CN113143949B - SiRNA of target SLC22A12 gene and application thereof in hyperuricemia accompanied with hyperlipidemia and hyperglycemia treatment - Google Patents

SiRNA of target SLC22A12 gene and application thereof in hyperuricemia accompanied with hyperlipidemia and hyperglycemia treatment Download PDF

Info

Publication number
CN113143949B
CN113143949B CN202110480769.8A CN202110480769A CN113143949B CN 113143949 B CN113143949 B CN 113143949B CN 202110480769 A CN202110480769 A CN 202110480769A CN 113143949 B CN113143949 B CN 113143949B
Authority
CN
China
Prior art keywords
group
sirna
slc22a12
uric acid
gene
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
CN202110480769.8A
Other languages
Chinese (zh)
Other versions
CN113143949A (en
Inventor
葛科立
张志霞
葛银林
贾少平
薛美兰
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.)
Qingdao University
Original Assignee
Qingdao 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 Qingdao University filed Critical Qingdao University
Priority to CN202110480769.8A priority Critical patent/CN113143949B/en
Publication of CN113143949A publication Critical patent/CN113143949A/en
Application granted granted Critical
Publication of CN113143949B publication Critical patent/CN113143949B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • 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
    • A61K31/713Double-stranded nucleic acids or oligonucleotides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/06Antigout agents, e.g. antihyperuricemic or uricosuric agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/06Antihyperlipidemics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • 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
    • 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
    • C12N2310/10Type of nucleic acid
    • C12N2310/14Type of nucleic acid interfering N.A.
    • 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
    • C12N2320/00Applications; Uses
    • C12N2320/30Special therapeutic applications

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Health & Medical Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Public Health (AREA)
  • Medicinal Chemistry (AREA)
  • Diabetes (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Biomedical Technology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Molecular Biology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Obesity (AREA)
  • Hematology (AREA)
  • Biochemistry (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • General Engineering & Computer Science (AREA)
  • Biotechnology (AREA)
  • Rheumatology (AREA)
  • Endocrinology (AREA)
  • Physics & Mathematics (AREA)
  • Biophysics (AREA)
  • Emergency Medicine (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Plant Pathology (AREA)
  • Microbiology (AREA)
  • Pain & Pain Management (AREA)
  • Epidemiology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)

Abstract

The invention belongs to the technical field of medicines for gene therapy, and particularly relates to siRNA targeting SLC22A12 gene and application thereof in hyperuricemia accompanied with hyperlipidemia and hyperglycemia treatment. The invention utilizes the effect of SLC22A12 gene in tubular and small intestine lumen cells, and further utilizes an RNA interference (RNAi) method to construct SLC22A12 gene small molecule interfering RNA, namely siRNA, to knock down SLC22A12 gene expression, reduce uric acid generation and reabsorption in tubular and small intestine lumen, thereby achieving the purpose of reducing blood uric acid.

Description

SiRNA of target SLC22A12 gene and application thereof in hyperuricemia accompanied with hyperlipidemia and hyperglycemia treatment
Technical Field
The invention belongs to the technical field of medicines for gene therapy, and particularly relates to siRNA targeting SLC22A12 gene and application thereof in hyperuricemia accompanied with hyperlipidemia and hyperglycemia treatment.
Background
Hyperuricemia (HUA) is a metabolic disease caused by purine metabolic disturbance and increased uric acid in blood, and is a fourth major underlying metabolic disease next to hypertension, hyperlipidemia and hyperglycemia. Clinical studies have found that: 70% of patients with hyperuricemia have abnormal glucose tolerance; 62.2% have lipid metabolic disorders, 58.2% have weight gain or obesity. Uric acid metabolic disorder is not only the pathological basis of gouty arthritis, kidney stones and kidney failure, but also is closely related to metabolic syndrome diseases such as hypertension, atherosclerosis, coronary atherosclerotic heart disease, disorder of blood lipid and blood glucose metabolism, obesity, insulin resistance and the like, so that the uric acid is controlled, and the development of hyperlipidemia and hyperglycemia diseases is favorably controlled.
The SLC22A12 gene codes for a protein of a urate transporter1 (Urate Transporter, URAT 1), is expressed in the apical membrane of tubular epithelial cells, small intestinal epithelial cells and the like, and plays an important role in the absorption of glucose and uric acid into cells from tubular lumens and intestinal lumens. The increase of SLC22A12 gene expression can promote the increase of uric acid reabsorption in the lumen, so that the increase of blood uric acid and blood sugar is realized, the decrease of SLC22A12 gene expression can reduce the uric acid reabsorption in the lumen, the increase of uric acid discharge is realized, and the decrease of blood uric acid is realized. The reduction of blood uric acid can be achieved by reducing blood lipid and blood sugar by the tricarboxylic acid cycle of purine nucleotide metabolic pathway, which is equivalent to the influence of the interconnection of sugar and lipid metabolism, and thus controlling body weight.
Disclosure of Invention
According to the defects existing in the prior art, the invention provides siRNA targeting SLC22A12 gene and application thereof in hyperuricemia accompanied by hyperlipidemia and hyperglycemia treatment, applies RNA interference technology to SLC22A12 gene combination, and provides a treatment thought and a method for targeted inhibition of SLC22A12 gene expression treatment to regulate uric acid increase accompanied by dyslipidemia.
The invention is realized by adopting the following technical scheme:
the invention provides application of a nucleic acid substance for inhibiting or silencing SLC22A12 gene expression in preparation of a medicine for treating hyperuricemia accompanied with hyperlipidemia and hyperglycemia.
Wherein, the nucleic acid substance for inhibiting or silencing SLC22A12 gene is siRNA for inhibiting URAT1 protein expression.
Specifically, the siRNA is formed by annealing a sense strand and an antisense strand; the nucleotide sequence of the sense strand of the siRNA is SEQ ID NO. 1, and the nucleotide sequence of the antisense strand is SEQ ID NO. 2.
The invention also provides an siRNA, which is the siRNA for inhibiting SLC22A12 gene expression.
The core of the invention is that the expression of SLC22A12 gene is knocked down by RNA interference (RNAi) method, the reabsorption of sugar uric acid in the tubular and intestinal lumens is reduced, and finally the purposes of reducing blood uric acid, reducing blood lipid and blood sugar and controlling weight are achieved by regulating uric acid generation, uric acid excretion and glycoprotein fat metabolism. According to the mRNA sequence of the SLC22A12 gene and the RNA interference principle, three pairs of double-stranded small interfering RNAs, namely SiRNA-1, siRNA-2 and SiRNA-3 sequences, are designed, and the sequences are shown in a table 2; the missense SCR siRNA (scRNA) without any homology with mouse mRNA is (Negative Control, NC) and the sequence is shown in Table 2, namely, the best knockdown effect is identified by screening in vitro (cell level), and in vitro experiments (animal experiments) with the best knockdown effect are performed.
Compared with the prior art, the invention has the beneficial effects that:
the siRNA sequence provided by the invention targets the SLC22A12 gene of encoding urate transporter1 (Urate Transporter, URAT 1) protein, and in-vitro experiments prove that the content of URAT1 protein can be reduced efficiently, and the effects of obviously reducing blood uric acid, reducing blood lipid, reducing blood sugar and controlling weight are achieved, so that the siRNA sequence can be used as a drug development thought for reducing blood uric acid, controlling blood sugar, blood lipid and controlling weight, and the accurate targeted therapeutic drug is developed.
Drawings
The present invention will become more fully understood from the detailed description given herein and the accompanying drawings, which are given by way of illustration only, and thus do not limit the intended scope of the invention.
FIG. 1 is a preliminary result of the design of siRNA targeting the SLC22A12 gene of example 1;
FIG. 2 is a graph showing the results of RNAStructure4.5 software in example 1 for predicting the partial secondary structure of mRNA for SLC22A12 to match three SiRNAs;
FIG. 3 is a graph showing comparison of mRNA transcription amounts of SLC22A12 detected by RT-PCR in 5 groups using TCMK1 as a target cell in example 2;
FIG. 4 is a graph showing comparison of the amounts of SLC22A12 protein expressed by Western blotting using TCMK1 as a target cell and 5-group detection in example 2, with β -actin as an internal reference;
FIG. 5 is a graph showing the comparison of uric acid concentration in and out of TCMK1 cells in example 3, where P <0.05 compared to the blank;
FIG. 6 shows the results of SUA changes at weeks 0, 4, 6 and 8 of the KM male mice 3 groups in examples 4 and 5;
FIG. 7 shows the results of UUA changes at weeks 0, 4, 6 and 8 of the KM male mice 3 groups in examples 4 and 5;
FIG. 8 shows the results of TG changes at 0, 4, 6 and 8 weeks in the 3 groups of KM male mice in examples 4 and 5;
FIG. 9 shows the results of T-CHO changes at weeks 0, 4, 6 and 8 of the KM male mice 3 group in examples 4 and 5;
FIG. 10 shows the results of Glu changes at weeks 0, 4, 6 and 8 for the KM male mice 3 groups of examples 4 and 5;
FIG. 11 shows the results of Weight changes at weeks 0, 4, 6 and 8 of the KM male mice 3 groups in examples 4 and 5;
FIG. 12 is a graph showing the serum creatinine Cr content of 8 th week KM male mice 3 groups in example 6 in FIG. 12 (A) and the blood urea nitrogen BUN content in FIG. 12 (B); changes in xanthine oxidase XOD fig. 12 (C);
FIG. 13 is a graph showing changes in the detection of proinflammatory factor L-1β by ELISAI method in the kidney cortex (FIG. 13 (A)) and small intestine ileum (FIG. 13 (B)) grouped by 8-week KM male mice 3 in example 6;
FIG. 14 is a graph showing changes in uric acid in the small intestine contents of 3 groups of 8-week KM male mice in example 6;
FIG. 15 is a HE staining immunohistochemical view of the kidney cortex (FIG. 15 (A)) and small intestine ileum (FIG. 15 (B)) groups of 8 th week KM male mice 3 in example 6;
FIG. 16 is a graph showing changes in the expression of immunohistochemical URAT1 proteins in the kidney cortex (FIG. 16 (A)) and the small intestine ileum (FIG. 16 (B)) grouped by 8 th week KM male mice 3 in example 6.
Detailed Description
The present invention will be described in further detail with reference to the accompanying drawings, in order to make the objects and technical solutions of the present invention more apparent. The experimental methods described in the following examples are all conventional methods unless otherwise specified; the specific techniques or conditions are not identified in the examples and are performed according to techniques or conditions described in the literature in this field or according to product specifications; the reagents and materials, unless otherwise specified, are commercially available.
1. The materials used in the following examples are as follows:
mouse tubular epithelial cells (TCMK 1) were passaged 1:3 at 80-90% confluency.
TCMK1 cell culture medium solution: the volume ratio is 87% dmem, high sugar +12% fbs +1% diabody. FBS, fetal bovine serum, available from Dalian Mei Lun Biotechnology Co., ltd., product number PWL001; double antibody 100U/ml penicillin/streptomycin solution, available from company under the trade designation; DMEM, high sugar, available from Thermo FisherScientific company, cat No. 11965118
RNA transfection reagent Entranster TM -R4000: purchased from england biosystems under the trade designation 4000-3.
RNA transfection reagent Entranster TM -in vivo: purchased from england biosystems under the trade designation 18668-11-1.
The high blood uric acid pellet feed is prepared from the following components in percentage by mass: 10% yeast powder (OXOID; LP 0021) +10% fructose (Macklin; F875004) +20% streaky pork+60% basal feed (Jinan Pengyue laboratory animal Breeding Co., ltd.).
Basal feed, purchased from Jinan Pengyue laboratory animal breeding limited company, common environment laboratory mouse feed.
Urine uric acid UUA, serum uric acid SUA, triglyceride TG, total cholesterol T-CHO, serum creatinine Cr, blood manipulating urea nitrogen BUN detection kit: the above kits were purchased from Nanjing Searching Biotechnology, inc., under the designations C012-2-1, A110-1-1, A111-2-1, C011-2-1, C013-2-1, A048-2-1, and A048-2-1, respectively, and were all performed as described in the Kit.
Blood glucose Glu detection kit: glucose assay kit (Glucose oxidase method; rsbio.com), all procedures were performed according to the kit instructions.
Mouse IL-1 beta ELISA kit: available from Liankebio under the catalog number 70-EK201B/3-96.
2. Experimental procedure
Extracellular RNA transfection procedure:
(A) In vitro transfection procedure: (1) 0.67. Mu.g (50 pmol) of siRNA was diluted with a predetermined amount of serum-free DMEM and mixed well to prepare an RNA dilution with a final volume of 25. Mu.l. (2) Mu.l of Entranster-R4000 was taken, and then 24. Mu.l of serum-free dilution was added thereto, followed by thoroughly mixing to prepare an Entranster-R4000 dilution, the final volume of which was 25. Mu.l. Standing at room temperature for 5 minutes. (3) The Entranster-R4000 dilution and the RNA dilution were mixed well by shaking with a shaker and allowed to stand at room temperature for 15 minutes. The preparation of the transfection complex is completed. (4) Mu.l of the transfection complex was added dropwise to cells in 0.45ml of whole medium (which may contain 10% serum and antibiotics), the dishes were moved back and forth, and mixed well. (5) And observing the cell state 6 hours after transfection, if the cell state is good, culturing for 48 hours continuously without changing a culture medium, lysing the cells, extracting RNA and protein, and performing Real-time PCR and Western blotting to obtain the result.
(B) In vivo transfection procedure: usage of EntransterTM-in vivo. SiRNA (μg) and Entranster-in vivo (μl) were used in a 1:2 ratio as exemplified below with 12.5 μg of nucleic acid and 25 μl of transfection reagent, a total injection volume of 100 μl,20g mouse tail intravenous injection. (1) dilution of nucleic acid. 12.5. Mu.g of nucleic acid was diluted to 1. Mu.g/. Mu.l with an appropriate amount of endotoxin-free pure water (if the concentration of the nucleic acid stock solution was small, the injection volume was increased), 12.5. Mu.l of water was added, 25. Mu.l of 10% glucose solution (w/v) was further added, and the final volume was 50. Mu.l, and thoroughly mixed. (2) dilution of transfection reagent. Mu.l of Entranster-in vivo reagent was diluted with 25. Mu.l of 10% glucose solution to a final volume of 50. Mu.l and thoroughly mixed. (3) transfection complex formation. The diluted transfection reagent is added into the diluted nucleic acid solution at room temperature and thoroughly mixed. (4) standing at room temperature for 15 minutes. The prepared transfection compound is ready to use just by being prepared and is not suitable for long-term storage.
Real-time PCR operation: cells were lysed with TRIzol (Sigma), extracted with isopropanol RNA, and then precipitated with ethanol. First strand cDNA (Nanjing Vazyme) was obtained by reverse transcription using 1. Mu.g RNA. According toIII RT Supermix qPCR (Nanjing Vazyme) kit instruction book a PCR reaction system is prepared as follows: mu.l of final volume reaction was performed with 2. Mu.l of cDNA product containing 10. Mu.l (Vazyme), 0.4. Mu.l of forward and antisense primers, ddH of 2 XChamQ-SYBR-Color-qPCR-Master mix 2 O7.2. Mu.l. Gene expression value normalization internal reference housekeeping gene GAPDH. The sequences of the gene-specific primers used for PCR and the reaction conditions used are shown in Table 1.
TABLE 1 RT-PCR specific primer sequences for SLC22A12 genes and reaction conditions used
Western blotting procedure: cells were lysed with RIPA (Solarbio) to extract proteins. Western blotting (Western blotting) was performed with 15. Mu.g of protein. Briefly, the membranes were blocked in 5% nonfat milk powder for 1h, then incubated with rabbit anti-human SLC22A12 (1:1000; ariobalo) overnight at 4 ℃. Incubation with horseradish enzyme-labeled goat anti-rabbit antibody (Zsbio) for 40min, detection of the bands by chemiluminescence method, image-pro plus 8. Software calculates grey scale values. Beta-actin is an internal reference protein.
Liver homogenate detection xanthine oxidase XOD procedure: mice were sacrificed, liver samples were rapidly removed, placed on ice, mixed with physiological saline 1:9, and homogenized. Centrifuging the homogenate at 4deg.C for 10min at 12,000r/min, collecting supernatant, centrifuging the supernatant at 4deg.C for 10min at 12,000r/min, collecting supernatant, and measuring the activity of Xanthine Oxidase (XOD) in mouse liver by using Xanthan kit (Colorimetric method) kit (Nanjing Biotechnology Co., A002-1-1), according to the description of the kit.
Detection of proinflammatory factor IL-1 beta ELISAI operation:
mice were sacrificed and the renal cortex and ileum portions of the small intestine were removed rapidly and homogenized, and the supernatant was removed and homogenized in the same manner as in liver. Detection of IL-1β content in renal cortex and ileum by enzyme-linked immunosorbent assay (ELISA):
the basic steps are as follows:
(1) Coating: the antibodies were diluted with carbonate coating buffer to a protein content of 1-10 μg/ml. Mu.l of the polystyrene ELISA plate was added to each well at 4℃overnight. The next day, the solution in the wells was discarded, and washed 3 times with wash buffer for 3min each. (the antibody is coated in the commercial kit, and this step can be omitted)
(2) Sealing: mu.l of blocking solution was added to each well and incubated at 37℃for 1-2h.
(3) Washing: carefully removing the sealing plate film, putting into a plate washer, and washing for 3-5 times. The plate can also be manually washed: the liquid was discarded, 300. Mu.l of wash solution was added to each well, soaked for 1-2min, and the mixture was dried on absorbent paper by beating, and repeated 3-5 times. (the first three steps can be omitted) the antibody is coated in a general commercial kit
(4) Sample adding: 100. Mu.l of a sample to be examined, which was properly diluted, was added to the above-mentioned coated reaction well. (blank wells, double diluted standard wells, conditional on addition of negative control wells and positive control wells as quality control points).
(5) Incubation: and (3) incubating for 1-2h at 37 ℃ after membrane sealing plates are used.
(6) Washing: and 3, the same step as the step.
(7) Adding an antibody: 100 μl of diluted biotinylated antibody working solution was added to each well.
(8) Incubation: the plates were then incubated at 37℃for 1h.
(9) Washing: and 3, the same step as the step.
(10) Adding enzyme conjugate: mu.l of diluted enzyme conjugate working solution was added to each well.
(11) Incubation: and (5) incubating for 30min at 37 ℃ in a dark place after the membrane sealing plate is used.
(12) Washing: and 3, the same step as the step.
(13) Adding a chromogenic substrate: 100 μl of TMB substrate solution was added to each well and reacted at 37deg.C in the dark for 10-30 min until a significant color gradient was observed in the diluted wells of the standard.
(14) Terminating the reaction: to each reaction well was added 100. Mu.l of 2M sulfuric acid, and the color changed from blue to yellow.
(15) And (3) measuring results: after 10min, the OD of each well was measured on a microplate reader at 450nm with a blank control well zeroed.
Uric acid content detection of the whole small intestine content: the mice of each group were sacrificed by a late fasting before the end of the experiment, and the entire small intestine was excised immediately (from the upper duodenum to the lower ileum). Then put on a sterile table, cut longitudinally, scrape all intestinal contents as much as possible, centrifuge at 12,000r/min for 10min at 4 ℃, take supernatant, and centrifuge the supernatant at 12,000r/min for 10min at 4 ℃. The method for determining uric acid by taking the supernatant is the same as described above.
HE staining immunohistochemistry:
(A) HE staining: groups of mice were sacrificed, kidney cortex and ileal tissue were fixed with 4% paraformaldehyde, embedded with paraffin, sectioned (4 μm) and patch, dewaxed, HE stained, dehydrated transparent, blocked, microscopic observations, scanned or photographed.
(B) Immunohistochemistry: mouse kidney cortex and ileum tissues were sectioned in 4 μm paraffin, immunoreactivity was identified by reaction with anti-URAT 1 (1:4000) polyclonal antibody (1:200, 37 ℃ C. For 1 h) and horseradish peroxidase-labeled streptavidin (1:200, 37 ℃ C., for 1 h), and observed microscopically, scanned or photographed.
3. KM male mice group
4-week-old KM male mice were grown at 12: alternating day and night for 12 hours, feeding at room temperature, and adaptively feeding for 1 week to obtain free diet. The model feed is prepared by simulating unhealthy life habits of high purine, high sugar, high fat and high protein of human: according to the mass fraction of 10 percent of yeast powder, 10 percent of fructose, 20 percent of pork streaky pork and 60 percent of basic feed. The experiments were grouped into three groups:
normal feed control group (a blank control group, BCG): a basal diet;
hyperuricemia model feed group (a hyperuricemia model group, MG): model feed diet;
SiSLC22A12 group (SiSLC 22A 12): 1-4 week model diet, 4-8 week model diet + SiSLC22a12 tail intravenous (1.3 mg/kg injected once every 3 days).
4. Statistics
All experimental data were calculated using SPSS statistics 26 software, corrected by Ponfroney and T-test, expressed as mean+ -SD. Statistical significance was determined using single factor anova pairwise comparisons (inter-group comparisons) and multi-factor anova pairwise comparisons (inter-group comparisons and intra-group comparisons), statistical significance was P <0.05. Significance was marked with an letter notation: if the group comparisons or the group comparisons have the same letter (or symbol), meaning there is no significant difference; if there is no identical letter (or symbol), this means a significant difference.
Example 1 design of siRNA targeting the SLC22A12 Gene
At NCBI: the full-length sequence of mRNA was obtained from the https:// www.ncbi.nlm.nih.gov/Gene module of the website Mus musculus solute carrier family (facilitated glucose transporter), membrane 12 (Slc 22a 12), (No. NM-009203.3), using BLOCK-iT TM RNAi Designer online design software, input No. NM_009203.3 in Accession number; selecting Open Reading Frame (ORF) as a homologous interference target; the Minimum G/C percentage is 35%, and the Maximum G/C percentage is 55%; alignment exclusion of homologous gene sequences BLAST was performed using Mouse-Mus musculus database. The results are shown in FIG. 1.
The 10 siRNA sequences obtained by the preliminary design were then verified. The site http:// sidirect2.Rnai. Jp/design. Cgi and http:// biodev. Extra. Sea. Fr/DSIR. Html were designed and verified on-line, respectively, and it was found that the three above-described SiRNA design software had inhibitory effects at three sites 510-550, 1360-1380 and 1820-1840 in the Open Reading Frame (ORF) of the SLC22a12 gene: the siRNA designed by the software is DNA base A, G, C, T, and T in the DNA sequence is converted into U according to the pairing principle of transcription by taking DNA as a template to generate RNA base A, G, C, U. RNAStructure4.5 software was used to predict mRNA secondary structure of SLC22A12, avoiding complex secondary structure, and matching local secondary structure to SiRNA as shown in FIG. 2.
SCR is a missense siRNA (scramble-siRNA), i.e., an out-of-order siRNA without any homology to mouse mRNA, as a negative control. The siRNA sequences were chemically synthesized by Shanghai Ji Ma biological Co., ltd, and the final designed target SLC22A12 gene siRNA sequences are shown in Table 2.
TABLE 2 SCR siRNA sequences and SiRNA double-stranded base sequences of three pairs of SLC22A12 genes
Example 2 SiRNAs screening
The experiments were grouped into 5 groups, blank (Blank Control Group, BC); SCRNA negative control group (ScRNA Negative Control, SCR); siRNA-1 group (SiRNA-1), siRNA-2 group (SiRNA-2), siRNA-3 group (SiRNA-3), gene delivery by RNA transfection reagent, total RNA and protein of TCMK1 cells were extracted 48 hours after transfection, and Real-time PCR and Western blotting test were performed, the results of which are shown in FIG. 3 and FIG. 4.
The results show that: Q-PCR statistical analysis, WB band gray scale analysis, siRNA-2, which is named SiSLC22A12 for subsequent experiments, has a significant down-regulation effect on mRNA and protein level expression.
Example 3 verification of the silencing Effect at the cellular level uric acid absorption experiment
TCMK1 cells were divided into 4 experimental groups: normal medium group (Normal Control Group, NCG); uric Acid Group (UAG): culturing TCMK1 cells in a medium containing 400. Mu. Mol/L uric acid; SCR group: culture medium with 400 mu mol/L uric acid+SCR siRNA transfection (final concentration of SiRNA is 100nM, siRNA: R4000 (100 pmol 1:2 mu L), siSLC22A12 group: culture with 400 mu mol/L uric acid+SiSLC 22A12 transfection. After 24h of transfection, extracellular medium and lysed cells were taken respectively, and the change of intracellular and extracellular uric acid concentration was measured, as shown in FIG. 5. The results show that the extracellular uric acid concentration of SiSLC22A12 group is significantly higher than that of MG group and SCR group, indicating that SiSLC22A12 has significant uric acid absorption inhibition effect.
Example 4 establishment and verification of hyperuricemia with hyperlipidemia and hyperglycemia mouse model
The 1-4 week model establishes a normal feed control group and a hyperuricemia model group, and at the end of week 4, all mice were prohibited from eating the diet the night before, tail bladder stimulation was performed to obtain urine, tail vein blood was performed, body Weight was measured weekly for Serum Uric Acid (SUA), urine Uric Acid (UUA), triglyceride (TG), total cholesterol (T-CHO), blood glucose (Glu), and Weight as shown in Table 3.
TABLE 3 Change Table of hyperuricemia with hyperlipidemia hyperglycemia for 4 weeks, normal feed control group and hyperuricemia model group SUA, UUA, TG, T-CHO, glu and Weight
The hyperuricemia model group is fed by model feed for 4 weeks, the food is forbidden in the evening before 4 th weekend, urine and blood are taken, various indexes are detected, and the measured body weight per week shows that: SUA, UUA, TG, T-CHO, glu and Weight of the MG group were significantly higher than BCG (P < 0.05). The modeling of mice with hyperuricemia and hyperglycemia is successful.
Example 5, mouse model SiSLC22A12 interference preliminary outcome verification
The test groups mice were subjected to the above parameters by taking blood from the tail vein at week 6, taking urine and measuring the weights SUA, UUA, TG, T-CHO, glu and Weight weekly, ending at week 8, and the test methods were the same as in example 4. The results are shown in FIGS. 6 to 11.
Animal models were established for 0-4 weeks, and as a result, SUA, UUA, TG, T-CHO, glu and Weight of the MG group were significantly higher than BCG (P < 0.05), indicating that mice with hyperuricemia and hyperglycemia were modeled successfully, and that the body Weight of the MG group mice was significantly higher than that of the BCG group.
Treatment efficacy was varied for each experimental group from 4-8 weeks: siSLC22A12 significantly reduced blood uric acid, triglyceride and blood glucose levels. As the diet time of the MG group mice model increased, UUA excretion decreased, while the SiSLC22a12 group mice UUA excretion increased. Weight gain in MG mice model diet time, weight gain, sisc 22a12 mice, weight loss.
Example 6, mouse model SiSLC22A12 interference end result verification
At the end of week 8, each experimental group of mice:
(1) The serum creatinine Cr content (fig. 12 (a)), the blood urea nitrogen BUN content (fig. 12 (B)) was examined; liver was taken as liver homogenate to detect changes in xanthine oxidase XOD (fig. 12 (C)); the results show that: the above 3 index BCG groups are lower than the MG group, SLC22a12 group (P < 0.05), MG group higher than BCG group and SLC22a12 group (P < 0.05), SLC22a12 group lower than MG group (P < 0.05) higher than BCG group (P < 0.05);
(2) Taking kidney cortex (FIG. 13 (A)) and small intestine ileum (FIG. 13 (B)) and detecting the change of proinflammatory factor L-1 beta by using an ELISAI method; the results show that: the content of proinflammatory factor IL-1 beta in kidney and small intestine is lower in BCG group than in MG group and SiSLC22A12 group (P < 0.05); MG group higher than BCG group sisc 22a12 (P < 0.05), sisc 22a12 group lower than MG group (P < 0.05) higher than BCG group (P < 0.05);
(3) Taking the whole small intestine content to detect the change of uric acid in the small intestine content (figure 14); the BCG group is higher than the MG group, the SiSLC22A12 group (P < 0.05), the MG group is lower than the BCG group SiSLC22A12 group (P < 0.05), the SiSLC22A12 group is lower than the BCG group (P < 0.05) and higher than the MG group (P < 0.05); the results show that: MG group, model diet mice reduced uric acid excretion from the small intestine, while simc 22a12 group mice increased small intestine uric acid excretion.
(4) The renal cortex and small intestine ileum were taken for HE staining immunohistochemistry to detect changes in kidney and small intestine structure (fig. 15 (a), 15 (B)), and changes in expression of the immunohistochemical SLC22a12 protein (fig. 16 (a), 16 (B)); expression of SLC22a12 protein in the kidney, small intestine: BCG group is lower than MG group (P < 0.05) and higher than sisc 22a12 group (P < 0.05), MG group is higher than BCG group sisc 22a12 group (P < 0.05), sisc 22a12 group is low BCG group, MG group (P < 0.05).
As can be seen from fig. 15 (a), the HE stained kidney structure was changed: dashed arrows refer to changes in glomeruli, kidney vesicles, MG groups show: the kidney small sac space is widened, and mucous exudation exists in the sac cavity. The solid arrows indicate the tubular epithelial cells and the arrangement disorder degree thereof, the arrangement of the MG group cells is disorder, and liquid seeps out from the lumen; BCG group shows that cells are orderly arranged, and no liquid seeps out from the lumen; the SiSLC22A12 cells were aligned and some fluid was exuded from the lumen. Open arrows indicate that fibroblasts, MG groups had more fibroblast proliferation than the other experimental groups.
As can be seen from fig. 15 (B), HE-stained small intestine villus structure changes: the dashed arrow indicates the degree of disorder of the arrangement of the small intestine epithelial cells, and the BCG group shows that the cells are orderly arranged; MG group showed villus swelling, cell arrangement disorder, visible multilayer arrangement; the SiSLC22A12 cells were aligned. The solid arrows indicate changes in capillary blood flow conditions in the villi, with changes in blood flow conditions in the MG group.
The results illustrate: the kidney and small intestine of the MG group mice have inflammation; siSLC22A12 can improve the change of kidney and small intestine structure caused by hyperuricemia, hyperlipidemia and hyperglycemia, and has protective effect on kidney and small intestine.
To sum up, the sisc 22a12 interference sequence of the present invention:
(1) Can reduce the transcription of mRNA of SLC22A12 gene and the expression of protein at the cellular level, and reduce the absorption of TCMK1 cells to uric acid;
(2) The expression of the kidney and small intestine URAT1 proteins of the model mice is increased, and SiSLC22A12 can reduce the expression of the kidney small intestine SLC22A12 gene;
(3) Can reduce uric acid, triglyceride and blood sugar levels of mice with hyperuricemia and hyperlipidemia, and has weight reducing effect;
(4) Can reduce the content of kidney and small intestine pro-inflammatory factors IL-1 beta and reduce the occurrence of kidney and small intestine inflammation; increase renal excretion of creatinine and urea nitrogen: can improve the structure of the kidney and small intestine, which shows that SiSLC22A12 has the functions of protecting the kidney and the small intestine;
(5) Can promote uric acid excretion from small intestine.
It should be understood that the foregoing description is only a preferred embodiment of the present invention, and the present invention is not limited to the above-described embodiment, but may be modified or substituted for some of the features described in the above-described embodiments. Any modification, equivalent replacement, improvement, etc. made within the spirit and principle of the present invention should be included in the protection scope of the present invention.

Claims (1)

1. Application of nucleic acid substance for inhibiting or silencing SLC22A12 gene expression in preparing medicine for treating hyperuricemia accompanied with hyperlipidemia and hyperglycemia;
the nucleic acid substance for inhibiting or silencing SLC22A12 gene is siRNA for inhibiting URAT1 protein expression;
the siRNA is formed by annealing a sense strand and an antisense strand; the nucleotide sequence of the sense strand of the siRNA is SEQ ID NO. 1, and the nucleotide sequence of the antisense strand is SEQ ID NO. 2.
CN202110480769.8A 2021-04-30 2021-04-30 SiRNA of target SLC22A12 gene and application thereof in hyperuricemia accompanied with hyperlipidemia and hyperglycemia treatment Active CN113143949B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202110480769.8A CN113143949B (en) 2021-04-30 2021-04-30 SiRNA of target SLC22A12 gene and application thereof in hyperuricemia accompanied with hyperlipidemia and hyperglycemia treatment

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202110480769.8A CN113143949B (en) 2021-04-30 2021-04-30 SiRNA of target SLC22A12 gene and application thereof in hyperuricemia accompanied with hyperlipidemia and hyperglycemia treatment

Publications (2)

Publication Number Publication Date
CN113143949A CN113143949A (en) 2021-07-23
CN113143949B true CN113143949B (en) 2023-10-10

Family

ID=76872729

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202110480769.8A Active CN113143949B (en) 2021-04-30 2021-04-30 SiRNA of target SLC22A12 gene and application thereof in hyperuricemia accompanied with hyperlipidemia and hyperglycemia treatment

Country Status (1)

Country Link
CN (1) CN113143949B (en)

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN113181377A (en) * 2021-04-30 2021-07-30 青岛大学 Application of ABCG2 protein in preparation of preparation for improving, treating or assisting in treating hyperuricemia with hyperlipidemia and hyperglycemia
CN114672485A (en) * 2021-08-24 2022-06-28 阿格纳生物制药有限公司 SiRNA molecule, shRNA molecule and application thereof

Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2006013904A1 (en) * 2004-08-03 2006-02-09 Mochida Pharmaceutical Co., Ltd. Medicinal composition containing meltrin antagonist
WO2011038700A1 (en) * 2009-09-29 2011-04-07 苏州瑞博生物技术有限公司 Inhibitors of fam3b gene, inhibitor compositions, inhibiting methods and applications of inhibitors in preparing pharmaceuticals
WO2012033720A1 (en) * 2010-09-08 2012-03-15 Wellstat Therapeutics Corporation Benzoic acid compounds for reducing uric acid
CN105399694A (en) * 2015-12-11 2016-03-16 浙江京新药业股份有限公司 Axially chiral enantiomers of drug Lesinurad
CN108004310A (en) * 2017-12-13 2018-05-08 深圳大学 The application of feritin (original) acceptor (P) RR genes and its inhibitor
CN109528812A (en) * 2019-01-11 2019-03-29 青岛大学 A kind of preparation method of composition for preventing and treating diabetes
CN111088268A (en) * 2019-03-05 2020-05-01 北京五加和分子医学研究所有限公司 Gene therapy medicine for hyperuricemia

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050197312A1 (en) * 2004-03-03 2005-09-08 Kevin Fitzgerald Transcription factor RNA interference reagents and methods of use thereof

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2006013904A1 (en) * 2004-08-03 2006-02-09 Mochida Pharmaceutical Co., Ltd. Medicinal composition containing meltrin antagonist
WO2011038700A1 (en) * 2009-09-29 2011-04-07 苏州瑞博生物技术有限公司 Inhibitors of fam3b gene, inhibitor compositions, inhibiting methods and applications of inhibitors in preparing pharmaceuticals
WO2012033720A1 (en) * 2010-09-08 2012-03-15 Wellstat Therapeutics Corporation Benzoic acid compounds for reducing uric acid
CN105399694A (en) * 2015-12-11 2016-03-16 浙江京新药业股份有限公司 Axially chiral enantiomers of drug Lesinurad
CN108004310A (en) * 2017-12-13 2018-05-08 深圳大学 The application of feritin (original) acceptor (P) RR genes and its inhibitor
CN109528812A (en) * 2019-01-11 2019-03-29 青岛大学 A kind of preparation method of composition for preventing and treating diabetes
CN111088268A (en) * 2019-03-05 2020-05-01 北京五加和分子医学研究所有限公司 Gene therapy medicine for hyperuricemia

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
Uric acid increases fibronectin synthesis through upregulation of lysyl oxidase expression in rat renal tubular epithelial cells;Zhou Yang et al.;Am J Physiol Renal Physiol;第299卷;F336-F346 *
Xu, YJ et al..Anti-inflammatory effect of low molecular weight fucoidan from Saccharina japonica on atherosclerosis in apoE-knockout mice.INTERNATIONAL JOURNAL OF BIOLOGICAL MACROMOLECULES.2018,第118卷365-374. *
Zhou Yang et al..Uric acid increases fibronectin synthesis through upregulation of lysyl oxidase expression in rat renal tubular epithelial cells.Am J Physiol Renal Physiol.2010,第299卷F336-F346. *
健脾化浊通络方合用苯溴马隆对糖尿病前期合并高尿酸血症患者尿酸及血糖、血脂的影响;林玺等;中国中医药科技;第28卷;409-410 *
郑征等.n-3多不饱和脂肪酸饮食对小鼠肥胖及相关细胞因子的影响.食品科学.2010,第31卷1-5. *

Also Published As

Publication number Publication date
CN113143949A (en) 2021-07-23

Similar Documents

Publication Publication Date Title
Bera et al. Reciprocal regulation of miR-214 and PTEN by high glucose regulates renal glomerular mesangial and proximal tubular epithelial cell hypertrophy and matrix expansion
Li et al. miR-103 promotes 3T3-L1 cell adipogenesis through AKT/mTOR signal pathway with its target being MEF2D
CN113143949B (en) SiRNA of target SLC22A12 gene and application thereof in hyperuricemia accompanied with hyperlipidemia and hyperglycemia treatment
Geng et al. MiRNA-99a can regulate proliferation and apoptosis of human granulosa cells via targeting IGF-1R in polycystic ovary syndrome
Yuan et al. microRNA‐30a inhibits the liver cell proliferation and promotes cell apoptosis through the JAK/STAT signaling pathway by targeting SOCS‐1 in rats with sepsis
Zhou et al. Let-7a is an antihypertrophic regulator in the heart via targeting calmodulin
Liu et al. LncRNA MALAT1 regulates diabetic cardiac fibroblasts through the Hippo–YAP signaling pathway
Cheng et al. MicroRNA-145 down-regulates mucin 5AC to alleviate airway remodeling and targets EGFR to inhibit cytokine expression
Huang et al. The miR‐140‐5p/KLF9/KCNQ1 axis promotes the progression of renal cell carcinoma
Yu et al. Long-chain non-coding RNA UCA1 inhibits renal tubular epithelial cell apoptosis by targeting microRNA-206 in diabetic nephropathy
Shi et al. Influence of LncRNA UCA1 on glucose metabolism in rats with diabetic nephropathy through PI3K-Akt signaling pathway.
Man et al. MiR-503 inhibits adipogenesis by targeting bone morphogenetic protein receptor 1a
Zhou et al. Obesity-induced upregulation of ZBTB7A promotes lipid accumulation through SREBP1
CN113584173B (en) Application of lncRNA SLC25A21-AS1 AS esophageal squamous cell carcinoma marker
CN108660212B (en) Application of WDR1 gene in preparation of non-small cell lung cancer treatment and detection products
Wang et al. Butyrate suppresses abnormal proliferation in colonic epithelial cells under diabetic state by targeting HMGB1
Zhao et al. MiRNA-337 leads to podocyte injury in mice with diabetic nephropathy.
Zhang et al. Knockdown of the Long Noncoding RNA LUCAT1 Inhibits High-Glucose-Induced Epithelial-Mesenchymal Transition through the miR-199a-5p–ZEB1 Axis in Human Renal Tubular Epithelial Cells
Zhao et al. miR‐101‐3p contributes to the progression of preeclampsia by suppressing WDR5‐mediated proliferation and invasion of trophoblast
Liu et al. The LIM-only protein FHL2 is involved in autophagy to regulate the development of skeletal muscle cell
Pan et al. Apolipoprotein H: a novel regulator of fat accumulation in duck myoblasts
CN113181206B (en) SiRNA of gene SLC2A9 related to hyperuricemia accompanied with hyperlipidemia and hyperglycemia treatment and application thereof
US9879258B2 (en) Treatment of insulin resistance through inhibitors of transcription factor TSC22D4
CN113913511B (en) Application of OTULINs in preparation of reagent for diagnosing and treating cilia-related diseases
Guo et al. MiR-146a upregulates FOXP3 and suppresses inflammation by targeting HIPK3/STAT3 in allergic conjunctivitis

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
GR01 Patent grant
GR01 Patent grant