CN111419860A - Glomerular lobular nephropathy modeling method - Google Patents

Glomerular lobular nephropathy modeling method Download PDF

Info

Publication number
CN111419860A
CN111419860A CN202010194856.2A CN202010194856A CN111419860A CN 111419860 A CN111419860 A CN 111419860A CN 202010194856 A CN202010194856 A CN 202010194856A CN 111419860 A CN111419860 A CN 111419860A
Authority
CN
China
Prior art keywords
adriamycin
day
ceftiofur
group
glomerular
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
CN202010194856.2A
Other languages
Chinese (zh)
Inventor
姜洪波
孙利伟
田玉玲
杜娟
刘宇奇
马英伟
贺岩
赵艳玲
张亮
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.)
CHANGCHUN CHILDREN S HOSPITAL
Original Assignee
CHANGCHUN CHILDREN S HOSPITAL
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 CHANGCHUN CHILDREN S HOSPITAL filed Critical CHANGCHUN CHILDREN S HOSPITAL
Priority to CN202010194856.2A priority Critical patent/CN111419860A/en
Publication of CN111419860A publication Critical patent/CN111419860A/en
Pending legal-status Critical Current

Links

Images

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
    • A61K31/7028Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages
    • A61K31/7034Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages attached to a carbocyclic compound, e.g. phloridzin
    • A61K31/704Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages attached to a carbocyclic compound, e.g. phloridzin attached to a condensed carbocyclic ring system, e.g. sennosides, thiocolchicosides, escin, daunorubicin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/54Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one sulfur as the ring hetero atoms, e.g. sulthiame
    • A61K31/542Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one sulfur as the ring hetero atoms, e.g. sulthiame ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/545Compounds containing 5-thia-1-azabicyclo [4.2.0] octane ring systems, i.e. compounds containing a ring system of the formula:, e.g. cephalosporins, cefaclor, or cephalexine
    • A61K31/546Compounds containing 5-thia-1-azabicyclo [4.2.0] octane ring systems, i.e. compounds containing a ring system of the formula:, e.g. cephalosporins, cefaclor, or cephalexine containing further heterocyclic rings, e.g. cephalothin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys

Abstract

The invention discloses a glomerular lobular nephropathy modeling method, which comprises the following steps: 6.5mg/kg of adriamycin is intravenously injected into a male Wistar rat body on the first day, and 30-70 mg/kg of ceftazidime is continuously injected for three days every day. The glomeruli of the obtained male Wistar rat kidney disease model appear to be lobulated. Provides a new animal kidney disease model for the research of pathophysiology mechanism, molecular cytology, molecular biology and the like of kidney diseases, and provides a basis for preventing and treating clinical patients earlier.

Description

Glomerular lobular nephropathy modeling method
Technical Field
The invention belongs to the technical field of animal model modeling, and particularly relates to a modeling method for glomerular lobular nephropathy.
Background
China is a large country with kidney diseases (nephropathy), the incidence rate of uremia is 100-. According to the disease progression rate, renal failure can be classified into Acute Renal Failure (ARF) and Chronic Renal Failure (CRF). The pathogenesis of renal failure is not clear, so no effective drug treatment exists. Research on pathogenesis of renal failure and development of safe and effective medicaments are always problems to be solved urgently in clinic. The preclinical research of drugs mainly takes the form of animal experiments of tested drugs, namely, animal models are established, and then pathogenesis or intervention (pharmaceutical) test research is carried out. For many years, renal disease researchers have developed animal models adapted to various renal diseases using rats as research subjects.
Acute renal failure refers to acute renal disease characterized by acute change of renal function within hours to days, resulting in loss of capability of kidney to discharge waste nitrogen and maintain stable function of water and electrolyte in vivo; they can be classified into pre-renal, neutral and post-renal. Prerenal refers to decreased perfusion of renal parenchyma due to various causes, renal indicates that renal parenchymal injury is present, with renal ischemia and Acute Tubular Necrosis (ATN) caused by nephrotoxic drugs or toxins being the most common, others include Acute Interstitial Nephritis (AIN), glomerular disease and renal vascular disease, etc., and obstruction can occur anywhere in the urinary tract from the renal pelvis to the urethra due to postrenal urinary tract infection.
Chronic renal failure is a long-time and progressive nephron injury, which causes dysfunction of renal excretion function, leads to retention of nitrogen metabolites caused by change of internal environment of an organism, and finally causes abnormal metabolism of the organism. The pathogenesis of the disease comprises: (1) nephron high perfusion, high filtration; (2) nephron hypermetabolism; (3) the effect of phenotypic transformation of epithelial cells of renal tissue; (4) the effects of cytokines and growth factors on promoting fibrosis; (5) and others.
Common molding methods for ARF and CRF are: physical method and chemical method, the ARF commonly used drugs include (1) glycerol type, (2) oleic acid type, (3) mercury chloride type, (4) aminoglycoside type, (5) snake venom induction, (6) sodium fluoride induction, (7) cisplatin induction, CRF commonly used drugs include adenine and adriamycin, and two drugs are used together to make animal model or two or more methods are used together. In recent years, the establishment methods of Acute Renal Failure (ARF) and Chronic Renal Failure (CRF) animal models are continuously improved, different animal models have respective characteristics and application ranges, each model cannot be completely matched with the characteristics of clinical diseases due to the influence of multiple factors, and the degree of renal injury is different. Therefore, the further exploration is more in line with the pathological change characteristics of human acute and chronic renal failure, and the search for a unified, stable and simple animal model is still a problem to be solved at present.
Adriamycin is a classic rodent nephritis injury inducer, can induce early stage focal segmental glomerulosclerosis, and can well simulate the chronic nephritis condition of a human body. Sternberg first reported in 1970 that anthracyclines could cause nephritis. The doxorubicin-induced rat nephritis model was first reported in 1976, after which the mouse doxorubicin nephritis model was reported in 1998. The doxorubicin nephritis rat model led the researchers to extensive and intensive studies to further reveal the pathological mechanism of kidney injury. Adriamycin (Adriamycin, ADR) has renal toxicity, can damage renal proximal convoluted tubule epithelial cells, and the exfoliated tubule epithelial cells, proteins and other cell components in a lumen form a tube shape to block the lumen, so that the intratubular pressure of the kidney is increased, the kidney injury is aggravated, and CRF is finally formed.
The mechanism of the antibiotic-induced kidney damage has two aspects, namely, the direct cytotoxicity effect is realized, the toxic effect is directly generated by the chemical structure of the medicine, the toxic kidney damage is caused, and the body has anaphylactic reaction to the medicine, such as acute interstitial nephritis, and the medicine is used as an antigen, so that the immune reaction is activated, and the kidney is influenced. Ceftiofur causes renal damage. It causes kidney damage parallel to the dose administered, which can cause toxic kidney damage. The drug can be injected into experimental animals through intramuscular, intravenous or intraperitoneal injection, and finally accumulated in renal tubular epithelial cells through a series of metabolism in the bodies. Due to its cytotoxic effects, it eventually leads to tubular epithelial cell injury and necrosis, inducing ARF production.
Disclosure of Invention
The invention aims to provide a glomerular lobular nephropathy modeling method.
Application of adriamycin and ceftazidime in preparation of glomerular lobular kidney disease animal model
A method for modeling glomerular lobular nephropathy, comprising: injecting adriamycin into the animal body 1); 2) injecting ceftiofur;
the injection in the step 1) is intravenous injection; the injection in the step 2) is intramuscular injection;
the animal is a rat;
the animal is a male Wistar rat;
the step 1) injecting adriamycin on the first day; 2) starting to inject ceftiofur on the next day, and continuously injecting for three days;
the adriamycin injection amount on the first day is 1.5-7.5 mg/kg; the ceftiofur is 30-70 mg/kg per day;
the adriamycin is 6.5mg/kg, and the cefotaxime is 50-70 mg/kg per day.
The invention provides a glomerular lobular nephropathy modeling method, which comprises the following steps: 6.5mg/kg of adriamycin is intravenously injected into a male Wistar rat body on the first day, and 50-70 mg/kg of ceftiofur is continuously injected for three days every day. The glomeruli of the obtained male Wistar rat kidney disease model appear to be lobulated. Provides a new animal kidney disease model for the research of pathophysiology mechanism, molecular cytology, molecular biology and the like of kidney diseases, and provides a basis for preventing and treating clinical patients earlier.
Drawings
FIG. 1 is a diagram of the steps of animal experiment; 1. weighing; 2. anaesthetizing; 3. disinfecting the skin at the tail part; 4. tail vein injection; 5. sterilizing; 6. intramuscular injection; 7. cutting open the abdominal cavity; 8. isolating the abdominal aorta; 9. taking blood from abdominal aorta; 10. taking out the kidney;
FIG. 2A-C are standard curves of phosphorus content;
FIG. 3D-F sets of standard curves for phosphorus content;
FIG. 4 microscopic observation of rat kidney section results; a-1, magnifying by 100 times and observing a normal control group; a-2, magnifying by 400 times and observing a normal control group; b-1, observing a doxorubicin 6.5mg/kg dose group under 100-fold magnification; b-2, observing a doxorubicin 6.5mg/kg dose group under 400-fold magnification; c-1, a group with 100 times of magnification of 60mg/kg of ceftazidime; c-2, a 400-fold magnification ceftazidime 60mg/kg dose group;
FIG. 5 microscopic observation of rat kidney section results; d-1, observing a dosage group of 6.5mg/kg of adriamycin and 30mg/kg of ceftazidime under the magnification of 100 times; d-2, observing a dosage group of 6.5mg/kg of adriamycin and 30mg/kg of ceftazidime under 400 times of magnification; e-1, observing a dosage group of 6.5mg/kg of adriamycin and 50mg/kg of ceftazidime under the magnification of 100 times; e-2, observing a dosage group of 6.5mg/kg of adriamycin and 50mg/kg of ceftazidime under 400-fold magnification; f-1, observing a dosage group of 6.5mg/kg of adriamycin and 70mg/kg of ceftazidime under the magnification of 100 times; f-2, observing the adriamycin 6.5mg/kg and ceftazidime 70mg/kg dosage group under 400-fold magnification.
Detailed Description
Example 1 Adriamycin in combination with ceftiofur in establishing rat model of chronic renal failure
1. Animal selection and grouping
Rats were selected because most species of rats were very sensitive to doxorubicin kidney damage, male Wistar rats required an doxorubicin dose in the range of 1.5-7.5 mg/kg.ba L B/c mice of 9.8-10.4 mg/kg, male BA L B/c SCID mice required only 5.3 mg/kg.c57b L/c mice highly resistant to doxorubicin-induced kidney damage, but high doses were able to induce kidney damage.
Grouping: a: a normal control group; b: doxorubicin at 6.5mg/kg dose group; c: ceftiofur 60mg/kg dose group; d: a 6.5mg/kg doxorubicin + 30mg/kg ceftiofur dose group; e: doxorubicin at 6.5mg/kg + ceftiofur 50 mg/kg; f: 6.5mg/kg of adriamycin and 70mg/kg of ceftazidime; there were 6 groups of 3, 18 each.
2. Drug selection
Doxorubicin is an anthracycline antibiotic, which is rapidly cleared from the plasma and significantly associated with tissue, primarily metabolized by the liver, after entering the body. 4 to 5 percent of adriamycin can be excreted by human body through urine 5d, and 40 to 50 percent of the administration amount can be excreted through bile 7 d. After intravenous administration in rats and mice, doxorubicin is rapidly cleared from plasma, deposited in tissues, and slowly excreted via urine and bile. Doxorubicin is slowly metabolized and accumulates primarily in the kidney. Ceftiofur is a first-generation cephalosporin, is a pyridine derivative of cephalothin, has stronger effect on gram-positive bacteria than the former and also has stronger effect on escherichia coli, is mainly used for respiratory tract infection, skin and soft tissue infection, urinary tract infection, biliary tract infection and thoracic cavity and abdominal cavity infection caused by sensitive bacteria, and can also be used for meningitis, septicemia and pleurisy. After injection, the blood concentration is higher than that of cephalothin, and the excretion is slower. There was significant nephrotoxicity, which parallels the dose administered to the kidney. Doxorubicin and ceftiofur have certain complementary action in vivo metabolism, can both generate residual renal toxicity, are combined to prepare the renal failure animal model, have definite and stable effect, can be said to be short of the time of simple doxorubicin magic property, and are reported at home and abroad.
3. Animal modeling
The animal rooms alternate day and night for 12h to 12h, the animals are kept drinking water and eating freely at the temperature of 23 ℃ to 25 ℃, the animals are raised in a metabolism cage, and the experimental animals enter the animal rooms for adaptive feeding for one week and then enter the experiment; the rats were weighed and anesthetized, placed flat on a laboratory bench, with the left thumb and index finger pinching the two sides of the tail to fill the vein, the middle finger was used to hold the tail from below, the ring finger was used to clamp the tip of the tail, and the right tail vein was selected for injection. B. D, E, F group is administered by tail vein injection with adriamycin once at a dose of 6.5 mg/kg; the experimental mouse is fixed the next day after tail vein injection, one hind limb is selected to be placed at the muscle part at the outer side of thigh, the skin surface is disinfected by 75% alcohol, the right hand-held injector forms 60 degrees with muscle, muscle is quickly punctured, and muscle is pumped back to carry out intramuscular injection after no backflow matter exists. C. D, E, F the ceftiofur is injected into the groups according to the dosage of 60mg/kg, 30mg/kg, 50mg/kg and 70mg/kg respectively for three days; normal group a rats were given equal volumes of saline tail vein or intramuscular injection.
Collecting a specimen: collecting urine 3 days, 5 days, 7 days and 2 weeks after intramuscular injection, calculating urine volume, and detecting urine protein volume; the rats were drunk with 28 Gastrodia elata on 14 days, and the femoral artery was sampled (operation process).
Example 2 detection of urine protein content
Taking urine: feeding mice in a metabolism cage, normally providing water and feed, collecting and calculating 24h urine of each rat on the 3 rd day, the 7 th day and the second week after intramuscular injection, and collecting the urine for biochemical detection; the grouping is as follows:
1) normal control group: a3, A7, A14
2) Doxorubicin 6.5mg/kg group: b3, B7, B14
3) Ceftiofur 60mg/kg group: c3, C7, C14
4) Doxorubicin 6.5mg/kg + ceftiofur 30mg/kg group: d3, D7, D14
5) Doxorubicin 6.5mg/kg + ceftiofur 50mg/kg group: e3, E7, E14
6) Doxorubicin 6.5mg/kg + ceftiofur 70mg/kg group: f3, F7, F14
3. 7, 14 represent 3 day, 7 day, 14 day time points, respectively; as a result, it was found (Table 1) that the urine protein content of the group containing doxorubicin at 6.5mg/kg and ceftiofur at 60mg/kg increased as the drug action time extended, as compared with the normal control group; compared with the group with single action of adriamycin and ceftazidime, the group with combined action of adriamycin and ceftazidime has increased urine protein content.
Figure DEST_PATH_IMAGE002
Example 3 serum marker detection
1. Grouping of rats
1) Normal control group: a14, A28
2) Doxorubicin 6.5mg/kg group: b14 and B28
3) Ceftiofur 60mg/kg group: c14, C28
4) Doxorubicin 6.5mg/kg + ceftiofur 30mg/kg group: d14 and D28
5) Doxorubicin 6.5mg/kg + ceftiofur 50mg/kg group: e14, E28
6) Doxorubicin 6.5mg/kg + ceftiofur 70mg/kg group: f14, F28;
14. 28 represent 14 day, 28 day time points, respectively;
2. serum creatinine assay
Under alkaline conditions, creatinine (Cr) in serum reacts with picric acid to produce an orange-colored picric acid creatinine complex. Detecting the change of absorbance at the wavelength of 505nm to obtain the concentration of creatinine in the detected serum; creatinine (CR) assay kit (Nanjing institute) was used; adding 0.2ml of serum into 2ml of tungstic acid protein testing agent, fully and uniformly mixing, rotating/min at 3500 rpm, centrifuging for 10 minutes, taking supernatant, and testing according to the following table;
Figure DEST_PATH_IMAGE004
mixing, water bathing at 37 deg.C for 10min, cooling with running water, and measuring absorbance at 510nm with enzyme-labeling instrument;
calculating the formula:
Figure DEST_PATH_IMAGE006
3. urea nitrogen detection
Urea is hydrolyzed under the action of urease to generate ammonia ions and carbon dioxide, the ammonia ions and a phenol color developing agent generate a blue substance in an alkaline medium, the generation amount of the substance is in direct proportion to the content of the urea, and the color comparison is carried out at the wavelength of 640 nm.
The operation steps are as follows:
Figure DEST_PATH_IMAGE008
mixing, water bathing at 37 deg.C for 10min, wavelength of 640nm, light path of 1cm, adjusting to zero with double distilled water, and measuring absorbance
Calculating the formula:
Figure DEST_PATH_IMAGE010
4. serum potassium detection
In an alkaline medium, reacting potassium ions in the sample treated by the protein precipitator with NA-TPB to generate turbid and stable suspension; the turbidity is in direct proportion to the concentration of potassium ions in the sample; detecting with potassium (K) test kit (built in Nanjing); taking 20ul serum and 180ul precipitator (prepared in situ), fully and uniformly mixing, 3500 rpm/min, centrifuging for 10min, and taking 50ul supernatant for determination; the operation steps are as follows:
Figure DEST_PATH_IMAGE012
mixing, standing for 5min at wavelength of 460nm and light path of 1cm, adjusting to zero with double distilled water, and measuring absorbance of each tube, wherein the calculation formula is as follows:
Figure DEST_PATH_IMAGE014
5. serum calcium detection
Calcium ions in the sample are combined with Methyl Thymol Blue (MTB) in an alkaline solution to generate a blue complex; comparing the color with a calcium standard treated in the same way to calculate the content of calcium in the sample; detecting with calcium content determination kit (Nanjing kit); serum procedure is as follows:
Figure DEST_PATH_IMAGE016
uniformly mixing, standing for 5 minutes, carrying out color comparison by an enzyme-labeling instrument at the wavelength of 610nm, and measuring the OD value of each hole; calculating the formula:
Figure DEST_PATH_IMAGE018
6. serum phosphorus detection
Inorganic phosphorus in the sample reacts with molybdic acid to generate phosphomolybdic acid, the phosphomolybdic acid is reduced to molybdenum blue, a maximum absorption peak is formed at 660nm, and the content of the inorganic phosphorus can be calculated through colorimetry; detecting with phosphorus content determination kit (Nanjing kit); taking 0.1ml of serum and 0.4ml of precipitator, fully and uniformly mixing, carrying out 3500 rpm centrifugation for 10 minutes, and taking the supernatant to be tested; the operation steps are as follows:
Figure DEST_PATH_IMAGE020
mixing, water bathing at 37 deg.C for 30 min, cooling to room temperature, wavelength of 660nm, optical path of 1cm, adjusting to zero with double distilled water, and measuring absorbance of each tube;
preparing and calculating standard curve by diluting 10 mmol/L phosphorus standard stock solution with deionized water to different concentrations (0.0625, 0.125, 0.25, 0.5, 1, 2 mmol/L), preparing standard curve according to operation table, drawing standard curve with the concentration of standard substance as abscissa and OD value as ordinate, and calculating linear regression equation y =0.4475x +0.006 (R) of standard curve by using the concentration of standard substance and OD value2= 0.9945), the OD value of the sample is substituted into the equation to calculate the concentration of the sample, and then the sample concentration is multiplied by the dilution factor to obtain the actual concentration of the sample, the OD value of the sample is measured, and the content of phosphorus (mmol/L) in the sample is calculated according to a standard curve, wherein the standard curve of the phosphorus content is shown in figure 2 and figure 3;
7. results
The experimental results are shown in Table 3, and it can be seen from the table that, compared with the normal control group, the serum creatinine, urea nitrogen, blood potassium and phosphorus contents of the adriamycin group of 6.5mg/kg and the ceftiofur group of 60mg/kg are increased, and the calcium content is reduced; the content of serum creatinine, urea nitrogen, blood potassium and phosphorus is continuously increased along with the prolonging of the action time of the medicine, and the content of calcium is reduced; compared with the group with simple action of the adriamycin and the ceftazidime, the group with combined action of the adriamycin and the ceftazidime has the advantages that the serum creatinine, the urea nitrogen, the serum potassium and the serum phosphorus content are increased, the calcium content is reduced, and the index values of the group with 6.5mg/kg of the adriamycin and 70mg/kg of the ceftazidime are increased or reduced the most.
Figure DEST_PATH_IMAGE022
Example 4 routine pathological examination of rat kidney
Anesthetizing rats in each group (A-F) on day 28, cutting off abdominal cavities, taking kidney tissues of the rats, fixing and embedding the kidney tissues, cutting the kidney tissues into paraffin sections with the thickness of 3 mu m, staining the kidney tissues by hematoxylin-eosin (HE), and observing pathological morphological changes of glomeruli and tubulointerstitial tissues under a light microscope; the method comprises the following specific steps:
1. tissue paraffin preparation
1) After the material was taken, the material was washed with PBS and fixed with 4% paraformaldehyde.
2) And (3) dehydrating: soaking the tissue in 75% ethanol, 85% ethanol, 95% ethanol I, 95% ethanol II, 100% ethanol I and 100% ethanol II for 10min each;
3) and (3) transparency: soaking the dehydrated tissue in 1/2 xylene, xylene I and xylene II for 10 min;
4) wax penetration: soaking the transparent treated tissue in melted paraffin for 3 h;
5) embedding;
6) slicing: slicing the tissue in the paraffin block into thin slices with the thickness of 5um by using a slicing machine, and flatly paving the thin slices on an anti-falling glass sheet;
7) baking slices: the slices were placed on a 55 ℃ slide-baking machine to allow the tissue pieces to cling to the anti-detachment slides.
2. Paraffin section dewaxing rehydration
1) Dewaxing: soaking the paraffin sections in xylene I, xylene II and 1/2 xylene respectively for 10 min;
2) rehydration: soaking the dewaxed paraffin section in 100% ethanol I, 100% ethanol II, 95% ethanol I, 95% ethanol II, 85% ethanol, and 75% ethanol for 5min respectively;
3) cleaning with double distilled water for 2min for 3 times;
3. HE staining
1) Dyeing with hematoxylin dye liquor for 10 min;
2) washing the redundant dyeing solution with tap water for about 5 min; washing with distilled water again (for several seconds);
3) performing microscopic examination, and decolorizing with 1% hydrochloric acid alcohol if hematoxylin is stained too deeply, and removing excessive hematoxylin staining solution in cytoplasm;
4) dyeing with eosin dye liquor for 30 s;
5) washing the excessive dye liquor with tap water for about 3 min; washing with distilled water again (i.e. microscopic examination);
4. dehydration seal
Dehydrating with 95% ethanol for 2min, replacing with fresh 95% ethanol, and dehydrating for 2 min; the xylene is transparent for 5min, fresh xylene is used for replacement, and the xylene is transparent for 5min again; sealing the neutral resin; microscopic examination, the nucleus turns blue, and the cytoplasm turns red or pink;
the experimental result shows that the kidney shape of the normal control group is normal, the kidney structure is clear, and the glomerulus is regular; compared with a normal control group, glomerular mesangial cells of an adriamycin group of 6.5mg/kg and a ceftazidime group of 60mg/kg are proliferated, and a basement membrane is thickened; part of glomeruli of the adriamycin and ceftiofur combined action group are lobulated, plasma proteins in renal capsules exude, and lesions have certain changes along with the increase of the concentration of the ceftiofur.

Claims (8)

1. Application of adriamycin and ceftazidime in preparation of glomerular lobular kidney disease animal models.
2. A method for modeling glomerular lobular nephropathy, comprising: injecting adriamycin into the animal body 1); 2) ceftiofur is injected.
3. The method of claim 1, wherein the method comprises: the injection in the step 1) is intravenous injection; the injection in the step 2) is intramuscular injection.
4. The method of claim 3, wherein the method comprises: the animal is a rat.
5. The method of claim 3 or 4, wherein the method comprises: the animal is a male Wistar rat.
6. The method of claim 5, wherein the method comprises: the step 1) injecting adriamycin on the first day; 2) ceftiofur was injected starting the next day for three consecutive days.
7. The method of claim 6, wherein the method comprises: the adriamycin injection amount on the first day is 1.5-7.5 mg/kg; the ceftiofur is 30-70 mg/kg per day.
8. The method of claim 7, wherein the method comprises: the adriamycin is 6.5mg/kg, and the cefotaxime is 50-70 mg/kg per day.
CN202010194856.2A 2020-03-19 2020-03-19 Glomerular lobular nephropathy modeling method Pending CN111419860A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202010194856.2A CN111419860A (en) 2020-03-19 2020-03-19 Glomerular lobular nephropathy modeling method

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202010194856.2A CN111419860A (en) 2020-03-19 2020-03-19 Glomerular lobular nephropathy modeling method

Publications (1)

Publication Number Publication Date
CN111419860A true CN111419860A (en) 2020-07-17

Family

ID=71548314

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202010194856.2A Pending CN111419860A (en) 2020-03-19 2020-03-19 Glomerular lobular nephropathy modeling method

Country Status (1)

Country Link
CN (1) CN111419860A (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN114190329A (en) * 2021-11-19 2022-03-18 华中农业大学 Method for establishing canine acute renal failure model

Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101686991A (en) * 2007-05-25 2010-03-31 韦罗制药有限公司 Compositions useful for reducing nephrotoxicity and methods of use thereof
CN102485255A (en) * 2010-12-02 2012-06-06 上海海天医药科技开发有限公司 Compound preparation used for preventing and treating nephrotic syndrome hypoproteinemia, preparation method thereof, and application thereof
CN104095865A (en) * 2014-07-16 2014-10-15 樊均明 CKD (chronic kidney disease) animal model establishment
CN106975070A (en) * 2008-02-26 2017-07-25 康奈尔大学 Method for preventing and treating acute injury of kidney
WO2018071701A1 (en) * 2016-10-13 2018-04-19 University Of Leicester Methods for reducing proteinuria in a human subject suffering from immunoglobulin a nephropathy
CN108495652A (en) * 2016-01-05 2018-09-04 莱斯特大学 Method for the fibrosis for inhibiting subject in need
CN110301398A (en) * 2019-07-03 2019-10-08 山西大学 A kind of construction method of mRNA IN ADRIAMYCIN NEPHROPATHY syndrome rat model

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101686991A (en) * 2007-05-25 2010-03-31 韦罗制药有限公司 Compositions useful for reducing nephrotoxicity and methods of use thereof
CN106975070A (en) * 2008-02-26 2017-07-25 康奈尔大学 Method for preventing and treating acute injury of kidney
CN102485255A (en) * 2010-12-02 2012-06-06 上海海天医药科技开发有限公司 Compound preparation used for preventing and treating nephrotic syndrome hypoproteinemia, preparation method thereof, and application thereof
CN104095865A (en) * 2014-07-16 2014-10-15 樊均明 CKD (chronic kidney disease) animal model establishment
CN108495652A (en) * 2016-01-05 2018-09-04 莱斯特大学 Method for the fibrosis for inhibiting subject in need
WO2018071701A1 (en) * 2016-10-13 2018-04-19 University Of Leicester Methods for reducing proteinuria in a human subject suffering from immunoglobulin a nephropathy
CN110301398A (en) * 2019-07-03 2019-10-08 山西大学 A kind of construction method of mRNA IN ADRIAMYCIN NEPHROPATHY syndrome rat model

Non-Patent Citations (12)

* Cited by examiner, † Cited by third party
Title
ATKINSON R.M.,等: "Acute toxicity of cephaloridine, an antibiotic derived from cephalosporin C", 《TOXICOLOGY AND APPLIED PHARMACOLOGY》 *
刘练,等: "阿霉素肾病小鼠的肾脏病理转变过程", 《中国实验动物学报》 *
包玉龙,等: "阿霉素大鼠肾病综合征模型最佳剂量的筛选阿霉素大鼠肾病综合征模型最佳剂量的筛选", 《动物医学进展》 *
姜洪波,等: "阿霉素联合头孢噻啶致大鼠慢性肾衰模型的建立及意义", 《中国实验诊断学》 *
孟凡翠,等: "药物重要脏器毒性及其早期预测方法的探讨", 《药物评价研究》 *
庞新路,等主编: "《现代肾脏病学理论与应用》", 30 June 2013, 河北科学技术出版社 *
徐积恩: "阿霉素诱发的肾病作为慢性进行性肾小球疾病的实验模型", 《国外药学(抗生素分册)》 *
李佳璐,等: "大鼠肾衰模型造模方法及比较研究进展", 《中国比较医学杂志》 *
杜宇,等: "阿霉素大鼠肾病模型的概", 《中国医药指南》 *
郭勇,等: "缬沙坦对肾小球硬化大鼠肾组织单核细胞趋化蛋白-1表达的影响", 《新乡医学院学报》 *
阎晓彩: "自由基与肾脏病的关系", 《陕西医学杂志》 *
韩峰,主编: "《药物毒理学》", 29 February 2020, 华中科技大学出版社 *

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN114190329A (en) * 2021-11-19 2022-03-18 华中农业大学 Method for establishing canine acute renal failure model
CN114190329B (en) * 2021-11-19 2024-02-09 华中农业大学 Method for establishing acute renal failure model of dog

Similar Documents

Publication Publication Date Title
Inada et al. The etiology, mode of infection, and specific therapy of Weil's disease (spirochaetosis icterohaemorrhagica)
CN111419860A (en) Glomerular lobular nephropathy modeling method
CN103891784A (en) Bacteriostatic agent for sterilization and disinfection, skin mucosa disinfection fluid and preparation method thereof
Jaturakan et al. Combination of vitamin E and vitamin C alleviates renal function in hyperoxaluric rats via antioxidant activity
Han et al. Immunomodulatory effect of Lycium barbarum polysaccharides against liver fibrosis based on the intelligent medical internet of things
Garrett et al. Renal malakoplakia. Experimental production and evidence of a link with interstitial megalocytic nephritis
CN101095743B (en) Exterior-applied medicine composition for treating gynecologic diseases and method of making the same and the use thereof
Saleh et al. Toxico-pathological study of gentamicin by intramuscular injection in experimental rabbits
CN108498546B (en) Traditional Chinese medicine composition for preventing or treating ulcerative colitis canceration and application
CN107007610B (en) Application of benzophenone compound in pharmacy
Molomut et al. A lymphocytopenic filterable agent derived from tissue cell cultures of murine carcinoma
Jonczyk et al. Rare cases of bezoars in the urinary tract of diabetic patients—a review of case reports
Kowsalya et al. Hepatoprotective activity of ethyl acetate of Mimosa pudica leaves against paracetamol induced in albino wister rats
CN114190329B (en) Method for establishing acute renal failure model of dog
Hanna et al. Impact of Fractionated Gentamicin Dosing on Induction of Nephrotoxicity: An Experimental Study in Rats.
Akman et al. Henoch-Schoenlein purpura and retroperitoneal fibrosis
Prakash et al. Sugar mill effluent induced histological changes in kidney of Channa punctatus
CN102210725B (en) Application of hypericum japonicum thunb general flavone in preparing medicament for treating hepatic fibrosis
Jameel et al. Protective role of gum arabic (Acacia Senegal) against adenine induced nephrotoxicity in male albino rats: Some histological, histochemical, and biochemical studies
TWI780460B (en) A use of manufacturing a composition of lactobacillus paracasei gks6 for preventing and treating the renal function impairment
CN114796107B (en) Tobramycin sulfate injection containing nicotin diphosphate and preparation method thereof
CN110101759B (en) Traditional Chinese medicine composition for treating acute kidney injury and application thereof
Oviosun et al. THERAPEUTIC PROPERTIES OF FICUS EXASPERATA EXTREACT ON GENTAMICIN INDUCED KIDNEY DAMAGE
EA010427B1 (en) Preparation for the treatment of prostrate gland diseases
CN115634245A (en) Veterinary drug composition, decoction thereof, preparation method and application

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
WD01 Invention patent application deemed withdrawn after publication

Application publication date: 20200717

WD01 Invention patent application deemed withdrawn after publication