CN117025594A - Oligodeoxyribonucleic acid Awcpdo for improving liver fibrosis and application thereof - Google Patents

Oligodeoxyribonucleic acid Awcpdo for improving liver fibrosis and application thereof Download PDF

Info

Publication number
CN117025594A
CN117025594A CN202310867041.XA CN202310867041A CN117025594A CN 117025594 A CN117025594 A CN 117025594A CN 202310867041 A CN202310867041 A CN 202310867041A CN 117025594 A CN117025594 A CN 117025594A
Authority
CN
China
Prior art keywords
awcpdo
oligodeoxyribonucleic acid
acid
oligodeoxyribonucleic
liver fibrosis
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
CN202310867041.XA
Other languages
Chinese (zh)
Inventor
倪毅然
张丽叶
吴江锋
孙悦
张艳琼
王英
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.)
China Three Gorges University CTGU
Original Assignee
China Three Gorges University CTGU
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 China Three Gorges University CTGU filed Critical China Three Gorges University CTGU
Priority to CN202310867041.XA priority Critical patent/CN117025594A/en
Publication of CN117025594A publication Critical patent/CN117025594A/en
Pending legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • 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
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • A61K31/711Natural deoxyribonucleic acids, i.e. containing only 2'-deoxyriboses attached to adenine, guanine, cytosine or thymine and having 3'-5' phosphodiester links
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • A61K31/7115Nucleic acids or oligonucleotides having modified bases, i.e. other than adenine, guanine, cytosine, uracil or thymine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • A61K31/712Nucleic acids or oligonucleotides having modified sugars, i.e. other than ribose or 2'-deoxyribose
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • 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
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/32Chemical structure of the sugar
    • C12N2310/3222'-R Modification
    • 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
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/33Chemical structure of the base

Abstract

The invention discloses an oligodeoxyribonucleic acid Awcpdo for improving liver fibrosis and application thereof, wherein the nucleotide sequence of the oligodeoxyribonic acid is SEQ ID NO. 1, can specifically enter activated hepatic stellate cells, can competitively bind with a transcription factor PU.1, further acts on a downstream target spot, and inhibits HSC activation and liver fibrosis. The oligodeoxyribonucleic acid provided by the invention can be used for preparing medicines for preventing and treating liver fibrosis.

Description

Oligodeoxyribonucleic acid Awcpdo for improving liver fibrosis and application thereof
Technical Field
The invention belongs to the technical field of biomedicine, and particularly relates to an oligodeoxyribonucleic acid Awcpdo for improving liver fibrosis and application thereof.
Background
Liver fibrosis is an inflammatory pathological process and result from excessive deposition of extracellular matrix (Extracellular matrix, ECM) and various chronic injuries in the post-injury repair of liver tissue caused by a variety of factors (e.g., hepatitis virus infection, alcohol abuse, non-alcoholic fatty liver, immune response, drug and chemical poison damage, etc.). ECM is produced by Myofibroblasts (MFB), which are mainly derived from activated hepatic stellate cells (hepatic stellate cell, HSC), and liver injury activates hepatic stellate cells and causes accumulation of extracellular matrix is recognized as a key link in liver fibrosis.
Hepatic stellate cells are located in the perihepatic sinus space and are the major cell types involved in the healing and scarring of liver injury. Hepatic stellate cells are normally in a quiescent state. Upon liver injury, resting hepatic stellate cells are transformed into activated myofibroblasts, resulting in upregulation of α -smooth muscle actin (α -smooth muscle actin, α -SMA) and extracellular matrix deposition. Activation and proliferation of hepatic stellate cells involves a variety of cell signaling pathways, ultimately leading to imbalance in extracellular matrix synthesis and degradation, increased extracellular matrix production, or decreased degradation, are the root causes of liver fibrosis.
PU.1 (Purine-rich box 1) is a member of ETS (E26 transformation-specific) transcription factor family, encoded by SPI1 (human/mouse), which is a major regulator of hematopoietic stem cell proliferation and differentiation, maintaining cell homeostasis, playing an important role in inflammation, cell reprogramming and immunity. Pu.1 inhibits the growth of hepatocytes through TGF- β/Smad signaling pathways, thereby inducing and promoting precursor cell interstitial transformation, causing excessive deposition of ECM, leading to the occurrence of liver fibrosis. After knocking out the pu.1 gene from fibroblasts of human fibrous tissue, collagen release from fibroblasts was reduced, and α -SMA and F-actin (Filamentous actin) expression levels were similar to resting fibroblasts. In contrast, overexpression of PU.1 in resting fibroblasts can induce a transition from resting fibroblasts to a highly activated pro-fibrotic phenotype, an up-regulation of collagen release and expression of alpha-SMA and F-actin. Upregulation of PU.1 in fibrotic disease causes fibrosis-related genes to be induced, fibroblast phenotypes to change, and ECM production to increase. Whereas inactivation of pu.1 is effective to restore the fibrotic phenotype of fibroblasts to a quiescent state and induce regression of tissue fibrosis. In liver fibrosis, pu.1 is highly expressed in activated hepatic stellate cells and not in resting hepatic stellate cells.
The technology of trap oligonucleotides (decoy oligodeoxynucleotide, decoy ODN) is a gene therapy strategy, the principle of which is: based on the core motif of the transcription factor binding site (Transcription Factor binding sites, TFBS), DNA fragments containing TFBS (i.e., decoy ODN) are designed, synthesized, and transfected into cells to competitively capture the transcription factor, prevent the transcription factor from binding to TFBS on the promoter, block transcription of DNA, and regulate expression of downstream target genes. It has the advantages of easy synthesis, high specificity, relatively low synthesis cost, is more stable than siRNA sequences, and is considered as an effective therapeutic tool, and can inhibit the expression of specific genes under various medical conditions. The decoy ODN strategy is an effective method of inhibiting specific gene expression both in vitro and in vivo, and has been studied to confirm its role in cancer and fibrosis. Such as N F- κb decoy ODN, SP1 decoy ODN, etc., but the number and kinds of substances in the decoy ODN technology that can be used for preventing and treating liver fibrosis are small at present, and further development is still required.
Disclosure of Invention
In the present invention, unless otherwise indicated, scientific and technical terms used herein have the meanings commonly understood by one of ordinary skill in the art. Moreover, the procedures of cell culture, molecular biology, nucleic acid chemistry, etc., as used herein are all conventional procedures widely used in the corresponding fields. Meanwhile, in order to better understand the present invention, definitions and explanations of related terms are provided below.
The term "decoy ODN" generally refers to a synthetic DNA fragment designed based on the core motif of the transcription factor binding site, with the ability to competitively capture transcription factors; the term "liver fibrosis" generally refers to a pathophysiological process that refers to the abnormal proliferation of connective tissue within the liver caused by various pathogenic factors.
In order to achieve the above object, the present invention provides an oligodeoxyribonucleic acid, which is an exogenously synthesized nucleotide chain that can specifically enter activated HSCs and inhibit expression of liver fibrosis-promoting genes, inhibiting activation of HSCs and liver fibrosis; the nucleotide sequence of the oligodeoxyribonucleic acid is SEQ ID NO:1, and a nucleotide sequence of 1.
The oligodeoxyribonucleic acid comprises at least one chemical modification: modification of ribose in the oligodeoxyribonucleic acid sequence, including 2' -H quilt F, NH 2 Substitution of OMe; modifying any base in the oligodeoxyribonucleic acid sequence by amino, carboxyl, sulfhydryl, biotin, cholesterol and polyethylene glycol groups; modification of polyethylene glycol (PEG) at the 5 'end and deoxyuracil (dU), deoxythymine (dT) and deoxyhypoxanthine (dL) at the 3' end in the nucleotide sequence of the oligodeoxyribonucleic acid; at least one nucleotide in the nucleotide sequence of the oligodeoxyribonucleic acid is a locked nucleic acid.
The invention also provides an application of the oligodeoxyribonucleic acid in preparing a medicament for preventing and treating liver fibrosis diseases; the prevention and treatment of liver fibrosis disease is based on decoy ODN technology.
The invention also provides a pharmaceutical composition comprising an effective dose of oligodeoxyribonucleotide and a pharmaceutically acceptable carrier or diluent.
Preferably, the effective dose of the oligodeoxyribonucleic acid is 100-400nM.
The invention also provides application of the pharmaceutical composition in preparing medicines for preventing and treating hepatic fibrosis cells and animal disease models.
Preferably, the cell is a mammalian cell.
The invention has the beneficial effects that: the invention screens and obtains an oligodeoxyribonucleic acid sequence, predicts a secondary structure in The UNAFold website, selects a structure with The lowest Gibbs free energy delta G, and generates a tertiary structure of The polydeoxyribonucleic acid in Discovery Studio, which is named Awcpdo. The invention verifies that Awcpdo can target and activate HSC cells in a cell line and an animal model, and verifies that Awcpdo regulates the expression of hepatic fibrosis related genes alpha-SMA, COL1 alpha 2 and TIMP1 in activated HSCs; the observation of Awcpdo in animal model to improve liver fibrosis proves that the oligodeoxyribonucleic acid Awcpdo can be used for preventing and treating liver fibrosis progress, provides a new substance for decoy ODN technology, and provides scientific basis for application and delivery strategy research in liver fibrosis diseases.
Drawings
FIG. 1 is a simulated docking result of the core base sequence of Awcpdo in example 1 and the PU.1 protein, wherein A is a potential base sequence specifically binding to the PU.1 protein, and the letter size indicates the binding potential of each base at the corresponding position; b is a simulated graph of the butt joint of the core base sequence of the Awcpdo and the PU.1 protein, a green structure in the graph is a PU.1 protein DNA binding domain, a double-helix structure is a fragment of the Awcpdo combined with the PU.1 protein, and red dots are water molecules participating in the simulated butt joint.
FIG. 2 shows the results of the optimization of the working concentration of Awcpdo in example 3.1.
FIG. 3 is a fluorescence microscopy image validating that Awcpdo targets activated HSC in example 3.2.
FIG. 4 is a Western Blot chart showing the inhibition of expression of liver fibrosis promoting genes by Awcpdo in example 4, wherein A is a Western Blot gel electrophoresis chart, B is a COL1α2 expression level histogram, C is an α -SMA expression level histogram, and D is a TIMP1 expression level histogram; ns represents no statistical difference, P <0.05, P <0.01, P <0.001, and the values are mean±sd.
FIG. 5 is a serum stability experiment of Awcpdo in example 5. Lanes are samples of DNA Ladder, awcpdo, serum, awcpdo incubated with serum for 0h,0.5h,1h,2h,4h,6h,8h, respectively from left to right.
FIG. 6 is a photograph of a small animal imaged to verify that Awcpdo targets the liver in an animal model as in example 6.1, wherein A is CCL after various times of injection of Awcpdo 4 Living body of molded mouseImaging photograph, B is CCL after injecting Awcpdo for different time 4 Tissue imaging photographs of the model mice, wherein in B, heart, spleen, liver, lung and kidney are sequentially arranged from left to right.
FIG. 7 is a laser scanning confocal microscopy image of Awcpdo targeted activated HSC in an animal model verified in example 6.2, wherein A is an immunofluorescence photograph of co-localization of alpha-SMA with Awcpdo, and B is the presence of Awcpdo in CCL 4 Immunofluorescence photographs of heart, spleen, lung and kidneys in the model mice.
FIG. 8 is a graph showing the effect of Awcpdo on anti-hepatic fibrosis in example 7, wherein A is the liver of a normal mouse and B is CCL 4 Model mouse liver, C is the mouse liver of control group, D is the mouse liver of Awcpdo treatment group.
FIG. 9 is a graph showing the effect of thiomodified Awcpdo on anti-hepatic fibrosis in example 8, wherein A is the liver of a normal mouse and B is CCL 4 Model mouse liver, C is the mouse liver of Awcpdo group, D is the mouse liver of Awcpdo group after being modified by thio.
Detailed Description
The technical solution of the present invention will be further explained below with reference to the accompanying drawings and specific embodiments, and it should be noted that the following embodiments are only preferred embodiments of the present invention, and should not be construed as limiting the present invention, and the scope of the present invention shall be defined by the claims. Modifications and substitutions made by those skilled in the art without the inventive effort fall within the scope of the present invention.
The main reagent comprises:
(1) Neonatal bovine serum (NBCS): holly bioengineering materials limited (Hangzhou);
(2) Fetal bovine serum, DMEM medium: gibco corporation (USA);
(3) Trypsin Trypsin, double antibody (penicillin/streptomycin): is of the science and technology limited company of the biological products of the ocean (Tianjin city);
(4) PBS powder (0.01M), ECL chemiluminescent kit, 5 x protein loading buffer: seville biotechnology Co., ltd;
(5) DMSO, EB, β -actin antibodies: sigma Co., USA;
(6) Cell transfection reagent (Touborfect), xhoI, sal I, ecoR I, protein Ladder, protein quantitative analysis kit, DNA Ladder: us Thermo Fisher scientific;
(7) Cell transfection reagent (Neofect): zero-passenger creating intelligent (Beijing) biotechnology limited company;
(8) RIPA lysate: solarbio (beijing);
(9) Awcpdo, scr, fluorescent probe (cy 5, FAM, etc.) labeled Awcpdo/Scr and thio modified Awcpdo synthesis: shanghai Bioengineering Co., ltd;
(10) Plasmid extraction kit: nanjinouzan biotechnology Co., ltd;
(11) Transfer ribonucleic acid (yeast): moeiy biotechnology limited;
(12) YEAST EXTRACT (YEAST EXTRACT), TRYPTONE (TRYPTONE): oxyid LTD (uk);
(13) Competent preparation kit: takara (Takara doctor materials technologies Co., ltd.);
(14) AGAROSE (agarsose), tween20 (Tween 20): bioFrox, germany;
(15) Sodium chloride, tris-base, glycine (Glycine): shanghai Miclin Biochemical technologies Co., ltd;
(16) Ammonium Persulfate (APS) and Sodium Dodecyl Sulfate (SDS): henan Huamei bioengineering Co., ltd;
(17) Skimmed milk powder: inner Mongolian illi Co., ltd;
(18) TEMED (tetramethyl ethylenediamine): life Technologies (U.S.);
(19) PVDF film: millipore (Germany);
(20) 30% acrylamide-methylene bisacrylamide (30% acr-Bis): boschia martensii;
(21) COL 1a 2 antibodies: santa cruz, inc. of America;
(22) alpha-SMA antibody, TIMP1 antibody: abcronal Biotechnology Inc.;
(23) HRP-labeled goat anti-mouse/goat anti-rabbit secondary, 75% alcohol, and 84 antiseptic solution: wuhan flying agent Co., ltd.
Animal experiment related materials:
(1) Sterile syringes (1 mL, 5mL, and 10 mL): hunan oasis Huikang development Co., ltd;
(2) Sterile insulin syringe (U-40): bidi medical instruments (Shanghai) Inc.;
(3) Pathological blade: shanghai Lianhui medical supplies Co;
(4) Microscope cover slip, microscope slide, and adhesive slide: jiangsu Shitai laboratory equipment company;
(5) Light-resistant slide moisture box: huasheng experiment consumables (Taobao shop);
(6) Dehairing instrument: ant nationality flagship (Taobao shop).
Animal experiment related drugs and reagents:
(1) Carbon tetrachloride (CCL) 4 ) Sucrose, chloral hydrate: shanghai Miclin Biochemical technologies Co., ltd;
(2) Edible olive oil: sanchang good office limited (hong Kong);
(3) Paraformaldehyde fixative, 6 XDNA loading buffer, RIPA lysate, 10 XTBE, gelRed, anti-fluorescence quenching caplet, DAPI staining reagent: seville biotechnology Co., ltd;
(4) OCT frozen section embedding agent: jiangsu Meiger Biotech Co., ltd;
(5) Triton X-100, 5% bsa blocking solution, masson/HE staining kit and FITC-labeled secondary antibody: solarbio (beijing);
(6) PBS powder (0.01M): whanbo Oute Biotechnology Co., ltd;
(7) Glycerol, boric acid, glacial acetic acid, methanol and EDTA: national pharmaceutical group chemical reagent company.
The common solution is prepared:
(1) Cell cryopreservation solution: DMEM (-): NBCS: volume ratio of DMSO was 7:2:1, preparing, uniformly mixing, and storing in a dark place at 4 ℃;
(2) PBS solution: pouring 1 package of PBS powder (0.01M) into a measuring cylinder, adding 1L of double distilled water, stirring until the PBS powder is fully dissolved, and fixing the volume to 2L; sterilizing under high pressure, cooling to room temperature, and storing in a refrigerator at 4deg.C;
(3) DMEM (-): high sugar culture solution (without serum and antibiotics) with the specification of 500 mL/bottle;
(4) DMEM (+): to 267mL of DMEM (-) medium, 3mL of penicillin/streptomycin mixed solution (1%) and 30mL of newborn calf serum were added respectively, and mixed uniformly to prepare 300mL of DMEM (+) medium;
(5) Ampicillin solution (Amp): weighing 3g of Amp powder, adding a proper amount of ddH 2 Dissolving O sufficiently, fixing volume to 30mL (100 μg/μl), filtering with 0.22 μm sterile filter, packaging, and storing at-20deg.C;
(6) LB (-) solid Medium: weighing agar powder 3g, yeast extract 1g, tryptone 2g and sodium chloride 2g, adding appropriate amount of ddH 2 After O is fully dissolved, the volume is fixed to 200mL; sterilizing under high pressure, rapidly pouring into a sterile culture dish after the temperature is reduced to about 55 ℃ in an ultra-clean bench, sealing the whole culture dish with a preservative film after the culture dish is cooled and solidified, and inverting the culture dish in a refrigerator at 4 ℃ for later use;
(7) LB (-) liquid Medium: weighing 2g of sodium chloride, 2g of tryptone, 1g of yeast extract and adding a proper amount of ddH 2 After O is fully dissolved, the volume is fixed to 200mL; sterilizing under high pressure, cooling to room temperature, and preserving at 4deg.C;
(8) Amp (+) solid medium: preparing 200mL of LB (-) culture medium solution according to the formula (6), sterilizing under high pressure, cooling to about 55 ℃, adding 200 mu L of Amp with concentration of (100 mu g/. Mu.L), rapidly pouring into a sterile culture dish, sealing the whole culture dish with a preservative film after cooling and solidifying, and inverting the culture dish in a refrigerator with the temperature of 4 ℃ for later use;
(9) Amp (+) liquid medium: 200mL of LB (-) medium solution was prepared according to the formula (7), autoclaved, cooled to about 55℃and then added with 200. Mu.L of Amp at a concentration of (100. Mu.g/. Mu.L),
cooling to room temperature, and preserving at 4deg.C;
(10) 50×tae solution: weigh 24.2g Tris-Base and 37.2g EDTA Na 2 ·2H 2 Adding O powder into a measuring cup, adding 57.1mL of glacial acetic acid, and adding a proper amount of ddH 2 After O is fully dissolved, the volume is fixed to 1L (ph=8.5), and the O is stored at room temperature and diluted to 1×tae when in use;
(11) 10% Sodium Dodecyl Sulfate (SDS): SDS powder (10 g) was weighed and a proper amount of ddH was added 2 Stirring O until the O is fully dissolved, and then keeping the volume to 100mL, and preserving at room temperature for later use;
(12) Agarose gel (1%): weighing 0.4g agarose powder in a conical flask, adding 40mL 1 xTAE into the conical flask, heating the conical flask in a microwave oven until the powder is completely dissolved, shaking the conical flask slightly, adding 1.5 mu L EB when the temperature of the solution is reduced to about 50 ℃, mixing uniformly, immediately pouring into a mold groove, inserting a comb into the hole teeth, and naturally solidifying the solution to obtain the agarose powder;
(13) Weak acid working solution: according to weak acid solution: ddH 2 O=1: 2, the volume ratio is configured and used at present;
(14)20% CCL 4 : CCL (CCL) 4 : olive oil at 1:4, respectively measuring CCL of the required volume 4 The solution and olive oil solution are stirred in a beaker until being fully and evenly mixed to prepare 20 percent CCL 4 The olive oil mixed solution is preserved in dark at 4 ℃;
(15) Chloral hydrate (10%): 3g of chloral hydrate powder is weighed and a proper amount of autoclaved ddH is added 2 After O is fully dissolved, the volume is fixed to 30mL (100 mg/mL), and the O is preserved in a dark place at 4 ℃;
(16) 70% ethanol solution: ddH sterilized in an amount of 30mL 2 Adding O into 70mL of absolute ethyl alcohol, fully and uniformly mixing, and storing at normal temperature;
(17) 100. Mu.M of Awcpdo and Scr stock solution were prepared: according to the instructions volume, the powder was dissolved in autoclaved ddH 2 O, keeping at-20 ℃ for standby;
(18) DAPI solution (10%): 200 mu L of DAPI solution is added into 2mL of sterile PBS solution, and the mixture is fully and uniformly mixed for use;
(19) Triton (2%): according to PBS: triton=50: 1, respectively measuring the solution with the required volume into a beaker, and fully stirring and uniformly mixing the solution to obtain the finished product;
(20) 1×running Buffer: weighing 75.2g glycine powder, 4g SDS powder, 12.12g Tris Base powder, adding a proper amount of ddH 2 O is stirred, after the dissolution is sufficient, the volume is fixed to 4L, and the mixture is preserved at the temperature of 4 ℃;
(21) 1 XTrans Buffer: 57.6g glycine powder, 4g SDS powder, 12.12g Tris Base powder were weighed and a proper amount of ddH was added 2 O is stirred, after the dissolution is complete, 800mL of methanol is added, the volume is fixed to 4L after the mixture is uniformly mixed, and the mixture is preserved at 4 ℃;
(22) 1 XTBST: weighing 35.2g NaCl,9.68g Tris Base powder, adding appropriate amount of ddH 2 O is stirred, after the solution is fully dissolved, 4mL of 0.05% Tween20 is added into the solution, the mixture is continuously mixed evenly, and then ddH is added 2 O is fixed to 4L, pH is regulated to 7.4, and the mixture is preserved at 4 ℃;
(23) 5% skim milk solution: weighing 2g of skimmed milk powder into a 50mL centrifuge tube, adding 40mL of 1 XTBST solution, and shaking by vortex until the skimmed milk powder is fully dissolved for preparation;
(24) 10% APS: 5g of ammonium persulfate powder was weighed into a beaker, and 50mL of ddH was added thereto 2 Stirring O until it is fully dissolved, packaging into 1.5mL EP tube (1 mL/tube), and storing at 20deg.C in dark place;
(25) 10 XTBE: weighing 216g of Tris-base, 110g of boric acid and 80mL of 0.5M EDTA, adding 1600mL of distilled water for complete dissolution, then, fixing the volume to 2L, adjusting the pH to 8.3, and preserving at normal temperature;
(26) 50% glycerol: taking 100mL of glycerol and 100mL of distilled water, fully and uniformly stirring, and preserving at high pressure and normal temperature;
(27) 2 XBuffer: tris-base 90.84g,10% SDS 20mL was weighed and dissolved in 400mL ddH 2 O, stirring until the mixture is completely dissolved, then, fixing the volume to 1000mL, and adjusting the pH to 8.8;
(28) Tris-HCl (1M): tris-base 48.44g was weighed and dissolved in 200mL ddH 2 O, after stirring to complete dissolution, constant volume to 400mL, ph=6.8.
Plasmid, cell line and animal:
plasmid PCDNA3.1 (+), PCDNA3.1-ALK3, PCDNA3.1-ALK5: tumor microenvironment at university of three gorges and professor Liu Changbai to the focus laboratory for immunotherapy;
HSC-T6 cells (rat hepatic stellate cells): giving by Song Yuhu professor (university of science and technology, china, same-aid Hospital);
specific pathogen free (Specific pathogen free, SPF) grade male C57BL/6 mice: 6-8 weeks old (body weight 20.+ -.2 g), purchased and fed to the university of three gorges laboratory animal center, laboratory animal production license number: SYXK 2022-0012, barrier system, drinking water, ingestion and illumination all meet the Experimental animal quality control Standard in Hubei province.
Example 1 design of Awcpdo
Potential base sequences for specific binding of PU.1 proteins were obtained by database Jaspar (https:// Jaspar. Geneg. Net /), the results are shown in FIG. 1A; the sequence of the obtained oligonucleotide sequence Awcpdo based on the sequence extension is SEQ ID NO. 1. The obtained Awcpdo was subjected to simulated docking with PU.1 protein, and as a result, awcpdo could bind to PU.1 protein as shown in FIG. 1B.
EXAMPLE 2 culture of HSC-T6 cells
(1) Taking out HSC-T6 cell frozen stock solution from-80deg.C refrigerator, rapidly placing into water bath of 37deg.C, rapidly shaking to defrost within 1min,
(2) Transferring the thawed cell suspension into a 15mL centrifuge tube with 3mL of DMEM (+) cell culture solution added in advance, lightly mixing, and centrifuging at 800rpm for 3 min;
(3) After the supernatant was aspirated, 3.5mL of DMEM (+) was added, gently mixed, transferred to a cell culture dish, and 5% CO was added 2 Culturing in an incubator at 37 ℃;
(4) HSC-T6 cells were harvested for use when cells grew on the wall and reached 80% density.
Example 3 validation of Awcpdo-targeted activated HSC in cell lines
3.1 optimization of the working concentration of Awcpdo
(1) The HSC-T6 cell suspension obtained in example 2 was taken at a cell amount of 1X 10 per well 5 Adding the cells into a 24-well plate to be subjected to transfection when the cells are attached and the fusion degree reaches 70-80%;
(2) Discarding old culture medium in the plate, adding PBS for washing twice, adding 400 mu L of DMEM (-) in a 24-pore plate, adding 900 mu L of DMEM (-) in a 6-pore plate, and performing starvation treatment;
(3) Taking sterile EP tubes (500 mu L) corresponding to each hole, adding 100 mu L of DMEM (-) into each tube, adding 1 mu g of plasmid pCDNA3.1-ALK5, gently mixing, and standing for 6min;
(4) The transfection reagent (Turbofect) was added to the EP tube of (3) in a volume ratio of: plasmid = 2:1, gently mixing, and standing for 20min;
(5) All the mixed liquid in the step (4) is dripped into the corresponding hole, and the culture plate is put into an incubator for continuous culture after being gently rocked left and right and up and down; after 6h, cell replacement is carried out, 500 mu LDMEM (+) is added into each hole of DMEM (-), and transfection is carried out for 24h;
(6) Awcpdo was added to each well of (5) to a final concentration of 0nM, 25nM, 50nM, 100nM, 150nM, 200nM, 300nM, 400nM, 500nM, co-cell incubation and after 24h, cell-line cytometry analysis was collected.
As a result, as shown in FIG. 2, the fluorescence intensity of Awcpdo reached the maximum at 300nM and as the concentration continued to increase, the fluorescence intensity in the cells did not increase any more; comprehensive analysis, 300nM is the optimum working concentration of Awcpdo.
3.2, fluorescence microscopy experiments with Awcpdo-targeted activated HSC verify
(1) Transfecting plasmid pCDNA3.1-ALK5 into HSC-T6 cells according to steps (1) - (5) in 3.1, obtaining activated HSC-T6 cells after 24 hours;
(2) FAM-labeled Atcpdo was added to the non-activated HSC-T6 cells (i.e., NC panel in FIG. 3) and activated HSC-T6 cells (i.e., ALK5 panel in FIG. 3) for 24h incubation at a final concentration of 20 nM; qualitative and quantitative analysis of HSC-T6 cells in ALK5 and NC groups was performed using fluorescence microscopy and flow cytometry. As a result, atcpdo can enter activated HSC-T6 cells without entering unactivated HSC-T6 cells, as shown in FIG. 3.
Example 4Awcpdo inhibits liver fibrosis Gene expression
The experimental groupings were as follows:
(1) Mock group: blank control, HSC-T6 cells were untreated;
(2) ALK5 group: transfecting plasmid pCDNA3.1-ALK5 into HSC-T6 cells according to steps (1) - (5) in 3.1, obtaining activated HSC-T6 cells after 24 hours;
(3) Scr group: transfecting plasmid pCDNA3.1-ALK5 into HSC-T6 cells according to steps (1) - (5) in 3.1, obtaining activated HSC-T6 cells after 24 hours; subsequently 300nM of Scr was added per well and incubated for a further 24h;
(4) Awcpdo group: transfecting plasmid pCDNA3.1-ALK5 into HSC-T6 cells according to steps (1) - (5) in 3.1, obtaining activated HSC-T6 cells after 24 hours; subsequently 300nM of Awcpdo was added per well and incubated for an additional 24h;
(5) ALK3 group: transfecting plasmid pCDNA3.1-ALK3 into HSC-T6 cells according to steps (1) - (5) in 3.1, obtaining activated and inhibited HSC-T6 cells after 24 hours;
collecting HSC-T6 cells obtained in the steps (1) - (5), extracting protein, performing Western blot to detect expression conditions of liver fibrosis related genes COL1α2, TIMP1 and alpha-SMA, and performing statistical analysis, wherein beta-actin is used as an internal reference.
As a result, as shown in FIG. 4, COL1α2, TIMP1, and α -SMA were all up-regulated in the ALK 5-treated activated HSC-T6, whereas the protein expression was down-regulated after the respective treatment with Awcpdo, and returned to be consistent with or slightly lower than that of the Mock group, whereas that of the Scr group did not. Taken together, the results demonstrate that Awcpdo is capable of inhibiting expression of genes associated with pro-hepatic fibrosis in activated HSC-T6 cells.
Example 5 serum stability experiments on Awcpdo
(1) Eyeball blood of a male C57BL/6 liver fibrosis model mouse is taken in a sterilized 1.5mL EP tube, centrifuged at room temperature at 3000rpm for 5min, and upper serum is taken in a new EP tube;
(2) Respectively mixing Awcpdo with the concentration of 10 mu M in serum with medium volume, incubating in a water bath kettle at 37 ℃, respectively collecting samples at 0h,0.5h,1h,2h,4h,6h,8h and 10h, and temporarily storing in a refrigerator at-80 ℃ for later use;
(3) Preparing 10% non-denaturing polyacrylamide gel (Native-PAGE), and the formulation of the gel is as follows;
10% Native-PAGE
(4) After vortex mixing, injecting the solution into two layers of glass plates, after the glue is completely solidified, placing the glue into an electrophoresis tank, filling the inner tank with 0.5 XTBE, adding a proper amount of 0.5 XTBE buffer solution into the outer tank, and pre-electrophoresis for 2 hours;
(5) Taking out the sample from the refrigerator at-80 ℃, thawing on ice, adding 6 XDNA loading with corresponding volume, mixing uniformly, and then instantly centrifuging for about 10 s;
(6) Adding sample, 100V electrophoresis for about 2 hours, and soaking and dyeing glue with GelRed working solution at room temperature for 30min; the position of the band and the extent of dispersion of the band were observed with a gel imaging system.
As a result, the Awcpdo was degraded at 0.5h and the degradation was substantially complete at 4h, as shown in FIG. 5.
Example 6 Small animal imaging and laser scanning confocal microscopy verification of Awcpdo-targeted activated HSC in animal models
6.1 imaging verification of Awcpdo targeting liver in animal model by small animals
(1) A20% CCL injection was subcutaneously administered to mice at 5mL/kg body weight 4 Injecting once every 3 days for 8 weeks to obtain CCL 4 Mice induced liver fibrosis for 8 weeks;
(2) 100 μl (10 μM) of the mice obtained in step (1) were injected via tail vein to form Cy 5-labeled Awcpdo for biological synthesis in Shanghai, and the mice were imaged in vivo and in tissue at 0h,0.5h,1h,2h,3h, and 5h, respectively.
As a result, awcpdo starts to aggregate in the liver at 0.5h and metabolize to the kidney as shown in FIG. 6A; the aggregation was evident in the liver at 1h, and the fluorescence at the kidney metabolism was not as strong as the fluorescence at the liver, and 5h was almost completed from the kidney metabolism. In the same period, hearts, spleens, lungs and kidneys of mice were taken out and photographed, and as a result, as shown in fig. 6B, fluorescence was not seen in the hearts, spleens, lungs, but was accumulated in the liver and gradually metabolized the kidneys with the lapse of time. Taken together, awcpdo has a certain targeting to the liver.
6.2, laser scanning confocal microscopy validation of Awcpdo-targeted activated HSC in animal models
To further verify whether Awcpdo is on CCL 4 The hepatic stellate cells activated by the model-making mice have a targeting effect, and the liver imaged by the tissue is adopted for fixation, dehydration and embedding of paraformaldehyde to prepare frozen sections for immunofluorescence. The activated hepatic stellate cell markers α -SMA were co-localized with Awcpdo, respectively. As shown in fig. 7a, the first column shows DAPI blue fluorescence, the nuclei are displayed, the second column shows Awcpdo red fluorescence, the third column shows α -SMA green fluorescence, and the fourth column shows three combined (merger) images, wherein representative regions of the 0.5h and 3h merger images are shown in column 5 after enlargement, and the combined images are visible: the green fluorescence of α -SMA was largely concentrated around the blood vessel and hepatic blood sinus, and red light of the Cy 5-labeled Awcpdo was found under the high power microscope to be concentrated near the hepatic stellate nucleus around the hepatic blood sinus, and approximately coincident with the green light of α -SMA and appeared yellow, consistent with the expected results. It was demonstrated that Awcpdo has a targeting effect on hepatic stellate cells activated by model-producing mice. The results of heart, spleen, lung and kidney are shown in FIG. 7B, and red fluorescence is not seen. Indicating that Awcpdo has no targeting effect on these organs.
Example 7 measurement of the anti-hepatic fibrosis Effect of Awcpdo
To verify whether Awcpdo has anti-fibrotic effect in mice, CCL was tested 4 Mice induced liver fibrosis for 8 weeks were given 2 times a week by tail vein injection of Awcpdo or Scr (negative control), respectively, with CCL4 modeling (on monday and friday CCL 4), monday, friday and friday Awcpdo or Scr) for 4 weeks, and normal mice, CCL were taken after 4 weeks 4 Model, awcpdo treatment group and Scr group mouse livers were observed and photographed as a whole, and H was performed&E staining and Masson staining, results are shown in FIG. 8, CCL compared to normal mice (A of FIG. 8) 4 The liver surface of the model (B of FIG. 8) was not smooth, had a hard texture, and had pronounced particulate nodules, H&E staining has massive inflammatory cell infiltration accompanied by massive cell necrosis, masson staining shows significant proliferation of collagen fibers, and sink region and bloodThe thickening of collagen fibers around the tube is obvious, the collagen fibers of the hyperplasia among the lobules extend along the inflammatory necrosis area, and a plurality of collagen fiber bundles are crosslinked to divide liver lobules. Scr group mice (C of FIG. 8) and CCL 4 No significant difference was seen from the general view of the liver to pathological examination compared to model mice. While Awcpdo treated mice (D of FIG. 8) and CCL 4 Compared with modeling, the particles on the surface of the liver are obviously reduced, the texture is softer, H&E staining showed reduced inflammatory cell infiltration and cell necrosis, masson staining showed a significant reduction in the proliferated collagen fibers. Taken together, it is demonstrated that Awcpdo has an anti-hepatic fibrosis effect in mice.
Example 8 detection of the anti-hepatic fibrosis Effect of thio-modified Awcpdo
Awcpdo binds to PU.1 through specific base sequence, and various chemical modifications to Awcpdo including but not limited to "modification of ribose in the oligodeoxyribonucleic acid sequence including 2' -H being F, NH 2 Substitution of OMe; modifying any base in the oligodeoxyribonucleic acid sequence by amino, carboxyl, sulfhydryl, biotin, cholesterol and polyethylene glycol groups; modification of polyethylene glycol (PEG) at the 5 'end and deoxyuracil (dU), deoxythymine (dT) and deoxyhypoxanthine (dL) at the 3' end in the nucleotide sequence of the oligodeoxyribonucleic acid; at least one nucleotide in the nucleotide sequence of the oligodeoxyribonucleic acid is a locked nucleic acid ", and can still have the anti-liver fibrosis effect of the original sequence.
To verify the anti-hepatic fibrosis effect of the chemically modified Awcpdo, we performed an Awcpdo thio modification between the deoxyguanosine at position 18 and the deoxyuracil at position 19, followed by animal experiments following the method of example 8, i.e.: to CCL (CCL) 4 Mice induced liver fibrosis for 8 weeks were administered 2 times a week by tail vein injection of either Awcpdo or Awcpdo after thioation, respectively, for a duration of CCL 4 Moulding (in Tuesday and friday CCL) 4 Administration, monday, wednesday and Saturday Awcpdo or Scr), and normal mice were harvested after 4 weeks 4 The model, the Awcpdo group and the mouse liver of the Awcpdo group after being thioated are integrally observed and photographedAnd Masson staining was performed, and the results were shown in FIG. 9, which shows CCL compared with normal mice (A of FIG. 9) 4 The liver surface of the model (B of FIG. 9) is not smooth, the texture is hard, obvious granular nodules are formed, masson staining shows that liver cell edema is obvious, collagen fibers are obviously proliferated, thickening of collagen fibers around a manifold area and blood vessels is obvious, and the proliferated collagen fibers between the leaflets extend along an inflammatory necrosis area, and a plurality of collagen fiber bundles are crosslinked to divide liver leaflets. Awcpdo group mice (C and D of FIG. 9) and CCL after thiolation of Awcpdo group 4 Compared with model mice, the particles on the surface of the liver are obviously reduced, the texture is softer, and the Masson dyeing shows that the collagen fibers which are proliferated are obviously reduced. Taken together, it is demonstrated that the thio-modified Awcpdo has an anti-hepatic fibrosis effect in mice similar to the original sequence.
The object of the present invention is to provide a novel oligodeoxyribonucleic acid, awcpdo, with specific binding and access to activated HSCs, inhibiting HSC activation and liver fibrosis. The present invention detects the optimum working concentration of Awcpdo, and the results show that Awcpdo has the best affinity for HSC-T6 at 300 nM; confirming that Awcpdo can enter activated HSC in a targeting way through means such as a laser scanning confocal microscope, small animal imaging and the like; through Western Blot and other means, it is confirmed that Awcpdo can inhibit liver fibrosis promotion gene expression; in the mouse liver fibrosis model, injection of Awcpdo significantly improved liver fibrosis. Awcpdo is expected to be applied to the prevention and treatment of hepatic fibrosis.

Claims (7)

1. An oligodeoxyribonucleic acid, characterized by: the nucleotide sequence of the oligodeoxyribonucleic acid is SEQ ID NO:1, said oligodeoxyribonucleic acid is capable of specifically entering activated HSC cells.
2. An oligodeoxyribonucleic acid according to claim 1, characterised in that: the oligodeoxyribonucleic acid comprises at least one chemical modification: modification of ribose in the oligodeoxyribonucleic acid sequence, including substitution of 2' -H by F, NH, OMe; modifying any base in the oligodeoxyribonucleic acid sequence by amino, carboxyl, sulfhydryl, biotin, cholesterol and polyethylene glycol groups; modification of polyethylene glycol (PEG) at the 5 'end and deoxyuracil (dU), deoxythymine (dT) and deoxyhypoxanthine (dL) at the 3' end in the nucleotide sequence of the oligodeoxyribonucleic acid; at least one nucleotide in the nucleotide sequence of the oligodeoxyribonucleic acid is a locked nucleic acid.
3. Use of the oligodeoxyribonucleic acid according to claim 1 or 2 for the preparation of a medicament for the prophylaxis and treatment of liver fibrosis diseases; the prevention and treatment of liver fibrosis disease is based on decoy ODN technology.
4. A pharmaceutical composition characterized by: the pharmaceutical composition comprises an effective amount of the oligodeoxyribonucleotide according to claim 1 or 2, and a pharmaceutically acceptable carrier or diluent.
5. The pharmaceutical composition of claim 4, wherein: the effective dose of the oligodeoxyribonucleic acid is 100-400nM.
6. Use of a pharmaceutical composition according to claim 4 or 5 for the preparation of a medicament for the prevention and treatment of hepatic fibrosis cells and animal disease models.
7. The use according to claim 6, characterized in that: the cell is a mammalian cell.
CN202310867041.XA 2023-07-14 2023-07-14 Oligodeoxyribonucleic acid Awcpdo for improving liver fibrosis and application thereof Pending CN117025594A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202310867041.XA CN117025594A (en) 2023-07-14 2023-07-14 Oligodeoxyribonucleic acid Awcpdo for improving liver fibrosis and application thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202310867041.XA CN117025594A (en) 2023-07-14 2023-07-14 Oligodeoxyribonucleic acid Awcpdo for improving liver fibrosis and application thereof

Publications (1)

Publication Number Publication Date
CN117025594A true CN117025594A (en) 2023-11-10

Family

ID=88601308

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202310867041.XA Pending CN117025594A (en) 2023-07-14 2023-07-14 Oligodeoxyribonucleic acid Awcpdo for improving liver fibrosis and application thereof

Country Status (1)

Country Link
CN (1) CN117025594A (en)

Similar Documents

Publication Publication Date Title
EP3553077B1 (en) Peptide for repairing cartilage and treating osteoarthritis
EP3336190A1 (en) Methods, systems, and compositions for cell-derived/vesicle-based microrna delivery
Zhou et al. Activated macrophage membrane-coated nanoparticles relieve osteoarthritis-induced synovitis and joint damage
CN101874111A (en) Utilize the endogenous chylomicron to send to be used to the system of the nucleic acid that suppresses expression of target gene
Carotenuto et al. Dietary flaxseed mitigates impaired skeletal muscle regeneration: in vivo, in vitro and in silico studies
Huang et al. Nanoparticle-mediated inhibition of notch signaling promotes mitochondrial biogenesis and reduces subcutaneous adipose tissue expansion in pigs
Zhou et al. Exosomes derived from dental pulp stem cells accelerate cutaneous wound healing by enhancing angiogenesis via the Cdc42/p38 MAPK pathway
US11622964B2 (en) Method for destroying cellular mechanical homeostasis and promoting regeneration and repair of tissues and organs, and use thereof
Zhao et al. Effect of Danshao Huaxian capsule on Gremlin and bone morphogenetic protein-7 expression in hepatic fibrosis in rats
CN107353333A (en) A kind of bone protectiveness molecule Semaphorine3A and its pharmacy application
Guo et al. Influence of serum collected from rat perfused with compound Biejiaruangan drug on hepatic stellate cells
CN110684774B (en) Aptamer specifically binding to DEK protein and application thereof
CN117025594A (en) Oligodeoxyribonucleic acid Awcpdo for improving liver fibrosis and application thereof
CN116904463A (en) Oligodeoxyribonucleic acid Atcpdo for improving liver fibrosis and application thereof
CN115896111A (en) Small interfering RNA for inhibiting ACSL4 expression and application thereof
CN111388499A (en) Application of miR-31 in preparation of colon-targeted nano-drug for preventing and treating ulcerative colitis
CN110229215A (en) A kind of medicament slow release polypeptide hydrogel and its preparation method and application
Xie et al. Effect of a subset of adipose‐derived stem cells isolated with liposome magnetic beads to promote cartilage repair
CN108403665B (en) Prostate cancer targeted drug delivery carrier modified by EpDT3 aptamer, delivery system, preparation method and application thereof
CN113521080A (en) Application of CX-5461 in preparation of PHF6 mutant acute myeloid leukemia medicine
CN112516318A (en) Application of CUL4B in treatment of type II diabetes
Peng et al. Locally administered lentivirus-mediated siRNA inhibits wear debris-induced inflammation in murine air pouch model
CN101524548A (en) Nanometer carrier combination and application thereof to preparation of anti-tumor drug
CN111388452A (en) Adipose tissue targeting peptide P3-chitosan oligomeric lactic acid-polyethylene glycol delivery system and application thereof in nucleic acid drug delivery
WO2023077711A1 (en) Screening method of medicament for preventing prostatic cancer and application of nitazoxanide in pharmacy

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