)培养细菌至对数生长期;
    2)收集菌液,菌液离心并取上清液1,所述上清液1用03-0.5μM滤器过滤除菌;
    3)将过滤后的所述上清液1用高速离心机离心,收集上清液2;
    4)将所述上清液2用超高速离心机离心,沉淀即得到膜囊泡。
  84. 根据权利要求83所述的方法,其特征在于,步骤2)的上清液用0.45μM滤器过滤除菌。
  85. 根据权利要求83所述的方法,其特征在于,步骤2)的离心速率为100-10000g;离心时间为10-60min。
  86. 根据权利要求83所述的方法,其特征在于,步骤3)的高速离心速率为5000-25000g;离心时间为10-100min。
  87. 根据权利要求83所述的方法,其特征在于,步骤4)的超高速离心速率为5000-150000g;离心时间为60-600min。
  88. 根据权利要求83所述的方法,其特征在于,所述金黄色葡萄球菌为灭活的金黄色葡萄球菌。
  89. 权利要求83-88所述任一方法得到的金黄色葡萄球菌膜囊泡。
  90. 制备权利要求80所述的生物颗粒的方法,其特征在于,所述方法包括如下步骤:
    1)培养细菌至对数生长期;
    2)收集菌体,将菌体用适量的磷酸盐缓冲液或无菌生理盐水重悬 并用电离射线辐照处理;
    3)收集辐照处理后的菌液,离心并取上清液1,所述上清液1用0.3-0.5μM滤器过滤除菌;
    4)将过滤后的所述上清液1用高速离心机离心,收集上清液2;
    5)将所述上清液2超高速离心,沉淀即得到膜囊泡。
  91. 根据权利要求90所述的方法,其特征在于,步骤1)中所述对数生长期的细菌的OD 600值为0.3-0.8。
  92. 根据权利要求90所述的方法,其特征在于,步骤2)中所加磷酸盐缓冲液或无菌生理盐水的量与菌体总量的比例为每1ml溶液含有的菌量为OD 600值20-80。
  93. 根据权利要求90所述的方法,其特征在于,步骤2)中辐照处理用的射线为X-ray射线;辐照剂量为500-3000Gy。
  94. 根据权利要求90所述的方法,其特征在于,步骤3)中的离心速率为100-10000g;离心时间为10-60min。
  95. 根据权利要求90所述的方法,其特征在于,步骤3)中上清液用0.45μM滤器过滤除菌。
  96. 根据权利要求90所述的方法,其特征在于,步骤4)中所述的高速离心速率为5000-25000g;离心时间为10-100min。
  97. 根据权利要求90所述的方法,其特征在于,步骤5)中所述的超高速离心速率为5000-150000g;离心时间为60-600min。
  98. 权利要求90-97所述任一方法制得的金黄色葡萄球菌膜囊泡。
  99. 一种提高金黄色葡萄球菌膜囊泡内容物含量的方法,所述内容物包括核酸和蛋白质,其特征在于,所述方法采用辐照设备处理金黄色葡萄球菌菌液,所述辐照设备的射线为X-ray射线,辐照剂量为500-1000Gy。
  100. 权利要求89或98所述的金黄色葡萄球菌膜囊泡在制备抗细菌感染疫苗中的应用,其特征在于,所述金黄色葡萄球菌膜囊泡可作为疫苗的佐剂使用。
  101. 根据权利要求100所述的应用,其特征在于,所述疫苗佐剂可以非特异性地改变或增强机体对抗原的特异性免疫应答。
  102. 根据权利要求100所述的应用,其特征在于,所述细菌感染疾病包括肺炎、泌尿道感染、脑膜炎、败血症,或皮肤、软组织感染。
  103. 根据权利要求100所述的应用,其特征在于,所述金黄色葡萄球菌膜囊泡还可作为疫苗的载体应用。
  104. 权利要求89或98所述的金黄色葡萄球菌膜囊泡作为免疫原的应用。
  105. 权利要求89或98所述的金黄色葡萄球菌膜囊泡作为抗原递呈细胞功能促进剂中的应用。
  106. 根据权利要去105所述的应用,其特征在于,抗原递呈细胞包括树突状细胞、噬细胞及B细胞。
  107. 权利要求98所述的金黄色葡萄球菌膜囊泡在制备DC细胞生长促进剂中的应用,其特征在于,所述金黄色葡萄球菌膜囊泡能刺激DC细胞表面共刺激分子CD80、CD86和MHCII显著上调,促进DC细胞成熟和分化。
  108. 权利要求89或98所述的金黄色葡萄球菌膜囊泡作为DC细胞抗原递呈能力促进剂中的应用。
  109. 一种促进CD4+T细胞增值的方法,其特征在于,将权利要求98所述的金黄色葡萄球菌膜囊泡刺激并吞噬OVA抗原的DCs与CFSE标记的CD4+T淋巴细胞在体外共培养。
  110. 权利要求89或98所述的金黄色葡萄球菌膜囊泡在制备兽药中的应用。
  111. 权利要求81或82所述的灭活金黄色葡萄球菌作为细菌疫苗的应用。
  112. 一种细菌膜囊泡的生产系统,其特征在于,所述生产系统依次设有发酵单元1和辐照单元2,所述发酵单元1由细菌发酵罐3和紫外分光光度计4构成,所述辐照单元2由辐照设备5构成。
  113. 根据权利要求112所述的生产系统,其特征在于,所述发酵罐3为通风发酵罐、鼓泡发酵罐、气升式发酵罐或喷嘴环流式发酵罐。
  114. 根据权利要求112所述的生产系统,其特征在于,所述辐照设备5中的射线发射器为X射线发生器、γ射线发生器或Co60同位素发生器。
  115. 一种用于细菌膜囊泡的纯化系统,其特征在于,所述纯化系统依次设有发酵单元1、辐照单元2和分离设备6,所述发酵单元1由细菌发酵罐3和紫外分光光度计4构成,所述辐照单元2由辐照设备5构成。
  116. 如权利要求115所述的纯化系统,其特征在于,所述分离设备6包括离心单元7、膜包单元和/或过柱单元。
  117. 根据权利要求115所述的纯化系统,其特征在于,所述发酵罐3为通风发酵罐、鼓泡发酵罐、气升式发酵罐或喷嘴环流式发酵罐。
  118. 根据权利要求115所述的纯化系统,其特征在于,所述辐照设备5中的射线发射器为X射线发生器、γ射线发生器或Co 60同位素发生器。
  119. 根据权利要求116所述的纯化系统,其特征在于,所述分离设备6包括离心单元7,所述离心单元7为普通离心机、高速离心机 或超高速离心机。
  120. 一种用于细菌膜囊泡的分离系统,其特征在于,所述分离系统依次设有发酵单元1和分离设备6,所述发酵单元1由细菌发酵罐3和紫外分光光度计4构成。
  121. 根据权利要求120所述的分离系统,其特征在于,所述分离设备6包括离心单元7、膜包单元和/或过柱单元。
  122. 根据权利要求121所述的分离系统,其特征在于,所述发酵罐3为通风发酵罐、鼓泡发酵罐、气升式发酵罐或喷嘴环流式发酵罐。
  123. 根据权利要求121所述的分离系统,其特征在于,所述分离设备包括离心单元7,所述离心单元7为普通离心机、高速离心机或超高速离心机。
  124. 采用权利要求112-114任一项所述的生产系统生产细菌膜囊泡的方法,其特征在于,包括如下步骤:
    1)培养细菌至对数生长期;
    2)收集菌体,将菌体用适量的磷酸盐缓冲液或无菌生理盐水重悬;
    3)用电离射线辐照处理。
  125. 根据权利要求124所述的方法,其特征在于,步骤1)中所述对数生长期的细菌的OD 600值为0.3-0.8;步骤2)中所加磷酸盐缓冲液或无菌生理盐水的量与菌体总量的比例为每1ml溶液含有的菌量为OD 600值20-80。
  126. 根据权利要求124所述的方法,步骤3)中辐照处理用的射线为X-ray射线;辐照剂量范围为500-3000Gy。
  127. 采用权利要求115-119任一项所述的纯化系统纯化细菌膜囊泡的方法,其特征在于,包括如下步骤:
    1)培养细菌至对数生长期;
    2)收集菌体,将菌体用适量的磷酸盐缓冲液或无菌生理盐水重悬并用电离射线辐照处理;
    3)收集辐照处理后的菌液,离心并取上清液1,所述上清液1用0.3-0.5μM滤器过滤除菌;
    4)将过滤后的所述上清液1用高速离心机离心,收集上清液2,去除鞭毛;
    5)将去除鞭毛后的所述上清液2超高速离心,沉淀即得到膜囊泡;
    6)收集纯化后的膜囊泡,用缓冲液重悬。
  128. 根据权利要求127所述的方法,其特征在于,步骤1)中所述对数生长期的细菌的OD 600值为0.3-0.8;步骤2)中所加磷酸盐缓冲液或无菌生理盐水的量与菌体总量的比例为每1ml溶液含有的菌量为OD 600值20-80。
  129. 根据权利要求127所述的方法,其特征在于,步骤3)中辐照 处理用的射线为X-ray射线;辐照剂量范围为500-3000Gy。
  130. 根据权利要求127所述的方法,其特征在于,步骤3)中辐照后菌液离心的速率为100-10000g;离心时间为10-60min。
  131. 根据权利要求127所述的方法,其特征在于,步骤4)中高速离心的速率为5000-25000g;离心时间为10-100min。
  132. 根据权利要求127所述的方法,其特征在于,步骤5)中超高速离心的速率为5000-150000g;离心时间为60-600min。
PCT/CN2019/106654 2019-08-22 2019-09-19 细菌膜囊泡及其分离制备系统和方法 WO2021031270A1 (zh)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US17/637,051 US20220378902A1 (en) 2019-08-22 2019-09-19 Bacterial membrane vesicles, and separation and preparation system and method therefor

Applications Claiming Priority (10)

Application Number Priority Date Filing Date Title
CN201910777473.5A CN112402601B (zh) 2019-08-22 2019-08-22 金黄色葡萄球菌膜囊泡及其制备方法与应用
CN201921369450.2U CN211394490U (zh) 2019-08-22 2019-08-22 一种细菌膜囊泡的生产系统、分离系统和纯化系统
CN201910777473.5 2019-08-22
CN201921369450.2 2019-08-22
CN201910777595.4A CN112410212B (zh) 2019-08-22 2019-08-22 一种细菌膜囊泡的生产系统和分离纯化系统及方法
CN201910777479.2 2019-08-22
CN201910777606.9A CN112410240B (zh) 2019-08-22 2019-08-22 铜绿假单胞菌膜囊泡及其制备方法与应用
CN201910777479.2A CN112410239B (zh) 2019-08-22 2019-08-22 细菌膜囊泡及其制备方法与应用
CN201910777606.9 2019-08-22
CN201910777595.4 2019-08-22

Publications (1)

Publication Number Publication Date
WO2021031270A1 true WO2021031270A1 (zh) 2021-02-25

Family

ID=74659947

Family Applications (3)

Application Number Title Priority Date Filing Date
PCT/CN2019/106654 WO2021031270A1 (zh) 2019-08-22 2019-09-19 细菌膜囊泡及其分离制备系统和方法
PCT/CN2019/118479 WO2021031409A1 (zh) 2019-08-22 2019-11-14 铜绿假单胞菌疫苗在呼吸系统疾病中的应用
PCT/CN2020/110383 WO2021032179A1 (zh) 2019-08-22 2020-08-21 铜绿假单胞菌疫苗在抗烧烫伤感染中的应用

Family Applications After (2)

Application Number Title Priority Date Filing Date
PCT/CN2019/118479 WO2021031409A1 (zh) 2019-08-22 2019-11-14 铜绿假单胞菌疫苗在呼吸系统疾病中的应用
PCT/CN2020/110383 WO2021032179A1 (zh) 2019-08-22 2020-08-21 铜绿假单胞菌疫苗在抗烧烫伤感染中的应用

Country Status (3)

Country Link
US (3) US20220378902A1 (zh)
CN (1) CN114364787B (zh)
WO (3) WO2021031270A1 (zh)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20220378902A1 (en) * 2019-08-22 2022-12-01 Sichuan University Bacterial membrane vesicles, and separation and preparation system and method therefor

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1688334A (zh) * 2002-08-30 2005-10-26 启龙有限公司 改进的细菌外膜囊泡
WO2008147816A2 (en) * 2007-05-22 2008-12-04 Cornell Research Foundation, Inc. Compositions and methods for the display of proteins on the surface of bacteria and their derived vesicles and uses thereof
WO2012097185A2 (en) * 2011-01-12 2012-07-19 The Administrators Of The Tulane Educational Fund Omv vaccine against burkholderia infections
WO2015144691A1 (en) * 2014-03-26 2015-10-01 Glaxosmithkline Biologicals Sa Compositions for immunising against staphylococcus aureus

Family Cites Families (39)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE2152112C3 (de) * 1970-10-20 1979-10-11 Yuzuru Homma Impfstoff gegen Pseudomonas aeruginosa
RU2155226C2 (ru) * 1993-06-07 2000-08-27 Чейл Фудз энд Кемикалз, Инк. Новый ослабленный штамм pseudomonas aeruginosa
EP0841944A2 (en) * 1995-08-04 1998-05-20 University Of Guelph Novel vaccines and pharmaceutical compositions using membrane vesicles of microorganisms, and methods for preparing same
US20020028215A1 (en) * 1999-08-09 2002-03-07 Jagath L. Kadurugamuwa Novel vaccines and pharmaceutical compositions using membrane vesicles of microorganisms, and methods for preparing same
CA2318536C (en) * 1998-11-25 2009-11-17 Mcw Research Foundation, Inc. Method of and compositions for immunization with the pseudomonas v antigen
GB9918319D0 (en) * 1999-08-03 1999-10-06 Smithkline Beecham Biolog Vaccine composition
CU23313A1 (es) * 2002-11-27 2008-09-25 Inst Finlay Ct De Investigacia Mã0/00todo de obtenciã"n de estructuras cocleares. composiciones vacunales y adyuvantes basados en estructuras cocleares y sus intermediarios
US7592326B2 (en) * 2004-03-15 2009-09-22 Karaolis David K R Method for stimulating the immune, inflammatory or neuroprotective response
CN102343086A (zh) * 2010-05-28 2012-02-08 四川大学 治疗或预防肿瘤的药物、肿瘤全细胞疫苗及其制备方法和用途
EP2897635A1 (en) * 2012-09-18 2015-07-29 Novartis AG Outer membrane vesicles
CN105163724A (zh) * 2013-02-07 2015-12-16 葛兰素史密丝克莱恩生物有限公司 包含囊泡的药物组合物
CN104338128B (zh) * 2013-07-31 2017-09-05 普莱柯生物工程股份有限公司 一种疫苗组合物及其制备方法和应用
CN104189898B (zh) * 2014-06-27 2017-09-08 四川大学 铜绿假单胞菌疫苗及其制备方法
CN105647841A (zh) * 2014-09-04 2016-06-08 苏静 铜绿假单胞菌突变株构建方法及其应用
KR101632962B1 (ko) * 2015-07-29 2016-06-23 한국생명공학연구원 변형 외막소포체를 포함하는 항감염성 면역증강용 조성물
CN105713916B (zh) * 2016-02-15 2018-08-24 重庆医科大学附属第二医院 一种铜绿假单胞菌基因及其dna疫苗
JP6854530B2 (ja) * 2016-02-16 2021-04-07 プレジデント アンド フェローズ オブ ハーバード カレッジ 病原体ワクチンならびにその製造および使用方法
CN105732818B (zh) * 2016-03-02 2019-06-07 中国人民解放军第三军医大学 一种铜绿假单胞菌重组蛋白pop及其制备方法和应用
CN105754978B (zh) * 2016-03-02 2018-12-11 中国人民解放军第三军医大学 一种铜绿假单胞菌重组蛋白Vac14及制备方法和应用
CN105709218A (zh) * 2016-03-18 2016-06-29 辽宁成大生物股份有限公司 一种奇异变形杆菌-金黄色葡萄球菌-铜绿假单胞菌吸附联合疫苗的制备方法
MX2019006104A (es) * 2016-11-25 2019-08-21 Glaxosmithkline Biologicals Sa Conjugados inmunogenicos y uso de los mismos.
CN107137427B (zh) * 2017-06-22 2020-09-04 四川大学华西医院 铜绿假单胞菌的新用途
CN107397956A (zh) * 2017-08-08 2017-11-28 南开大学 一种铜绿假单胞菌外膜蛋白疫苗的制备方法及应用
WO2019178487A2 (en) * 2018-03-15 2019-09-19 Evelo Biosciences, Inc. Compositions and methods for treating disease using klebsiella quasipneumoniae subsp. similipneumoniae
CN108743931B (zh) * 2018-05-02 2022-08-16 成都威斯克生物医药有限公司 抗结核病疫苗及其制备方法和用途
US10953081B2 (en) * 2018-10-03 2021-03-23 The Administrators Of The Tulance Educational Fund Methods and compositions for inducing an immune response to Pseudomonas aeruginosa pulmonary infections
US20200281969A1 (en) * 2019-03-05 2020-09-10 Wonder Spray, LLC Inhibiting viral and bacterial activity using low concentration hypochlorous acid solutions
CN110028559B (zh) * 2019-04-15 2022-06-10 中国人民解放军陆军军医大学 一种铜绿假单胞菌疫苗重组蛋白及其编码基因和它们的应用
US20220378902A1 (en) * 2019-08-22 2022-12-01 Sichuan University Bacterial membrane vesicles, and separation and preparation system and method therefor
CN112402601B (zh) * 2019-08-22 2022-09-13 四川大学 金黄色葡萄球菌膜囊泡及其制备方法与应用
CN114364396B (zh) * 2019-08-22 2024-01-23 四川大学 铜绿假单胞菌疫苗在抗烧烫伤感染中的应用
CN112410239B (zh) * 2019-08-22 2023-03-24 四川大学 细菌膜囊泡及其制备方法与应用
CN112410240B (zh) * 2019-08-22 2022-10-18 四川大学 铜绿假单胞菌膜囊泡及其制备方法与应用
US20230124862A1 (en) * 2020-03-24 2023-04-20 Jeffrey W. MILLARD Bismuth thiol compounds and compositions and methods of treating microbial co-infections
CN111701015A (zh) * 2020-07-19 2020-09-25 北京易思腾翻译服务有限公司 可防治新冠病毒感染等呼吸系统疾病和其他疾病的组合物
CN112755180B (zh) * 2020-12-30 2022-01-28 中国医学科学院医学生物学研究所 一种通用的细菌疫苗的制备方法和应用
CN112646046B (zh) * 2021-01-09 2023-03-28 中国人民解放军陆军军医大学 用于预防铜绿假单胞菌感染的多表位融合蛋白及其编码基因、表达载体和应用
WO2023142201A1 (zh) * 2022-01-29 2023-08-03 成都威斯克生物医药有限公司 铜绿假单胞菌疫苗的工业化生产方法
CN115161377A (zh) * 2022-08-26 2022-10-11 四川大学华西医院 一种耐碳青霉烯铜绿假单胞菌的鉴定培养基及其制备方法和应用

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1688334A (zh) * 2002-08-30 2005-10-26 启龙有限公司 改进的细菌外膜囊泡
WO2008147816A2 (en) * 2007-05-22 2008-12-04 Cornell Research Foundation, Inc. Compositions and methods for the display of proteins on the surface of bacteria and their derived vesicles and uses thereof
WO2012097185A2 (en) * 2011-01-12 2012-07-19 The Administrators Of The Tulane Educational Fund Omv vaccine against burkholderia infections
WO2015144691A1 (en) * 2014-03-26 2015-10-01 Glaxosmithkline Biologicals Sa Compositions for immunising against staphylococcus aureus

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
CAI WEI, KESAVAN DINESH KUMAR, WAN JIE, ABDELAZIZ MOHAMED HAMED, SU ZHAOLIANG, XU HUAXI: "Bacterial outer membrane vesicles, a potential vaccine candidate in interactions with host cells based", DIAGNOSTIC PATHOLOGY, vol. 13, 95, 11 December 2018 (2018-12-11), pages 1 - 12, XP055782531, DOI: 10.1186/s13000-018-0768-y *
GAMALIER JULIANA P; SILVA THIAGO P; ZARANTONELLO VICTOR; DIAS FELIPE F; MELO ROSSANA C N: "Increased production of outer membrane vesicles by cultured freshwater bacteria in response to ultraviolet radiation", MICROBIOLOGICAL RESEARCH, vol. 194, 3 November 2016 (2016-11-03), pages 38 - 46, XP029837118, ISSN: 0944-5013, DOI: 10.1016/j.micres.2016.08.002 *
JIE YI; QING LIU; QINGKE KONG: "Advances in Outer Membrane Vesicles of Gram-Negative Bacteria as Sub-Unit Vaccines-A Review", ACTA MICROBIOLOGICA SINICA, vol. 56, no. 6, 11 January 2016 (2016-01-11), pages 911 - 921, XP009526240, ISSN: 0001-6209, DOI: 10.13343/j.cnki.wsxb.20150377 *

Also Published As

Publication number Publication date
WO2021031409A1 (zh) 2021-02-25
WO2021032179A1 (zh) 2021-02-25
US20220378902A1 (en) 2022-12-01
US20220378901A1 (en) 2022-12-01
CN114364787B (zh) 2024-03-01
CN114364787A (zh) 2022-04-15
US20220370588A1 (en) 2022-11-24

Similar Documents

Publication Publication Date Title
CN112410239B (zh) 细菌膜囊泡及其制备方法与应用
CN112402601B (zh) 金黄色葡萄球菌膜囊泡及其制备方法与应用
JP4243349B2 (ja) インフルエンザウイルスの複製のための動物細胞および方法
US6140122A (en) Process for preparing macrophages, kits, and compositions for the use of this process
CN112410240B (zh) 铜绿假单胞菌膜囊泡及其制备方法与应用
WO1996023060A1 (en) Method for isolating dendritic cells
WO2014021070A1 (ja) 単球またはnk細胞を入手する方法
EP2491115B1 (en) Cell populations having immunoregulatory activity, methods for the preparation and uses thereof
CN112608896A (zh) 一种nk细胞的培养方法及其应用
WO2021031270A1 (zh) 细菌膜囊泡及其分离制备系统和方法
JP2002045174A (ja) ナチュラルキラー細胞増殖法
Miyake Establishment of a human oligodendroglial cell line
JP2015504423A (ja) 腫瘍免疫療法のためのワクチン
Waner et al. Behavior of human cytomegaloviruses in cell cultures of bovine and simian origin
CN115094034B (zh) 一种人nkt细胞系及其应用
CN114990075B (zh) 一株可适用于人用疫苗细胞基质培养的柯萨奇病毒a组10型疫苗株及其应用
US3616203A (en) Virus culture and method
CN114364396B (zh) 铜绿假单胞菌疫苗在抗烧烫伤感染中的应用
DE60130206T2 (de) Zusammensetzungen und verfahren zur herstellung von antigen-präsentierenden zellen
CN107841486A (zh) 树突状细胞子集cd103+dc的体外培养方法及鉴定方法
CN113546162A (zh) 一种支原体疫苗及其制备方法
NL2031523B1 (en) Cell culture method of chlamydia abortus and use thereof
US20040152191A1 (en) Process for the maturation of dendritic cells and for the activation of macrophages with RU 41740
CN112410212B (zh) 一种细菌膜囊泡的生产系统和分离纯化系统及方法
WO2023208212A1 (zh) 基于三维多孔微载体的病毒载体生产方法

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 19942289

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 19942289

Country of ref document: EP

Kind code of ref document: A1