CN114560947A - Fusion protein for inhibiting angiogenesis - Google Patents

Fusion protein for inhibiting angiogenesis Download PDF

Info

Publication number
CN114560947A
CN114560947A CN202011366707.6A CN202011366707A CN114560947A CN 114560947 A CN114560947 A CN 114560947A CN 202011366707 A CN202011366707 A CN 202011366707A CN 114560947 A CN114560947 A CN 114560947A
Authority
CN
China
Prior art keywords
fusion protein
ala
cells
glu
lys
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
CN202011366707.6A
Other languages
Chinese (zh)
Inventor
李红玉
李婷
李洋
王欣
王梅竹
段建功
白璐
史彦斌
郭玎玎
王如玥
张涵姿
刘怡瑶
张倩
侯迎港
李文博
夏润洁
黎凯科
宋美娟
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Lanzhou University
Original Assignee
Lanzhou University
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Lanzhou University filed Critical Lanzhou University
Priority to CN202011366707.6A priority Critical patent/CN114560947A/en
Publication of CN114560947A publication Critical patent/CN114560947A/en
Pending legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/78Connective tissue peptides, e.g. collagen, elastin, laminin, fibronectin, vitronectin or cold insoluble globulin [CIG]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • 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
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/80Vectors or expression systems specially adapted for eukaryotic hosts for fungi
    • C12N15/81Vectors or expression systems specially adapted for eukaryotic hosts for fungi for yeasts
    • C12N15/815Vectors or expression systems specially adapted for eukaryotic hosts for fungi for yeasts for yeasts other than Saccharomyces
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/31Fusion polypeptide fusions, other than Fc, for prolonged plasma life, e.g. albumin
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Genetics & Genomics (AREA)
  • Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Zoology (AREA)
  • Biotechnology (AREA)
  • Biophysics (AREA)
  • Wood Science & Technology (AREA)
  • Medicinal Chemistry (AREA)
  • Molecular Biology (AREA)
  • Biochemistry (AREA)
  • Mycology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Engineering & Computer Science (AREA)
  • Biomedical Technology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • General Chemical & Material Sciences (AREA)
  • Plant Pathology (AREA)
  • Physics & Mathematics (AREA)
  • Microbiology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Toxicology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Peptides Or Proteins (AREA)

Abstract

The invention discloses a fusion protein for inhibiting angiogenesis, belongs to the field of genetic engineering pharmacy, and particularly relates to a fusion protein capable of being used as an angiogenesis inhibitor. The fusion protein has a long-acting effect of inhibiting angiogenesis, and the technical scheme disclosed by the invention improves the anti-angiogenesis activity of the angiogenesis endostatin and the inhibition effect on melanoma on one hand, prolongs the half-life period of the angiogenesis endostatin and further enhances the stability on the other hand.

Description

Fusion protein for inhibiting angiogenesis
Technical Field
The invention belongs to the field of biotechnology pharmacy, and particularly relates to a fusion protein with angiogenesis inhibiting activity, which can be applied to the field of antitumor drugs.
Background
According to the latest national research report in 2018, the number of new cases of malignant tumors in China is 380.4 ten thousands, and the number of cancer deaths in China is 229.6 thousands, so that the malignant tumors are one of the main diseases which seriously affect the health of human beings and threaten the life of human beings at present, and form three causes of death in all countries in the world together with cardiovascular and cerebrovascular diseases and accidents. With the continuous development of socioeconomic, the prevalence rate of tumors is increasing year by year, so that the world health organization and the government health departments of all countries take cancer attack as a first task.
The current means for treating tumors comprise physical therapy (radiotherapy and surgical removal) and chemical therapy (anti-tumor drugs), wherein the physical therapy has large damage to the body of a patient, and the chemical therapy has serious side effects including nausea, vomiting, hair loss and the like in most patients, and the patients are easy to have drug resistance. All the above methods cannot completely cure the tumor because tumor cells often have infiltrative and metastatic properties, which brings difficulty to treatment.
The growth and the metastasis of primary tumors are completed depending on new blood vessels, the blood vessels of the tumors can provide nutrition for the growth of the tumors and mediate tumor cell metastasis through the blood vessels, the blood vessels of the tumors play an important role in the whole growth stage of the tumors, and if the formation of the tumor new blood vessels can be inhibited, the nutrition supply channels of the tumor cells and the metastasis channels of the tumor cells can be blocked, and the defect that the tumors are difficult to treat due to the metastatic properties of the tumors is overcome, so that the treatment method for the tumor angiogenesis is paid attention in recent years, and the good application prospect is shown.
It has been found that benign tumors have rare angiogenesis and slow growth of blood vessels, while most malignant tumors have dense angiogenesis and rapid growth, indicating that the number of tumor blood vessels is closely related to the development degree of tumors. The formation of new blood vessels depends mainly on the proliferation, migration and lumen formation of vascular endothelial cells, which is the main process of angiogenesis and is the result of the coordination of pro-angiogenic factors and inhibitory angiogenic factors in the body. The specific process of tumor angiogenesis is as follows: tumor cells release angiogenesis promoting factors (such as VEGF, bFGF and the like), activate angiogenesis promoting factor receptors on vascular endothelial cells, activate the endothelial cells and increase vascular permeability; secondly, the activated endothelial cells release protease to degrade basement membrane; endothelial cells proliferate and migrate to form a surrounding matrix and a sprouting mechanism; the bud-shaped mechanism is further expanded to be annular, and finally a blood vessel cavity is formed; the blood vessel cavity matures and stabilizes to form new blood vessel branch. The overall angiogenic process is highly ordered. According to the process of tumor neovascularization, Judah Folkman firstly proposes the importance of angiogenesis on the growth and metastasis of tumors in 1971, and the theory of tumor angiogenesis is continuously proved and enriched so far, researches show that a large amount of oxygen and nutrients are needed for the growth of tumors, the channel for providing the nutrients for the tumor cells is blood vessels, the tumor blood vessels can transport harmful metabolites of the tumor cells out to stimulate the growth of the tumors, and when the tumors grow to a certain stage, the tumors can be metastasized through the blood vessels connected with the tumor blood vessels. The invasion and metastasis of tumors are difficult points in tumor treatment, and the inhibition of tumor angiogenesis can effectively inhibit the transportation and metastasis channels of nutrients required by tumor growth, so that the mode of inhibiting tumors by taking tumor neovascularization as a treatment target is the main direction of current research. At present, there are many drugs for inhibiting tumor angiogenesis, including drugs for inhibiting degradation of basement membrane, drugs for inhibiting endothelial cells (drugs for directly inhibiting endothelial cells and drugs for inhibiting specific integrin on vascular endothelial cells), and drugs for vascular growth factors. The medicine for inhibiting endothelial cells comprises 2 medicines, namely directly inhibiting the growth of the endothelial cells, such as endostatin; the second is a drug for inhibiting specific integrin on endothelial cells, which can act on different ligands of the integrin, block subsequent channels opened by the integrin, inhibit proliferation and migration of the endothelial cells, and inhibit tumor angiogenesis. At present, the most developed drugs taking the angiogenesis promoting factor as a target point can inhibit the signal of a receptor of the angiogenesis promoting factor and block the opening of a subsequent signal path and the secretion of the angiogenesis promoting factor, so that the aim of inhibiting tumors is fulfilled.
Compared with the traditional anticancer drugs, the drug for inhibiting the tumor angiogenesis has the following advantages: firstly, the growth and the metastasis of solid tumors depend on tumor blood vessels, so that the angiogenesis targeted medicine has broad spectrum and can play a role in various solid tumors; secondly, the medicine for partially inhibiting the generation of tumor angiogenesis acts on endothelial cells, and compared with cancer cells, the endothelial cells have the advantage of low mutation rate and low probability of drug resistance of organisms; thirdly, the medicine can directly reach the target site through blood transportation, and has quick effect taking time and less toxic and side effects.
In the process of angiogenesis, vascular endothelial cell adhesion molecules are important substances and can adhere various cell components together to accelerate the generation and stabilization of new blood vessels. The integrin family belongs to one of vascular endothelial cell adhesion molecules, is transmembrane protein, can mediate adhesion of cells and extracellular matrix, and plays an important role in the processes of tumor angiogenesis and tumor metastasis. Integrin is composed of two subunits, alpha and beta, and there are currently reported 18 alpha subunits and 8 beta subunits, with different subunits being capable of composing 24 different integrins. Integrins regulate the bidirectional signaling pathway of cells, recognize and bind to extracellular ligands, open intracellular signaling pathways, and mediate adhesion and migration of endothelial and tumor cells, e.g., integrins can participate in MAPK signaling pathways, regulate VEGF expression, and promote VEGF binding to its receptors. At present, α v β 3 and α v β 5 are two types of integrins which are researched more, and reports about participation of the two types of integrins in tumor deterioration are numerous, on one hand, the integrins can regulate and control tumor cells to express matrix metalloproteinase, accelerate degradation of extracellular matrix, promote angiogenesis, on the other hand, the integrins can promote the tumors to secrete adhesion molecules, and the tumor cells can diffuse through the adhesion molecules to realize metastasis of the tumor cells. Therefore, the integrin plays an important role in tumor angiogenesis and the growth and metastasis of tumor cells, and becomes one of the important targets for treating tumors.
The alpha v beta 3 is expressed in various cells, but the alpha v beta 3 is only highly expressed on the surfaces of neovascular endothelial cells and tumor cells, and is low expressed on the surfaces of quiescent endothelial cells and normal cells, so that the integrin alpha v beta 3 can be used as an important target point for resisting angiogenesis. α v β 3 recognizes the Arg-Gly-Asp (RGD) sequence in the ligand, so RGD can be used as a targeting peptide for targeting integrin in vivo. However, the RGD sequence consists of only 3 amino acids and is easily degraded by proteases in vivo if present alone. ZL2005100403785 introduces a fusion peptide HM-3 which is an integrin blocker and has the amino acid sequence as follows: Ile-Val-Arg-Arg-Ala-Asp-Arg-Ala-Ala-Val-Pro-Gly-Gly-Gly-Gly-Arg-Gly-Asp. HM-3 is composed of 2 parts, endostatin active peptide ED (Ile-Val-Arg-Arg-Ala-Ala-Asp-Arg-Ala-Ala-Val-Pro) and integrin ligand sequence (Arg-Gly-Asp) are connected by linker (Gly-Gly-Gly-Gly), and the structural formulas are ED-L-RGD and RGD-L-ED. The active peptide ED of endostatin is a 60 th-70 th amino acid segment of endostatin, and researches show that the segment has the activity of inhibiting angiogenesis in vitro, but has lower in vivo activity and is possibly related to shorter half life of small peptide; HM-3 combines the two sequences, and researches show that the fusion small peptide has good anti-angiogenesis effect, not only retains the anti-angiogenesis activity of ED, but also can play the targeting role of RGD. Research proves that HM-3 has the action targets of alpha v beta 3 and alpha 5 beta 1, mainly takes alpha v beta 3 as the main target, and in vivo and in vitro experiments prove that the small peptide can effectively inhibit the migration and adhesion of vascular endothelial cells and the generation of tumor vessels, and has better tumor inhibition effect. Currently, HM-3 has begun clinical trials in China (accession number: CTR20150368), and the intended clinical medication is daily intravenous drip.
The daily injection administration not only brings great pain to patients, but also increases the treatment cost, because HM-3 belongs to short peptide and has short half-life period, in order to solve the problem, patent 201110370529.9 discloses a method for modifying HM-3 by polyethylene glycol, and obtains HM-3 with long half-life period, but the PEGylated small peptide belongs to chemical synthesis products, inevitably has the defects of uneven products and the like, can not strictly control the quality of the products, and brings certain difficulty to clinic. The Beijing Saiyi pharmaceutical industry of 2018 discloses a long-acting HM-3, and the technology is to perform fatty acid modification and maleimide modification on the HM-3, so that the anti-tumor effect of the HM-3 is maintained, and the half-life period of the HM-3 is prolonged. Researches show that polyethylene glycol modification, fatty acid modification or maleimide modification belong to chemical modification, the operation is complex, the defect of non-uniform products exists, biological expression cannot be carried out, and the cost is reduced.
Under the circumstance that chemical modification brings a series of problems, in order to prolong the half-life of a drug, a better drug modification method is sought, and in recent years, fusion protein technology is becoming more mature, wherein the Fc fusion protein drug and HSA related fusion protein technology are developed more rapidly, and HSA has slight targeting compared with Fc and can realize monomer fusion, so that great attention is paid to researchers. Research finds that HSA related fusion protein has larger molecular weight because HSA is a macromolecular long peptide, the molecular weight of the HSA related fusion protein is greatly increased when the HSA is connected with a medicament, and the clearance efficiency of the kidney of an organism to the macromolecular medicament is low, so that the clearance efficiency of the organism to the related fusion protein medicament is reduced, and the half-life period of the medicament is prolonged; furthermore, HSA belongs to endogenous molecules, and when being combined with drugs, HSA can play a certain role in modifying the structure of the small-molecule polypeptide drugs, weaken the immunological rejection of organisms to exogenous small-molecule polypeptide drugs, and weaken the cytotoxic effect of the organisms caused by the exogenous small-molecule polypeptide drugs while delaying the elimination of the drugs.
Although much work has been done by researchers, only 2 HSA fusion protein drugs are on the market to date. Admittedly, the half-life of small molecule polypeptide drugs can be prolonged to a certain extent by means of fusion proteins, but what is not satisfactory is that after small peptides are prepared into fusion proteins, the drug effect of specific tumors is not as good as that of short peptides used alone, so that how to increase the expression efficiency of the fusion proteins and enhance the drug effect of the fusion proteins is the key point for improving the drug effect of the fusion proteins in the process of fusing the short peptides with the target.
Disclosure of Invention
The invention constructs the fusion protein of the angiogenesis endostatin HM-3, not only solves the problems of reduced drug effect and non-ideal expression quantity caused by adopting HSA fusion strategy in the prior art, but also improves the problem of low activity of the existing angiogenesis endostatin. The inventor also unexpectedly finds that although the small molecule polypeptide HM-3 has no proliferation inhibition effect on the melanoma cell B16F10 in vitro, the HM-3 fusion protein can obviously inhibit the proliferation of the melanoma cell B16F10 in vitro. In order to solve the problems, the technical scheme adopted by the invention is as follows:
the invention provides a fusion protein, wherein the amino acid sequence of the fusion protein is shown as SEQ ID NO:1, and the DNA sequence for coding the amino acid sequence is shown as SEQ ID NO: 2, respectively.
The fusion protein is prepared by yeast cell expression.
The yeast is Pichia methanolica (Pichia pastoris).
The preparation method of the fusion protein comprises the following steps:
synthesizing DNA of fusion protein by using whole gene;
secondly, connecting the target segment with a vector by a genetic engineering technology to obtain a recombinant yeast expression vector containing a DNA sequence for coding the fusion protein;
and thirdly, the recombinant yeast expression vector in the step two is transformed into an escherichia coli competence, and after the sequencing is successful, the plasmid is transformed into a host expression system for expression to obtain the fusion protein, wherein the host expression system is pichia methanolica.
Another object of the present invention is to provide a recombinant expression vector containing a DNA sequence encoding the fusion protein.
Another object of the present invention is to provide a host expression system comprising the recombinant expression vector.
The invention also aims to provide application of the fusion protein in preparing anti-tumor drugs.
Preferably, the fusion protein is used for preparing antitumor drugs, and particularly can be applied to preparing drugs for inhibiting solid tumor cells such as melanoma cells, liver cancer cells, lung cancer cells, pancreatic cancer cells, cervical cancer cells and the like.
The dosage form of the anti-tumor medicine is injection, capsule, tablet, pill, nasal spray or aerosol. Compared with the prior art, the invention has the beneficial effects that:
(1) the invention discovers an angiogenesis endostatin fusion protein with more obvious tumor proliferation inhibition activity;
(2) the yeast expression system is adopted, the yeast expression system can process the protein, most of the protein can form a correct high-level structure, the yield of the yeast expression protein is high, the purity is high, the purification is simple, and the host expression efficiency is improved;
(3) the fusion protein in vitro experiment proves that the fusion protein has selective inhibition effect on the proliferation of tumor cells.
(4) The fusion protein in vitro experiment proves that the fusion protein has small influence on normal cells.
(5) The fusion protein of the invention is proved to have the inhibition effect on the migration of tumor cells by in vitro experiments.
Drawings
FIG. 1 map of vector pPink alpha-HC
FIG. 2 map of vector pPink alpha-HC/Fusion protein gene
FIG. 3SDS-PAGE electrophoresis to detect protein expression
FIG. 4 SDS-PAGE of purified fusion proteins
FIG. 5 shows the inhibition of tumor cell proliferation by the fusion protein, CK represents blank control
FIG. 6 inhibition of Normal cell proliferation by fusion protein, CK stands for blank control
FIG. 7 Tanswell results for the fusion protein, CK stands for blank control
Detailed Description
Example 1
Construction of pPINK alpha-HC/fusion protein vector
The fusion protein gene segment is synthesized by the whole gene, the amino acid sequence of the fusion protein is shown as SEQ ID NO 1, and the DNA sequence for coding the amino acid sequence is shown as SEQ ID NO: 2 is shown in the specification;
carrying out double enzyme digestion on pPINK alpha-HC (product of Invitrogen) plasmid and fusion protein fragment by KpnI and StuI, carrying out gel recovery to obtain pPINK alpha-HC (Kpn I/Stu I) vector and protein fragment, carrying out recombination reaction on the pPINK alpha-HC (Kpn I/Stu I) vector fragment and fusion protein gene fragment obtained by gel recovery to obtain a recombinant yeast expression vector containing a DNA sequence for coding the fusion protein. Coli competent TOP10 was transformed, plated on ampicillin-resistant LB plates and cultured overnight at 37 ℃ to select positive clones. The obtained clone is sent to a company for sequencing;
secondly, digesting and recovering the pPINK alpha-HC/fusion protein plasmid DNA with correct sequencing by Afl II to obtain a linear recombinant plasmid fragment, transforming pichia methanolica, inoculating the transformed bacterial liquid to a PAD plate, culturing for 5-7 days at 30 ℃, and selecting positive clones. And respectively inoculating the obtained positive clones to a BMGY liquid culture medium, culturing at 30 ℃ for 48 hours, then transferring to a BMMY culture medium for induced expression, continuously centrifuging at 8000rpm for 30 minutes after 96 hours, taking supernatant, and detecting the protein expression condition by SDS-PAGE electrophoresis.
As shown in FIG. 3, the fusion protein has a molecular weight of about 68kDa, a high expression level and a high purity.
EXAMPLE 2 purification of fusion proteins
According to the nature of the fusion protein, DEAE Sepharose Fast Flow was first selected for purification, and the experimental procedure was as follows:
firstly, sample pretreatment: the expression supernatant was dialyzed against 20mM PB buffer; column balancing: loading DEAE Sepharose fast flow purified packing into a purification column, and balancing with 20mM PB buffer; ③ sample loading: loading a proper amount of sample, collecting a flow-through sample, and naming LC; fourthly, washing the filler by using a proper amount of 20mM PB buffer, and collecting flow liquid which is named as W; fifth, elution: eluting with 0.1M NaCl, 0.3M NaCl, 0.5M NaCl and 1M NaCl, respectively, and collecting eluates named as E1, E2, E3 and E4; sixthly, washing the filler by using 0.1M NaOH, collecting the flow-through liquid and naming the liquid as N; seventhly, washing the filler with a large amount of distilled water, and preserving the filler with 20% ethanol. Protein samples were analyzed by SDS-PAGE.
Phenyl Sepharose Fast Flow was selected for subsequent purification, the experimental procedure was as follows:
firstly, sample pretreatment: the sample was E1 eluate collected after DEAE Sepharose Fast Flow purification, and ammonium sulfate was added to make the sample contain 1.5M (NH)4)2SO4
Column balancing: phenyl Sepharose Fast Flow purification packing was loaded onto a purification column using 1.5M (NH)4)2SO4+0.1M NaCl buffer for equilibration;
③ sample loading: loading a proper amount of sample, collecting a flow-through sample, and naming LC;
fourthly, using proper amount of 1.5M (NH)4)2SO4+0.1M NaCl buffer washes the packing, collects the flow-through fluid, named W;
fifth, elution: respectively using 1.0M (NH)4)2SO4,0.8M(NH4)2SO4,0.6M(NH4)2SO4,0.5M(NH4)2SO4,0.3M(NH4)2SO4,0.1M(NH4)2SO4,H2Eluting O, and collecting eluates named as E1, E2, E3, E4, E5, E6 and E7;
sixthly, washing the filler by using 0.1M NaOH, collecting the flow-through liquid and naming the liquid as N;
seventhly, washing the filler with a large amount of distilled water, and preserving the filler with 20% ethanol. Protein samples were analyzed by SDS-PAGE. The 10kDa concentration tube was used to concentrate the protein sample and the protein concentration was determined by Lowry method.
The results are shown in fig. 4, obtaining fusion proteins with a purity > 90%.
Example 3 inhibition of tumor cells by fusion proteins
HM-3 was synthesized by Shanghai Gill polypeptide Ltd (HM-3 amino acid sequence: IVRRADRAAVPGGGGRGD), and was more than 95% pure.
MTT method is adopted to detect the fusion protein pairs B16F10 (mouse melanoma cells), SMMC-7721 (human hepatoma cells), A549 (human lung cancer cells), Hela (human cervical cancer cells) and PANC-1 (human pancreatic cancer cells)Cell) proliferation inhibition of tumor cells. Tumor cells were treated at 37 ℃ with 5% CO2The cells were collected by trypsinization when cultured in the incubator (2) to a density of 90% or more, and the cells were resuspended in the culture medium and counted under a microscope to adjust the cell concentration to 3.0X 104Cell suspension was seeded into 96-well plates at 100. mu.L per well at 37 ℃ in 5% CO2The culture was carried out overnight in an incubator. The fusion proteins were diluted with PBS to each of the predetermined concentrations. After the cells were fully adherent, each dilution was added to a 96-well plate at 100 μ L per well. The fusion protein was added as an administration group, the small peptide HM-3 was used as a control group, and PBS of the same volume was used as a blank control group at 37 ℃ with 5% CO2Incubate for 48 hours. To each well of the 96-well plate, 15. mu.L of 5mg/mL MTT was added and the culture was continued for 4 hours. The medium was aspirated off and dissolved by adding 100. mu.L DMSO per well. Detecting with enzyme-labeling instrument at 490nm, calculating growth inhibition rate, and repeating the experiment for 3 times. The results of the experiment are shown in FIG. 5.
The experiment selects cells as tumor cells, the cells are administrated at different concentrations, and the light absorption value is detected after 48 hours, MTT experiment proves that the proliferation inhibition effect of the small peptide HM-3 and the protein HSA on 5 tumor cells is not obvious, and the fusion protein has selective inhibition on the proliferation of the tumor cells, wherein the proliferation inhibition effect on B16F10 cells is most obvious and is concentration and time dependent.
Example 4 inhibition of Normal cells by fusion proteins
MTT method is adopted to detect the proliferation inhibition effect of the fusion protein on BEAS-2B (human normal lung epithelial cells), H9c2 (rat cardiac muscle cells), HIEC (human normal intestinal epithelial cells) and HL-7702 (human normal liver cells). Cells were incubated at 37 ℃ with 5% CO2The cells were collected by trypsinization after culturing in the incubator of (1) until the density became 90% or more, and the cells were resuspended in a culture medium and counted under a microscope to adjust the cell concentration to 3.0X 104Cell suspension was seeded into 96-well plates at 100. mu.L per well at 37 ℃ in 5% CO2The culture was carried out overnight in an incubator. The fusion proteins were diluted with PBS to each of the predetermined concentrations. After the cells were fully adherent, each dilution was added to a 96-well plate at 100 μ L per well. To add withAdding fusion protein as administration group, small peptide HM-3 as control group, and PBS with equal volume as blank control group at 37 deg.C and 5% CO2Incubate for 48 hours. To each well of the 96-well plate, 15. mu.L of 5mg/mL MTT was added, and the culture was continued for 4 hours. The medium was aspirated off and dissolved by adding 100. mu.L DMSO per well. Detecting with enzyme-labeling instrument at 490nm, calculating growth inhibition rate, and repeating the experiment for 3 times. The results of the experiment are shown in FIG. 6.
The experiment selects cells as human normal tissue cells and rat normal tissue cells, the administration is carried out at different concentrations, the light absorption value is detected after 48 hours, and MTT experiments prove that the proliferation inhibition effect of 3 medicaments on 4 normal cells is obviously lower than that of tumor cells B16F10, which indicates that the toxicity of the fusion protein on the normal cells is smaller.
Example 5Transwell assay for the Activity of fusion proteins to inhibit tumor cell migration
B16F10 cells were cultured in 1640 medium containing 10% fetal bovine serum at 37 ℃ with 5% CO2When the fusion protein is cultured in the incubator to reach the confluency of more than 80 percent, the activity of the fusion protein for inhibiting the migration of the tumor cells is detected by adopting a Transwell method, which comprises the following specific operations:
cells cultured to logarithmic growth phase were digested with 0.2% EDTA trypsin, collected, washed with PBS, diluted with serum-free 1640 medium, counted under a microscope, and the cell concentration was adjusted to 5X 105Per mL;
② inoculating cells into upper chamber of Transwell, 100 mul/well, adding test solution into upper chamber, adding fusion protein as administration group, small peptide HM-3(4.5 muM) as positive control group, using physiological saline with equal volume as blank control group, adding 0.6mL 1640 cell culture solution containing 10% fetal calf serum into lower chamber to stimulate cell migration, adding 5% CO2Incubating for 48h at 37 ℃;
and thirdly, discarding culture solution in the hole, fixing the culture solution with 4% paraformaldehyde at normal temperature for 30min, dyeing the culture solution with 0.1% crystal violet at normal temperature for 10min, rinsing the culture solution with PBS for 3 times, slightly wiping off non-migrated cells on the upper layer with a cotton swab, observing the cells under a microscope, selecting four fields for photographing and counting, calculating the migration inhibition rate, and repeating the experiment for 3 times.
It is reported in the literature that 4.5. mu.M HM-3 is able to achieve the highest mobility inhibition, so we chose 4.5. mu.M HM-3 as the control. The Transwell experiment (fig. 7) demonstrated that the higher the mobility of B16F10 cells was inhibited with increasing doses of the fusion protein drug. The inhibition rate reaches 32.2% at 250nM, while the mobility inhibition rate of 4.5 μ M HM-3 is 33.6%, which is equivalent to the drug effect of 250nM HM-3-HSA fusion protein, thus proving that the drug effect of the fusion protein is higher than that of the small peptide HM-3.
Sequence listing
<110> Lanzhou university
<120> a fusion protein for inhibiting angiogenesis
<160> 2
<170> SIPOSequenceListing 1.0
<210> 1
<211> 1842
<212> DNA
<213> human (Homo sapiens)
<400> 1
attgttagaa gagctgatag agctgctgtt ccaggtggtg gtggtagagg tgacggtggt 60
ggtggttctg gtggtggtgg ttctgatgct cataagtctg aagttgctca tagatttaag 120
gatttgggtg aagaaaactt taaggctttg gttttgattg cttttgctca atacttgcaa 180
caatgtccat ttgaagatca tgttaagttg gttaacgaag ttactgaatt tgctaagact 240
tgtgttgctg atgaatctgc tgaaaactgt gataagtctt tgcatacttt gtttggtgat 300
aagttgtgta ctgttgctac tttgagagaa acttacggtg aaatggctga ttgttgtgct 360
aagcaagaac cagaaagaaa cgaatgtttt ttgcaacata aggatgataa cccaaacttg 420
ccaagattgg ttagaccaga agttgatgtt atgtgtactg cttttcatga taacgaagaa 480
acttttttga agaagtactt gtacgaaatt gctagaagac atccatactt ttacgctcca 540
gaattgttgt tttttgctaa gagatacaag gctgctttta ctgaatgttg tcaagctgct 600
gataaggctg cttgtttgtt gccaaagttg gatgaattga gagatgaagg taaggcttct 660
tctgctaagc aaagattgaa gtgtgcttct ttgcaaaagt ttggtgaaag agcttttaag 720
gcttgggctg ttgctagatt gtctcaaaga tttccaaagg ctgaatttgc tgaagtttct 780
aagttggtta ctgatttgac taaggttcat actgaatgtt gtcatggtga tttgttggaa 840
tgtgctgatg atagagctga tttggctaag tacatttgtg aaaaccaaga ttctatttct 900
tctaagttga aggaatgttg tgaaaagcca ttgttggaaa agtctcattg tattgctgaa 960
gttgaaaacg atgaaatgcc agctgatttg ccatctttgg ctgctgattt tgttgaatct 1020
aaggatgttt gtaagaacta cgctgaagct aaggatgttt ttttgggtat gtttttgtac 1080
gaatacgcta gaagacatcc agattactct gttgttttgt tgttgagatt ggctaagact 1140
tacgaaacta ctttggaaaa gtgttgtgct gctgctgatc cacatgaatg ttacgctaag 1200
gtttttgatg aatttaagcc attggttgaa gaaccacaaa acttgattaa gcaaaactgt 1260
gaattgtttg aacaattggg tgaatacaag tttcaaaacg ctttgttggt tagatacact 1320
aagaaggttc cacaagtttc tactccaact ttggttgaag tttctagaaa cttgggtaag 1380
gttggttcta agtgttgtaa gcatccagaa gctaagagaa tgccatgtgc tgaagattac 1440
ttgtctgttg ttttgaacca attgtgtgtt ttgcatgaaa agactccagt ttctgataga 1500
gttactaagt gttgtactga atctttggtt aacagaagac catgtttttc tgctttggaa 1560
gttgatgaaa cttacgttcc aaaggaattt aacgctgaaa cttttacttt tcatgctgat 1620
atttgtactt tgtctgaaaa ggaaagacaa attaagaagc aaactgcttt ggttgaattg 1680
gttaagcata agccaaaggc tactaaggaa caattgaagg ctgttatgga tgattttgct 1740
gcttttgttg aaaagtgttg taaggctgat gataaggaaa cttgttttgc tgaagaaggt 1800
aagaagttgg ttgctgcttc tcaagctgct ttgggtttgt aa 1842
<210> 2
<211> 613
<212> PRT
<213> human (Homo sapiens)
<400> 2
Ile Val Arg Arg Ala Asp Arg Ala Ala Val Pro Gly Gly Gly Gly Arg
1 5 10 15
Gly Asp Gly Gly Gly Gly Ser Gly Gly Gly Gly Ser Asp Ala His Lys
20 25 30
Ser Glu Val Ala His Arg Phe Lys Asp Leu Gly Glu Glu Asn Phe Lys
35 40 45
Ala Leu Val Leu Ile Ala Phe Ala Gln Tyr Leu Gln Gln Cys Pro Phe
50 55 60
Glu Asp His Val Lys Leu Val Asn Glu Val Thr Glu Phe Ala Lys Thr
65 70 75 80
Cys Val Ala Asp Glu Ser Ala Glu Asn Cys Asp Lys Ser Leu His Thr
85 90 95
Leu Phe Gly Asp Lys Leu Cys Thr Val Ala Thr Leu Arg Glu Thr Tyr
100 105 110
Gly Glu Met Ala Asp Cys Cys Ala Lys Gln Glu Pro Glu Arg Asn Glu
115 120 125
Cys Phe Leu Gln His Lys Asp Asp Asn Pro Asn Leu Pro Arg Leu Val
130 135 140
Arg Pro Glu Val Asp Val Met Cys Thr Ala Phe His Asp Asn Glu Glu
145 150 155 160
Thr Phe Leu Lys Lys Tyr Leu Tyr Glu Ile Ala Arg Arg His Pro Tyr
165 170 175
Phe Tyr Ala Pro Glu Leu Leu Phe Phe Ala Lys Arg Tyr Lys Ala Ala
180 185 190
Phe Thr Glu Cys Cys Gln Ala Ala Asp Lys Ala Ala Cys Leu Leu Pro
195 200 205
Lys Leu Asp Glu Leu Arg Asp Glu Gly Lys Ala Ser Ser Ala Lys Gln
210 215 220
Arg Leu Lys Cys Ala Ser Leu Gln Lys Phe Gly Glu Arg Ala Phe Lys
225 230 235 240
Ala Trp Ala Val Ala Arg Leu Ser Gln Arg Phe Pro Lys Ala Glu Phe
245 250 255
Ala Glu Val Ser Lys Leu Val Thr Asp Leu Thr Lys Val His Thr Glu
260 265 270
Cys Cys His Gly Asp Leu Leu Glu Cys Ala Asp Asp Arg Ala Asp Leu
275 280 285
Ala Lys Tyr Ile Cys Glu Asn Gln Asp Ser Ile Ser Ser Lys Leu Lys
290 295 300
Glu Cys Cys Glu Lys Pro Leu Leu Glu Lys Ser His Cys Ile Ala Glu
305 310 315 320
Val Glu Asn Asp Glu Met Pro Ala Asp Leu Pro Ser Leu Ala Ala Asp
325 330 335
Phe Val Glu Ser Lys Asp Val Cys Lys Asn Tyr Ala Glu Ala Lys Asp
340 345 350
Val Phe Leu Gly Met Phe Leu Tyr Glu Tyr Ala Arg Arg His Pro Asp
355 360 365
Tyr Ser Val Val Leu Leu Leu Arg Leu Ala Lys Thr Tyr Glu Thr Thr
370 375 380
Leu Glu Lys Cys Cys Ala Ala Ala Asp Pro His Glu Cys Tyr Ala Lys
385 390 395 400
Val Phe Asp Glu Phe Lys Pro Leu Val Glu Glu Pro Gln Asn Leu Ile
405 410 415
Lys Gln Asn Cys Glu Leu Phe Glu Gln Leu Gly Glu Tyr Lys Phe Gln
420 425 430
Asn Ala Leu Leu Val Arg Tyr Thr Lys Lys Val Pro Gln Val Ser Thr
435 440 445
Pro Thr Leu Val Glu Val Ser Arg Asn Leu Gly Lys Val Gly Ser Lys
450 455 460
Cys Cys Lys His Pro Glu Ala Lys Arg Met Pro Cys Ala Glu Asp Tyr
465 470 475 480
Leu Ser Val Val Leu Asn Gln Leu Cys Val Leu His Glu Lys Thr Pro
485 490 495
Val Ser Asp Arg Val Thr Lys Cys Cys Thr Glu Ser Leu Val Asn Arg
500 505 510
Arg Pro Cys Phe Ser Ala Leu Glu Val Asp Glu Thr Tyr Val Pro Lys
515 520 525
Glu Phe Asn Ala Glu Thr Phe Thr Phe His Ala Asp Ile Cys Thr Leu
530 535 540
Ser Glu Lys Glu Arg Gln Ile Lys Lys Gln Thr Ala Leu Val Glu Leu
545 550 555 560
Val Lys His Lys Pro Lys Ala Thr Lys Glu Gln Leu Lys Ala Val Met
565 570 575
Asp Asp Phe Ala Ala Phe Val Glu Lys Cys Cys Lys Ala Asp Asp Lys
580 585 590
Glu Thr Cys Phe Ala Glu Glu Gly Lys Lys Leu Val Ala Ala Ser Gln
595 600 605
Ala Ala Leu Gly Leu
610

Claims (8)

1. A fusion protein, characterized in that: the amino acid sequence of the fusion protein is shown as SEQ ID NO. 1, and the DNA sequence for coding the amino acid sequence is shown as SEQ ID NO: 2, respectively.
2. The fusion protein of claim 1, wherein the fusion protein is produced by yeast cell expression.
3. The fusion protein of claim 4, wherein the yeast is Pichia methanolica (Pichia pastoris).
4. A method of producing the fusion protein of claim 1, comprising the steps of:
synthesizing DNA of fusion protein by using whole gene;
secondly, connecting the target segment with a vector by a genetic engineering technology to obtain a recombinant yeast expression vector containing a DNA sequence for coding the fusion protein;
and thirdly, the recombinant yeast expression vector in the step two is transformed into a competent escherichia coli cell, and the plasmid is transformed into a host expression system for expression, so that the fusion protein is obtained, wherein the host expression system is pichia methanolica.
5. A recombinant expression vector comprising a DNA sequence encoding the fusion protein of claim 1.
6. A host expression system comprising the recombinant expression vector of claim 5.
7. The use of the fusion protein of claim 1 in the preparation of an anti-tumor medicament.
8. The use of the fusion protein as claimed in claim 7 in the preparation of an antitumor drug, wherein the antitumor drug is in the form of an injection, a capsule, a tablet, a pill, a nasal spray or an aerosol.
CN202011366707.6A 2020-11-27 2020-11-27 Fusion protein for inhibiting angiogenesis Pending CN114560947A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202011366707.6A CN114560947A (en) 2020-11-27 2020-11-27 Fusion protein for inhibiting angiogenesis

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202011366707.6A CN114560947A (en) 2020-11-27 2020-11-27 Fusion protein for inhibiting angiogenesis

Publications (1)

Publication Number Publication Date
CN114560947A true CN114560947A (en) 2022-05-31

Family

ID=81712094

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202011366707.6A Pending CN114560947A (en) 2020-11-27 2020-11-27 Fusion protein for inhibiting angiogenesis

Country Status (1)

Country Link
CN (1) CN114560947A (en)

Similar Documents

Publication Publication Date Title
EP2210904B1 (en) Method of preparing a fusion protein comprising tumor necrosis factor related apoptosis inducing ligand and integrin ligand and use thereof
CN107261124B (en) Preparation method and application of angiogenesis inhibiting peptide and hyaluronic acid modifier thereof
US8664368B2 (en) Fully human monoclonal antibody to VEGF, preparation method and use thereof
CN101717449B (en) Recombinant TRAIL-Fc fusion protein as well as preparation and application thereof
CN112386678B (en) Use of polypeptides or derivatives thereof
CN111995686B (en) Medicine with anti-angiogenesis activity and preparation method thereof
CN113416260B (en) Claudin18.2-targeted specific chimeric antigen receptor cell and preparation method and application thereof
CN112851791B (en) Novel FGF analogue for resisting metabolic disorder and application thereof
CN107236046B (en) Recombinant human endostatin fusion protein and preparation method and application thereof
CN107417772B (en) Polypeptide HIP-20 capable of antagonizing RNA binding activity of hnRNPU protein and application thereof
EP0550769B1 (en) Use of hepatocyte growth factors for the manufacture of a hemopoietic stem cell augmenting agent
CN106589130B (en) A kind of Slit2D2-HSA recombinant protein and its application in treatment pyemia
CN107540739A (en) A kind of tumor-targeting polypeptide
CN110483647B (en) Anti-tumor polypeptide and application thereof
CN104193828A (en) Recombinant fusion protein capable of simultaneously blocking HER2 and VEGFR signal paths
CN114560947A (en) Fusion protein for inhibiting angiogenesis
CN111205361B (en) Interleukin 21 protein (IL21) mutant and application thereof
CN113583095B (en) Antitumor polypeptide and application thereof
CN116003627A (en) NKG2D-NKp46 cell adaptor molecules and uses thereof
CN113980133B (en) Antibody and application thereof in anti-tumor
WO2012019423A1 (en) Angiogenesis inhibitor, method for purifying thereof and pharmaceutical composition comprising thereof
CN109400711A (en) A kind of PDGFR β targeting tumor necrosin relative death inducing ligand variant and its preparation method and application
CN108864258A (en) With the PEGylated polypeptide and the preparation method and application thereof for inhibiting tumour function
CN101092452A (en) Preparation method for both of micromolecule polypeptide of tumor chalone for anti angiogenesis, and fusion protein
EP4261230A1 (en) Recombinant human serum albumin-collagen binding domain fusion protein for tumor-specific targeting matrix and application thereof

Legal Events

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