WO2015172746A1 - Use of ginsenoside m1 for inhibiting renal fibrosis - Google Patents

Use of ginsenoside m1 for inhibiting renal fibrosis Download PDF

Info

Publication number
WO2015172746A1
WO2015172746A1 PCT/CN2015/079156 CN2015079156W WO2015172746A1 WO 2015172746 A1 WO2015172746 A1 WO 2015172746A1 CN 2015079156 W CN2015079156 W CN 2015079156W WO 2015172746 A1 WO2015172746 A1 WO 2015172746A1
Authority
WO
WIPO (PCT)
Prior art keywords
renal
ginsenoside
subject
fibrosis
uuo
Prior art date
Application number
PCT/CN2015/079156
Other languages
French (fr)
Inventor
Sheau-Long Lee
Yu-Chieh Lee
Ann Chen
Kuo-Feng Hua
Shuk-Man Ka
Original Assignee
Wellhead Biological Technology Corp.
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 Wellhead Biological Technology Corp. filed Critical Wellhead Biological Technology Corp.
Priority to KR1020167035138A priority Critical patent/KR102360315B1/en
Priority to CN201580019639.7A priority patent/CN106795546A/en
Priority to DK15793585.9T priority patent/DK3146062T3/en
Priority to US15/311,683 priority patent/US10357507B2/en
Priority to ES15793585T priority patent/ES2739203T3/en
Priority to JP2017512092A priority patent/JP6626094B2/en
Priority to EP15793585.9A priority patent/EP3146062B1/en
Publication of WO2015172746A1 publication Critical patent/WO2015172746A1/en

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
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0053Mouth and digestive tract, i.e. intraoral and peroral administration
    • 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
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12PFERMENTATION OR ENZYME-USING PROCESSES TO SYNTHESISE A DESIRED CHEMICAL COMPOUND OR COMPOSITION OR TO SEPARATE OPTICAL ISOMERS FROM A RACEMIC MIXTURE
    • C12P19/00Preparation of compounds containing saccharide radicals
    • C12P19/44Preparation of O-glycosides, e.g. glucosides

Definitions

  • the present invention relates to a new use of ginsenoside M1 for inhibiting renal fibrosis, particularly interstitial fibrosis.
  • Renal fibrosis is the final stage of renal diseases.
  • the causes of renal fibrosis include ureteral obstruction.
  • Ureteral obstruction is one of the most common problems confronting the urologist. Ureteral obstruction causes impedance to the flow of urine in the ureter. Most commonly, obstruction occurs at the ureteropelvic junction.
  • the broader term “obstructive uropathy” can be used to indicate any obstruction to urinary flow occurring between the renal pelvis and the urethra, which causes a developing hydronephrosis and associated renal impairment. Urethral strictures and benign prostatic hypertrophy are cases in point.
  • the incidence of unilateral ureteric obstruction (UUO) is reported as 1/1,000 in adults. Ureteric calculi are the most common cause, and acute obstruction typically presents with significant renal colic as the major symptom.
  • TGF- ⁇ 1 TGF- ⁇ 1
  • TGF- ⁇ 1 TGF- ⁇ 1
  • TGF- ⁇ 1 TGF- ⁇ 1
  • EMT epithelial–mesenchymal transition
  • Angiotensin II (ANG II) , produced by the activation of monocytes, stimulates the production of nuclear factor- ⁇ B (NF- ⁇ B) , which leads to the recruitment of more macrophages, as well as to the production of reactive oxygen species (ROS) , which aggravates renal tubular injury.
  • ROS reactive oxygen species
  • endothelial cells can undergo endothelial–mesenchymal transition (EndMT) or apoptosis, which leads to capillary loss and secondary renal ischemia and hypoxia. Resident pericytes and infiltrating hematopoietic stem cells can also differentiate into fibroblasts.
  • cytokines such as TGF- ⁇ 1 produced by macrophages or other cells
  • fibroblasts synthesize stress fibers and undergo further differentiation to become myofibroblasts.
  • the myofibroblasts are contractile and augment the deposition of the extracellular matrix (ECM) , leading to progressive interstitial fibrosis.
  • ECM extracellular matrix
  • This process finally leads to progressive renal fibrosis and irreversible renal failure. See, for example, Chevalier et al., Ureteral obstruction as a model of renal interstitial fibrosis and obstructive nephropathy, Kidney Int. 2009 Jun; 75 (11) : 1145-52.
  • Ginsenosides the main active ingredients of ginseng, are known to have a variety of pharmacological activities, e.g. antitumor, antidiabetic, antifatique, antiallergic and antioxidant activities. Ginsenosides share a basic structure, composed of gonane steroid nucleus having 17 carbon atoms arranged in four rings. Ginsenosides are metalized in the body, and a number of recent studies suggest that ginsenoside metabolites, rather than naturally occurring ginsenosides, are readily absorbed in the body and act as the active components.
  • pharmacological activities e.g. antitumor, antidiabetic, antifatique, antiallergic and antioxidant activities.
  • Ginsenosides share a basic structure, composed of gonane steroid nucleus having 17 carbon atoms arranged in four rings. Ginsenosides are metalized in the body, and a number of recent studies suggest that ginsenoside metabolites, rather than naturally
  • ginsenoside M1 is known as one metabolite of protopanaxadiol-type ginsenosides via the gypenoside pathway by human gut bacteria.
  • ginsenoside M1 is known as one metabolite of protopanaxadiol-type ginsenosides via the gypenoside pathway by human gut bacteria.
  • no prior art references report the effect of ginsenoside M1 in treatment of renal fibrosis.
  • the present invention provides a new approach for treatment or prevention of renal fibrosis in a subject.
  • the present invention provides a method for inhibiting renal fibrosis in a subject in need thereof comprising administering an effective amount of ginsenoside M1 to the subject.
  • the subject is a patient with an obstructive nephropathy.
  • the method of the invention is effective in reducing or alleviating one or more symptoms or conditions, including but not limited to, mononuclear leukocyte inflammation, tubular dilation, tubular atrophy, proliferation of tubular epithelial cells, activation of fibroblast or myofibroblast and deposition of collagens (e.g. III or IV) , particularly in the tubulointerstitial compartment of the subject.
  • one or more symptoms or conditions including but not limited to, mononuclear leukocyte inflammation, tubular dilation, tubular atrophy, proliferation of tubular epithelial cells, activation of fibroblast or myofibroblast and deposition of collagens (e.g. III or IV) , particularly in the tubulointerstitial compartment of the subject.
  • the method of the invention is effective in reducing or alleviating mononuclear leukocyte infiltration or fibrosis in renal interstitial in the subject.
  • the ginsenoside M1 is administered by parenteral or enteral route.
  • the ginsenoside M1 is administered prior to or at the time of occurrence of renal fibrosis.
  • the ginsenoside M1 is administered after occurrence of renal fibrosis.
  • ginsenoside M1 for manufacturing a medicament for treating renal fibrosis.
  • Fig. 1 shows the establishment of the UUO animal model, including (A) the experimental procedure, (B) the surgical procedure of the UUO animal model wherein the left is the diseased kidney after ligation of ureter and the right is the normal kidney without ligation of ureter.
  • Fig. 2 shows the kidney histopathological evaluation by H&E staining wherein the arrows indicate dilated tubule.
  • A H&E staining for renal histopathological evaluation. “#” indicates dilated renal tubule, asterisk indicates monocytes, and “*” indicates atrophic renal tubule. The original magnification was x400 for histopathology.
  • B Semi-quantitative shows tubulointerstitial injury scores (TIS) . White, black and gray columns represent normal control mice, disease-control mice and LCHK168+UUO mice, respectively. ***p ⁇ 0.005.
  • Fig. 3 shows renal cell apoptosis and proliferation evaluation, including (A) detection of PCNA by immunohistochemical staining wherein the arrows indicate positively stained cells, and (B) semi-quantitative results.
  • Original magnification is ⁇ 400.
  • White, black and gray columns represent normal control mice, disease-control mice and LCHK168+UUO mice, respectively. ***p ⁇ 0.005.
  • Fig. 4 shows fibrosis expression evaluation, including (A) expression of fibrotic collagens in renal tissues by Masson’s trichome staining, (B) expression of smooth muscle ⁇ -actin ( ⁇ -SMA) in renal tissues by immunohistochemical staining, (C) expression of collagen III in renal tissues by immunohistochemical staining, (D) expression of collagen IV in renal tissues by immunohistochemical staining, wherein the arrows indicate positively stained cells, and (E) semi-quantitative results.
  • Original magnification is ⁇ 400.
  • White, black and gray columns represent normal control mice, disease-control mice and LCHK168+UUO mice, respectively. *p ⁇ 0.05, **p ⁇ 0.01, ***p ⁇ 0.005.
  • Fig. 5 shows (A) expression of NF- ⁇ B in renal tissues by immunohistochemical staining, wherein the arrows indicate positively stained cells, and (B) semi-quantitative results.
  • Original magnification is ⁇ 400.
  • White, black and gray columns represent normal control mice, disease-control mice and LCHK168+UUO mice, respectively. ***p ⁇ 0.005.
  • Fig. 6 shows mononuclear leukocyte infiltration in renal tissues, including detection of (A) F4/80 + monocytes/macrophages and (B) CD3 + T cells by immunohistochemical staining, wherein the arrows indicate positively stained cells, and (C) semi-quantitative results.
  • Original magnification is ⁇ 400.
  • White, black and gray columns represent normal control mice, disease-control mice and LCHK168+UUO mice, respectively. ***p ⁇ 0.005.
  • Fig. 7 shows that LCHK168 inhibits the expression level of MCP-1, TNF- ⁇ and IL-1 ⁇ in pelvis urine.
  • MCP-1, TNF- ⁇ , and IL-1 ⁇ are detected in urine samples by ELISA.
  • White, black and gray columns represent normal control mice, disease-control mice and LCHK168+UUO mice, respectively. *p ⁇ 0.05, **p ⁇ 0.01, ***p ⁇ 0.005.
  • Fig. 8 shows evaluation for activation of cellular immunity, including detection of (A) CD3 + CD69 + T cells and (B) CD19 + CD69 + T cells by flow cytometry.
  • White, black and gray columns represent normal control mice, disease-control mice and LCHK168+UUO mice, respectively.
  • Fig. 9 shows evaluation of activation of hematopoietic stem cells, including detection of (A) CD34 + stem cells and (B) Sca-1 + hematopoietic stem cells by flow cytometry.
  • White, black and gray columns represent normal control mice, disease-control mice and LCHK168+UUO mice, respectively.
  • an element means one element or more than one element.
  • ginsenoside M1 is effective in inhibiting renal fibrosis.
  • the renal damage/inflammation is reduced and the progression to renal fibrosis is inhibited or alleviated.
  • the surgical-intervented UUO model on rodents has been used for decades as a high-throughput experimental model for UUO.
  • This model is not complicated by hypertension, proteinuria or hyperlipidemia, and does not involve any apparent immune or toxic renal insult.
  • UUO can mimic various stages of obstructive nephropathy that ultimately lead to tubulointerstitial fibrosis.
  • the animal’s lifespan is not compromised, because the function of the contralateral kidney is maintained or even increased as a result of compensatory functional and anatomic hypertrophy.
  • Other Advantages of this model include the fact that the progression of renal fibrosis is highly predictable and reproducible and leads to significant fibrosis and nephron loss in a relatively short period (7–14 days) .
  • ginsenoside M1 shows the unexpected effects of ginsenoside M1, including (1) a reduction in cell apoptosis and tubular damage in renal interstitial tissues (2) prevention of fibrosis and collage deposition in renal interstitial tissues, (3) a reduction of NF- ⁇ B activation, (4) prevention of macrophage and T cell infiltration of the kidney, and (5) maintenance of normal function of hematopoietic stem cell and systemic cellular immunity.
  • the present invention provides a new approach for inhibition of renal fibrosis in a subject in need by administering an effective amount of ginsenoside M1 to the subject.
  • Ginsenoside M1 20-O- ⁇ -D-glucopyranosyl-20 (S) -protopanaxadiol, is one of saponin metabolites known in the art.
  • the chemical structure of ginsenoside M1 is as follows:
  • Ginsenoside M1 is known as one metabolite of protopanaxadiol-type ginsenosides via the gypenoside pathway by human gut bacteria. Ginsenoside M1 can be found in blood or urine after intake. Ginsenoside M1 may be prepared from ginseng plants through fungi fermentation by methods known in the art, such as Taiwan Patent Application No. 094116005 (I280982) and U.S. Patent No. 7,932,057, the entire content of which is incorporated herein by reference. In certain embodiments, the ginseng plants for preparing the ginsenoside M1 include Araliaceae family, Panax genus, e.g. P. ginseng and P.
  • the method of preparation of ginsenoside M1 includes the steps of (a) providing powder of ginseng plant materials (e.g. leaves or stems) ; (b) providing a fungus for fermenting the ginseng plant materials, wherein the fermentation temperature is ranged from 20-50°C., the fermentation humidity is ranged from 70-100%, the pH value is ranged from 4.0-6.0, and the fermentation period is ranged from 5-15 days; (c) extracting and collecting the fermentation products; and (d) isolating 20-O- ⁇ -D-glucopyranosyl-20 (S) -protopanaxadiol from the fermentation products.
  • ginseng plant materials e.g. leaves or stems
  • ginsenoside M1 When ginsenoside M1 is described as “isolated” or “purified” in the present invention, it should be understood as not absolutely isolated or purified, but relatively isolated or purified.
  • purified ginsenoside M1 refers to one that is more purified compared to its naturally existing form.
  • a preparation comprising purified ginsenoside M1 may comprise ginsenoside M1 in an amount of more than 50%, more than 60%, more than 70%, more than 80%, more than 90%, or 100% (w/w) of the total preparation. It should be understood that when a certain number was used herein to show a ratio or dosage, said number generally includes that within the range of 10%more and less, or more specifically, the scope of 5%more and less than the number.
  • the present invention provides a method for inhibiting renal fibrosis comprising administering an effective amount of ginsenoside M1 to a subject in need thereof. Also provided is use of ginsenoside M1 for manufacturing a medicament for inhibiting renal fibrosis in a subject in need thereof.
  • the medicament of the invention is effective in reducing or alleviating one or more symptoms or conditions, including but not limited to, mononuclear leukocyte inflammation, tubular dilation, tubular atrophy, proliferation of tubular epithelial cells, activation of fibroblast or myofibroblast and deposition of collagens (e.g. III or IV) , particularly in the tubulointerstitial compartment of the subject.
  • the medicament of the invention is effective in reducing or alleviating mononuclear leukocyte infiltration or fibrosis in renal interstitial in the subject.
  • the term “individual” or “subject” used herein includes human and non-human animals such as companion animals (such as dogs, cats and the like) , farm animals (such as cows, sheep, pigs, horses and the like) , or laboratory animals (such as rats, mice, guinea pigs and the like) .
  • the subject to be treated by the method of the invention is identified as having obstructive nephropathy.
  • treating refers to the application or administration of a composition including one or more active agents to a subject afflicted with a disorder, a symptom or condition of the disorder, or a progression of the disorder, with the purpose to cure, heal, alleviate, relieve, alter, remedy, ameliorate, improve, or affect the disorder, the symptom or condition of the disorder, the disabilities induced by the disorder, or the progression of the disorder or the symptom or condition thereof.
  • preventing or prevention refers to application or administration of a composition including one or more active agents to a subject who is susceptible or predisposed to a disorder or a symptom or condition thereof and thus relates to prevention of the occurrence of the disorder or the symptom or condition thereof or underlying causes thereof.
  • an effective amount for treating or preventing renal fibrosis refers to the amount of an active ingredient to confer a desired therapeutic effect in a treated subject.
  • an effective amount for treating or preventing renal fibrosis can be an amount that can prohibit, improve, alleviate, reduce or prevent one or more symptoms or conditions or progression thereof such as mononuclear leukocyte inflammation, tubular dilation, tubular atrophy, proliferation of tubular epithelial cells, activation of fibroblast or myofibroblast and deposition of collages (III or IV) , particularly in the tubulointerstitial compartment.
  • the symptoms may be determined and evaluated using methods known in the art based on various disease progress-related indexes, for example by analyzing renal sections via staining.
  • the oral dosage of ginsenoside M1 used in the present invention is 10 to 1,000 mg/kg daily.
  • the oral dosage of ginsenoside M1 used in the present invention is 100 to 300 mg/kg daily, 50 to 150 mg/kg daily, 25 to 100 mg/kg daily, 10 to 50 mg/kg daily, or 5 to 30 mg/kg daily.
  • ginsenoside M1 is administered periodically for a certain period of time, for example, daily administration for at least 5 days, 10 days or 15 days, one month or two months or longer.
  • ginsenoside M1 may be used as an active ingredient for treating or preventing renal fibrosis.
  • an effective amount of the active ingredient may be formulated with a pharmaceutically acceptable carrier into a pharmaceutical composition of an appropriate form for the purpose of delivery and absorption.
  • the pharmaceutical composition of the present invention preferably comprises about 0.1%by weight to about 100%by weight of the active ingredient, wherein the percentage by weight is calculated based on the weight of the whole composition.
  • “pharmaceutically acceptable” means that the carrier is compatible with the active ingredient in the composition, and preferably can stabilize said active ingredient and is safe to the individual receiving the treatment.
  • Said carrier may be a diluent, vehicle, excipient, or matrix to the active ingredient.
  • excipients include lactose, dextrose, sucrose, sorbose, mannose, starch, Arabic gum, calcium phosphate, alginates, tragacanth gum, gelatin, calcium silicate, microcrystalline cellulose, polyvinyl pyrrolidone, cellulose, sterilized water, syrup, and methylcellulose.
  • the composition may additionally comprise lubricants, such as talc, magnesium stearate, and mineral oil; wetting agents; emulsifying and suspending agents; preservatives, such as methyl and propyl hydroxybenzoates; sweeteners; and flavoring agents.
  • lubricants such as talc, magnesium stearate, and mineral oil
  • wetting agents such as talc, magnesium stearate, and mineral oil
  • emulsifying and suspending agents such as methyl and propyl hydroxybenzoates
  • preservatives such as methyl and propyl hydroxybenzoates
  • sweeteners such as methyl and propyl hydroxybenzoates
  • the form of said composition may be tablets, pills, powder, lozenges, packets, troches, elixers, suspensions, lotions, solutions, syrups, soft and hard gelatin capsules, suppositories, sterilized injection fluid, and packaged powder.
  • composition of the present invention may be delivered via any physiologically acceptable route, such as oral, parenteral (such as intramuscular, intravenous, subcutaneous, and intraperitoneal) , transdermal, suppository, and intranasal methods.
  • parenteral administration it is preferably used in the form of a sterile water solution, which may comprise other substances, such as salts or glucose sufficient to make the solution isotonic to blood.
  • the water solution may be appropriately buffered (preferably with a pH value of 3 to 9) as needed.
  • Preparation of an appropriate parenteral composition under sterile conditions may be accomplished with standard pharmacological techniques well known to persons skilled in the art, and no extra creative labor is required.
  • ginsenoside M1 or compositions comprising ginsenoside M1 as the active ingredient may be used in treating or preventing renal fibrosis.
  • ginsenoside M1 or compositions comprising ginsenoside M1 as the active ingredient may be administered to individuals with renal fibrosis or individuals with the risk of acquiring renal fibrosis so as to prevent occurrence of the disease or improve the symptoms or delay deterioration of the symptoms.
  • the ginsenoside M1 can be administered prior to or at the time of occurrence of renal fibrosis, or the ginsenoside M1 can be administered after occurrence of renal fibrosis.
  • ginsenoside M1 or compositions comprising ginsenoside M1 as the active ingredient may be used in combination with existing therapeutic methods or medicaments, such as pharmaceutical treatment, including but not limited to corticosteroids (such as prednisolone) , non-steriodal anti-inflammatory drugs (NSAIDs) , cytotoxic drugs (such as cyclophosphamide, chlorambucil, and azathioprine) , immunosuppressants (such as cyclosporine and Mycophenolate Mofetil) , and vasodilators (such as angiotensin-converting-enzyme inhibitors (ACE inhibitors) ) .
  • corticosteroids such as prednisolone
  • NSAIDs non-steriodal anti-inflammatory drugs
  • cytotoxic drugs such as cyclophosphamide, chlorambucil, and azathioprine
  • immunosuppressants such as cyclosporine and Mycophenolate Mofetil
  • the medicament or therapeutic method used in combination may be used simultaneously (parallel) or sequentially.
  • the medicaments may be mixed in the same formula or put in different formulas separately, such as separate capsules, pills, tablets, and injections.
  • mice Male 8-week-old C57BL/6 mice were purchased form National Laboratory Animal Breeding and Research Center (Taipei, Taiwan) and maintained at the animal center of the National Defense Medical Center (Taipei, Taiwan) . All animal experiments were performed with the approval of the Institutional Animal Care and Use Committee of The National Defense Medical Center, Taiwan, and were consistent with the National Institutes of Health Guide for the Care and Use of Laboratory Animals.
  • mice were first anesthetized with sodium pentobarbital (50 mg/kg, i.p. ) . After a left flank incision was taken, the left ureter was exposed, ligated with 6–0 silk sutures at two points, and cut between the two ligatures. Lastly, the peritoneal membrane and skin were sutured. Sham surgery was performed as control by following all steps of UUO-procedure except ligation and cut of ureter. (Xiao et al. Adenosine A2A receptor: a target for regulating renal interstitial fibrosis in obstructive nephropathy, PLoS One. 2013 Apr 9; 8 (4) : e60173) .
  • mice received preoperative analgesia (subcutaneous injection of 50 mg/kg buprenorphin (Temgesic, Shering-Plough) ) and the right ureter was subsequently ligated with 6.0 silk through a small abdominal incision under 2.0%isoflurane-induced anesthesia. The abdomen was closed in two layers and mice were allowed to recover from surgery for 12 hours at 28°C in a ventilated stove. Mice were sacrificed 7 or 14 days after surgery (Pulskens, et al. Nlrp3 prevents early renal interstitial edema and vascular permeability in unilateral ureteral obstruction, PLoS One. 2014 Jan 15; 9 (1) : e85775) .
  • Ginsenoside M1 20-O- ⁇ -D-glucopyranosyl-20 (S) -protopanaxadiol (named LCHK168 below) , was prepared by methods known in the art, such as those described in Taiwan Patent Application No. 094116005 (I280982) and U.S. Patent No. 7,932,057.
  • LCHK168 was dissolved in 3%cremophore (Sigma-Aldrich) . Starting from one day before UUO operation, mice were given a daily dose of LCHK168 (30 mg/kg) by intraperitoneal injection till sacrifice.
  • Paraffin-embedded kidney tissues were sectioned and stained with Masson’s trichrome and hematoxylin and eosin (H&E) using standard protocols. For histopathology, the tissues were fixed in 10%buffered formalin and embedded in paraffin, and then sections (3 ⁇ m) were prepared and stained with hematoxylin and eosin (H&E) .
  • Hematoxylin was used to counterstain nuclei.
  • terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling (TUNEL) was used.
  • Formalin-fixed tissue sections were stained using an ApopTag Plus Peroxidase In Situ Apoptosis detection kit (Chemicon, Temecula, CA, USA) according to the manufacturer’s instructions.
  • Splenocytes from the mice were treated with Tris-buffered ammonium chloride to eliminate erythrocytes, washed, and resuspended in RPMI1640 medium supplemented with 10%fetal calf serum, Hepes buffer, L-glutamine, and penicillin/streptomycin (all from Invitrogen/Life Technologies, Grand Island, NY, USA) .
  • the cells were then stained for surface markers of T or B cells, using FITC-conjugated anti-mouse CD3 antibodies (17A2) for T cells, anti-mouse CD19 antibodies (1D3) for B cells, and phycoerythrin-conjugated anti-mouse CD69 antibodies (H1.2F3; all antibodies from BD Biosciences) .
  • bone marrow cells analysis bone marrow from mice were isolated as described (Takeshita S et al., 2014) , and were stained with antibodies againt mouse Sca-1 (BD Biosciences) or CD34 (eBioscience, San Diego, CA, USA) . Flow cytometric analysis was carried out using a FACSCalibur (BD Biosciences) .
  • Urine samples are collected from calyx and pelvis by 29G insulin syringe. As normal control, urine is collected by metabolic cages. MCP-1, TNF- ⁇ , and IL-1 ⁇ are then detected by ELISA following instruction manual (eBioscience and R&D) . Briefly, capture antibody is coated in 96well plates over night. After wash with PBST, blocking buffer, samples, detection antibody, avidin-HRP are loaded in order. Colors are developed after TMB substrate step, and then add 2N H 2 SO 4 to stop reaction. Finally, O. D. 450 nm is measure.
  • Renal tissues were collected, fixed and underwent H&E staining.
  • Fig. 2 On 7 days of UUO, disease control mice showed significant mononuclear leukocyte infiltration and tubular dilation in the interstitial area, and this histopathology was inhibited by LCHK168 administration.
  • LCHK168 administration On 14 days of UUO, fibrosis and tubular atrophy are observed, which are significantly inhibited after LCHK168 administration.
  • PCNA proliferating cell nuclear antigen
  • fibrosis is a maladaptive response to renal tissue damage and will develop to irreversible renal dysfunction if obstruction persisted.
  • Histochemical staining of renal section with Masson’s trichrom staining showed a significant increase of fibrosis tissue in the UUO interstitial area compared to sham control, and was decreased after LCHK168 administration.
  • Fig. 4A Further examination were performed by immunohistochemical staining of renal section for ⁇ -SMA, collagen III and IV, which strongly expressed in UUO interstitial area and decreased after LCHK168 administration.
  • Fig. 4B-E These data obtained showed that LCHK168 might ameliorates myofibroblasts activation and renal fibrosis in renal interstitial.
  • NF- ⁇ B is an essential nuclear transcript factor for the modulation of inflammatory related factors for its stimulation of cytokine and cell-adhesion molecules.
  • cytokine and chemokines are release focally in renal and could recruit immune cells. And the recruited immune cells then release more and more inflammatory factors. MCP-1, TNF- ⁇ , and IL-1 ⁇ in renal pelvis are detected. Both UUO and UUO+LCHK group express higher MCP-1, TNF- ⁇ , and IL-1 ⁇ compare to normal control group at day 7. Although the level of these cytokine and chemokines are significant decreased at day 14 on UUO+LCHK group compare to UUO group. The figure shows: LCHK168 could significant reduce urine MCP-1, TNF- ⁇ , and IL-1 ⁇ level.
  • LCHK168 To examine LCHK168 whether prevents local inflammation and fibrosis through inhibition of systemic cellular immunity, we examined B cells and T cells activation in splenocytes by flow cytometry. As shown in Fig. 8A and 8B, there was no significant change in the percentage of CD3 + CD69 + (activated pan B) within three groups, nor was CD19 + CD69 + (activated pan T) . The data obtained indicated nephropathy caused by UUO might have little effect on systemic cellular immunity, and LCHK168 may not prevent local inflammation and fibrosis through inhibition of systemic cellular immunity.
  • LCHK168 To examine LCHK168 whether prevents local inflammation and fibrosis through inhibition of hematopoietic stem cell activation, we examined stem cells and hematopoietic stem cell activation in bone marrow cells by flow cytometry. As shown in Fig. 9A and 9B, there was no significant change in the percentage of Stem-cell antigen-1-positive cells (hematopoietic stem cell) and CD34 + (stem cell) cells within three groups. The data obtained indicated nephropathy caused by UUO might have little effect on hematopoietic stem cell activity, and LCHK168 may not prevent local inflammation and fibrosis through inhibition of hematopoietic stem cell activity.
  • ginsenoside M1 is effective in preventing development of renal fibrosis in the UUO model.
  • the results shows (1) a reduction in cell apoptosis and tubular damage in renal interstitial tissues (2) prevention of fibrosis and collage deposition in renal interstitial tissues, (3) a reduction of NF- ⁇ B activation, (4) prevention of macrophage and T cell infiltration of the kidney, and (5) normal function of hematopoietic stem cell and systemic cellular immunity. All these findings suggest that ginsenoside M can be further developed to a candidate new drug for treatment or prevention of renal fibrosis.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Animal Behavior & Ethology (AREA)
  • Epidemiology (AREA)
  • Organic Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Molecular Biology (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • General Engineering & Computer Science (AREA)
  • Microbiology (AREA)
  • Biotechnology (AREA)
  • Biochemistry (AREA)
  • Genetics & Genomics (AREA)
  • Nutrition Science (AREA)
  • Physiology (AREA)
  • Dermatology (AREA)
  • Urology & Nephrology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

A method of inhibiting renal fibrosis in a subject in need thereof is provided.

Description

USE OF GINSENOSIDE M1 FOR INHIBITING RENAL FIBROSIS FIELD OF THE INVENTION
The present invention relates to a new use of ginsenoside M1 for inhibiting renal fibrosis, particularly interstitial fibrosis.
BACKGROUND OF THE INVENTION
Renal fibrosis is the final stage of renal diseases. The causes of renal fibrosis include ureteral obstruction.
Ureteral obstruction is one of the most common problems confronting the urologist. Ureteral obstruction causes impedance to the flow of urine in the ureter. Most commonly, obstruction occurs at the ureteropelvic junction. The broader term “obstructive uropathy” can be used to indicate any obstruction to urinary flow occurring between the renal pelvis and the urethra, which causes a developing hydronephrosis and associated renal impairment. Urethral strictures and benign prostatic hypertrophy are cases in point. The incidence of unilateral ureteric obstruction (UUO) is reported as 1/1,000 in adults. Ureteric calculi are the most common cause, and acute obstruction typically presents with significant renal colic as the major symptom. See, for example, Docherty et al., evidence that inhibition of tubular cell apoptosis protects against renal damage and development of fibrosis following ureteric obstruction, Am J Physiol Renal Physiol. 2006 Jan; 290 (1) : F4-13.
In the beginning of UUO, the interstitium is infiltrated by monocytes, which are ‘classically’ activated to macrophages that release cytokines such as TGF-β1 and tumor necrosis factor-α (TNF-α) . In turn, TGF-β1 promotes a phenotypic response of tubular epithelial cells either to undergo apoptosis (leading to tubular atrophy) or to undergo epithelial–mesenchymal transition (EMT) , becoming fibroblasts that migrate to the interstitium. Angiotensin II (ANG II) , produced by the activation of monocytes, stimulates the production of nuclear factor-κB (NF-κB) , which leads to the recruitment of more macrophages, as well as to the production of reactive oxygen species (ROS) , which aggravates renal tubular injury. In contrast, alternatively activated macrophages can enhance tubular cell survival and proliferation. Endothelial cells can undergo endothelial–mesenchymal transition (EndMT) or apoptosis, which leads to capillary loss and secondary renal ischemia and hypoxia. Resident pericytes and infiltrating hematopoietic stem cells can also  differentiate into fibroblasts. Under the stimulus of cytokines, such as TGF-β1 produced by macrophages or other cells, fibroblasts synthesize stress fibers and undergo further differentiation to become myofibroblasts. The myofibroblasts are contractile and augment the deposition of the extracellular matrix (ECM) , leading to progressive interstitial fibrosis. This process finally leads to progressive renal fibrosis and irreversible renal failure. See, for example, Chevalier et al., Ureteral obstruction as a model of renal interstitial fibrosis and obstructive nephropathy, Kidney Int. 2009 Jun; 75 (11) : 1145-52.
Ginsenosides, the main active ingredients of ginseng, are known to have a variety of pharmacological activities, e.g. antitumor, antidiabetic, antifatique, antiallergic and antioxidant activities. Ginsenosides share a basic structure, composed of gonane steroid nucleus having 17 carbon atoms arranged in four rings. Ginsenosides are metalized in the body, and a number of recent studies suggest that ginsenoside metabolites, rather than naturally occurring ginsenosides, are readily absorbed in the body and act as the active components. Among them, ginsenoside M1 is known as one metabolite of protopanaxadiol-type ginsenosides via the gypenoside pathway by human gut bacteria. Until now, no prior art references report the effect of ginsenoside M1 in treatment of renal fibrosis.
BRIEF SUMMARY OF THE INVENTION
In the present invention, it is unexpected found that ginsenoside M1 is effective in inhibiting renal fibrosis. Therefore, the present invention provides a new approach for treatment or prevention of renal fibrosis in a subject.
In particular, the present invention provides a method for inhibiting renal fibrosis in a subject in need thereof comprising administering an effective amount of ginsenoside M1 to the subject.
In some embodiments, the subject is a patient with an obstructive nephropathy.
In some embodiments, the method of the invention is effective in reducing or alleviating one or more symptoms or conditions, including but not limited to, mononuclear leukocyte inflammation, tubular dilation, tubular atrophy, proliferation of tubular epithelial cells, activation of fibroblast or myofibroblast and deposition of  collagens (e.g. III or IV) , particularly in the tubulointerstitial compartment of the subject.
In some embodiments, the method of the invention is effective in reducing or alleviating mononuclear leukocyte infiltration or fibrosis in renal interstitial in the subject.
In some embodiments, the ginsenoside M1 is administered by parenteral or enteral route.
In some embodiments, the ginsenoside M1 is administered prior to or at the time of occurrence of renal fibrosis.
In some embodiments, the ginsenoside M1 is administered after occurrence of renal fibrosis.
Also provided is use of ginsenoside M1 for manufacturing a medicament for treating renal fibrosis.
The details of one or more embodiments of the invention are set forth in the description below. Other features or advantages of the present invention will be apparent from the following detailed description of several embodiments, and also from the appending claims.
BRIEF DESCRIPTION OF THE DRAWINGS
For the purpose of illustrating the invention, there are shown in the drawings embodiments. It should be understood, however, that the invention is not limited to the preferred embodiments shown. In the drawings:
Fig. 1 shows the establishment of the UUO animal model, including (A) the experimental procedure, (B) the surgical procedure of the UUO animal model wherein the left is the diseased kidney after ligation of ureter and the right is the normal kidney without ligation of ureter.
Fig. 2 shows the kidney histopathological evaluation by H&E staining wherein the arrows indicate dilated tubule. (A) H&E staining for renal histopathological evaluation. “#” indicates dilated renal tubule, asterisk indicates monocytes, and “*” indicates atrophic renal tubule. The original magnification was x400 for histopathology. (B) Semi-quantitative shows tubulointerstitial injury scores (TIS) . White, black and gray columns represent normal control mice, disease-control mice and LCHK168+UUO mice, respectively. ***p<0.005.
Fig. 3 shows renal cell apoptosis and proliferation evaluation, including (A) detection of PCNA by immunohistochemical staining wherein the arrows indicate positively stained cells, and (B) semi-quantitative results. Original magnification is ×400. White, black and gray columns represent normal control mice, disease-control mice and LCHK168+UUO mice, respectively. ***p<0.005.
Fig. 4 shows fibrosis expression evaluation, including (A) expression of fibrotic collagens in renal tissues by Masson’s trichome staining, (B) expression of smooth muscle α-actin (α-SMA) in renal tissues by immunohistochemical staining, (C) expression of collagen III in renal tissues by immunohistochemical staining, (D) expression of collagen IV in renal tissues by immunohistochemical staining, wherein the arrows indicate positively stained cells, and (E) semi-quantitative results. Original magnification is×400. White, black and gray columns represent normal control mice, disease-control mice and LCHK168+UUO mice, respectively. *p<0.05, **p<0.01, ***p<0.005.
Fig. 5 shows (A) expression of NF-κB in renal tissues by immunohistochemical staining, wherein the arrows indicate positively stained cells, and (B) semi-quantitative results. Original magnification is ×400. White, black and gray columns represent normal control mice, disease-control mice and LCHK168+UUO mice, respectively. ***p<0.005.
Fig. 6 shows mononuclear leukocyte infiltration in renal tissues, including detection of (A) F4/80+ monocytes/macrophages and (B) CD3+ T cells by immunohistochemical staining, wherein the arrows indicate positively stained cells, and (C) semi-quantitative results. Original magnification is ×400. White, black and gray columns represent normal control mice, disease-control mice and LCHK168+UUO mice, respectively. ***p<0.005.
Fig. 7 shows that LCHK168 inhibits the expression level of MCP-1, TNF-α and IL-1β in pelvis urine. (A) MCP-1, (B) TNF-α, and (C) IL-1β are detected in urine samples by ELISA. White, black and gray columns represent normal control mice, disease-control mice and LCHK168+UUO mice, respectively. *p<0.05, **p<0.01, ***p<0.005.
Fig. 8 shows evaluation for activation of cellular immunity, including detection of (A) CD3+ CD69+ T cells and (B) CD19+ CD69+ T cells by flow cytometry. White, black and gray columns represent normal control mice, disease-control mice and LCHK168+UUO mice, respectively.
Fig. 9 shows evaluation of activation of hematopoietic stem cells, including detection of (A) CD34+ stem cells and (B) Sca-1+ hematopoietic stem cells by flow cytometry. White, black and gray columns represent normal control mice, disease-control mice and LCHK168+UUO mice, respectively.
DETAILED DESCRIPTION OF THE INVENTION
Unless defined otherwise, all technical and scientific terms used herein have the meaning commonly understood by a person skilled in the art to which this invention belongs. As used herein, the following terms have the meanings ascribed to them unless specified otherwise.
The articles “a” and “an” are used herein to refer to one or more than one (i.e., at least one) of the grammatical object of the article. By way of example, “an element” means one element or more than one element.
In the present invention, it is unexpectedly found that ginsenoside M1 is effective in inhibiting renal fibrosis. In particular, it is found that in the mice UUO model, the renal damage/inflammation is reduced and the progression to renal fibrosis is inhibited or alleviated.
The surgical-intervented UUO model on rodents has been used for decades as a high-throughput experimental model for UUO. This model is not complicated by hypertension, proteinuria or hyperlipidemia, and does not involve any apparent immune or toxic renal insult. Depending on the duration of obstruction, UUO can mimic various stages of obstructive nephropathy that ultimately lead to tubulointerstitial fibrosis. The animal’s lifespan is not compromised, because the function of the contralateral kidney is maintained or even increased as a result of compensatory functional and anatomic hypertrophy. Other Advantages of this model include the fact that the progression of renal fibrosis is highly predictable and reproducible and leads to significant fibrosis and nephron loss in a relatively short period (7–14 days) . See, for example, Eddy et al., Investigating mechanisms of chronic kidney disease in mouse models, Pediatr Nephrol. 2012 Aug; 27 (8) : 1233-47; and Grande MT and López-Novoa JM, Fibroblast activation and myofibroblast generation in obstructive nephropathy, Nat Rev Nephrol. 2009 Jun; 5 (6) : 319-28.
The examples and data below show the unexpected effects of ginsenoside M1, including (1) a reduction in cell apoptosis and tubular damage in renal interstitial tissues (2) prevention of fibrosis and collage deposition in renal interstitial tissues, (3)  a reduction of NF-κB activation, (4) prevention of macrophage and T cell infiltration of the kidney, and (5) maintenance of normal function of hematopoietic stem cell and systemic cellular immunity.
Accordingly, the present invention provides a new approach for inhibition of renal fibrosis in a subject in need by administering an effective amount of ginsenoside M1 to the subject.
Ginsenoside M1, 20-O-β-D-glucopyranosyl-20 (S) -protopanaxadiol, is one of saponin metabolites known in the art. The chemical structure of ginsenoside M1 is as follows:
Figure PCTCN2015079156-appb-000001
Ginsenoside M1 is known as one metabolite of protopanaxadiol-type ginsenosides via the gypenoside pathway by human gut bacteria. Ginsenoside M1 can be found in blood or urine after intake. Ginsenoside M1 may be prepared from ginseng plants through fungi fermentation by methods known in the art, such as Taiwan Patent Application No. 094116005 (I280982) and U.S. Patent No. 7,932,057, the entire content of which is incorporated herein by reference. In certain embodiments, the ginseng plants for preparing the ginsenoside M1 include Araliaceae family, Panax genus, e.g. P. ginseng and P. pseudo-ginseng (also named Sanqi) . In general, the method of preparation of ginsenoside M1 includes the steps of (a) providing powder of ginseng plant materials (e.g. leaves or stems) ; (b) providing a fungus for fermenting the ginseng plant materials, wherein the fermentation temperature is ranged from 20-50℃., the fermentation humidity is ranged from 70-100%, the pH value is ranged from 4.0-6.0, and the fermentation period is ranged from 5-15 days; (c) extracting and collecting the fermentation products; and (d)  isolating 20-O-β-D-glucopyranosyl-20 (S) -protopanaxadiol from the fermentation products.
When ginsenoside M1 is described as “isolated” or “purified” in the present invention, it should be understood as not absolutely isolated or purified, but relatively isolated or purified. For example, purified ginsenoside M1 refers to one that is more purified compared to its naturally existing form. In one embodiment, a preparation comprising purified ginsenoside M1 may comprise ginsenoside M1 in an amount of more than 50%, more than 60%, more than 70%, more than 80%, more than 90%, or 100% (w/w) of the total preparation. It should be understood that when a certain number was used herein to show a ratio or dosage, said number generally includes that within the range of 10%more and less, or more specifically, the scope of 5%more and less than the number.
The present invention provides a method for inhibiting renal fibrosis comprising administering an effective amount of ginsenoside M1 to a subject in need thereof. Also provided is use of ginsenoside M1 for manufacturing a medicament for inhibiting renal fibrosis in a subject in need thereof. The medicament of the invention is effective in reducing or alleviating one or more symptoms or conditions, including but not limited to, mononuclear leukocyte inflammation, tubular dilation, tubular atrophy, proliferation of tubular epithelial cells, activation of fibroblast or myofibroblast and deposition of collagens (e.g. III or IV) , particularly in the tubulointerstitial compartment of the subject. Specifically, the medicament of the invention is effective in reducing or alleviating mononuclear leukocyte infiltration or fibrosis in renal interstitial in the subject.
The term “individual” or “subject” used herein includes human and non-human animals such as companion animals (such as dogs, cats and the like) , farm animals (such as cows, sheep, pigs, horses and the like) , or laboratory animals (such as rats, mice, guinea pigs and the like) . In certain embodiments, the subject to be treated by the method of the invention is identified as having obstructive nephropathy.
The term “treating” or “treatment” as used herein refers to the application or administration of a composition including one or more active agents to a subject afflicted with a disorder, a symptom or condition of the disorder, or a progression of the disorder, with the purpose to cure, heal, alleviate, relieve, alter, remedy, ameliorate, improve, or affect the disorder, the symptom or condition of the disorder, the disabilities induced by the disorder, or the progression of the disorder or the symptom  or condition thereof. The term “preventing” or “prevention” as used herein refers to application or administration of a composition including one or more active agents to a subject who is susceptible or predisposed to a disorder or a symptom or condition thereof and thus relates to prevention of the occurrence of the disorder or the symptom or condition thereof or underlying causes thereof..
The term “effective amount” used herein refers to the amount of an active ingredient to confer a desired therapeutic effect in a treated subject. For example, an effective amount for treating or preventing renal fibrosis can be an amount that can prohibit, improve, alleviate, reduce or prevent one or more symptoms or conditions or progression thereof such as mononuclear leukocyte inflammation, tubular dilation, tubular atrophy, proliferation of tubular epithelial cells, activation of fibroblast or myofibroblast and deposition of collages (III or IV) , particularly in the tubulointerstitial compartment. The symptoms may be determined and evaluated using methods known in the art based on various disease progress-related indexes, for example by analyzing renal sections via staining. The effective amount may change depending on various reasons, such as administration route and frequency, body weight and species of the individual receiving said pharmaceutical, and purpose of administration. Persons skilled in the art may determine the dosage in each case based on the disclosure herein, established methods, and their own experience. For example, in certain embodiments, the oral dosage of ginsenoside M1 used in the present invention is 10 to 1,000 mg/kg daily. In some examples, the oral dosage of ginsenoside M1 used in the present invention is 100 to 300 mg/kg daily, 50 to 150 mg/kg daily, 25 to 100 mg/kg daily, 10 to 50 mg/kg daily, or 5 to 30 mg/kg daily. In addition, in some embodiments of the invention, ginsenoside M1 is administered periodically for a certain period of time, for example, daily administration for at least 5 days, 10 days or 15 days, one month or two months or longer.
According to the present invention, ginsenoside M1 may be used as an active ingredient for treating or preventing renal fibrosis. In one embodiment, an effective amount of the active ingredient may be formulated with a pharmaceutically acceptable carrier into a pharmaceutical composition of an appropriate form for the purpose of delivery and absorption. Depending on the mode of administration, the pharmaceutical composition of the present invention preferably comprises about 0.1%by weight to about 100%by weight of the active ingredient, wherein the percentage by weight is calculated based on the weight of the whole composition.
As used herein, “pharmaceutically acceptable” means that the carrier is compatible with the active ingredient in the composition, and preferably can stabilize said active ingredient and is safe to the individual receiving the treatment. Said carrier may be a diluent, vehicle, excipient, or matrix to the active ingredient. Some examples of appropriate excipients include lactose, dextrose, sucrose, sorbose, mannose, starch, Arabic gum, calcium phosphate, alginates, tragacanth gum, gelatin, calcium silicate, microcrystalline cellulose, polyvinyl pyrrolidone, cellulose, sterilized water, syrup, and methylcellulose. The composition may additionally comprise lubricants, such as talc, magnesium stearate, and mineral oil; wetting agents; emulsifying and suspending agents; preservatives, such as methyl and propyl hydroxybenzoates; sweeteners; and flavoring agents. The composition of the present invention can provide the effect of rapid, continued, or delayed release of the active ingredient after administration to the patient.
According to the present invention, the form of said composition may be tablets, pills, powder, lozenges, packets, troches, elixers, suspensions, lotions, solutions, syrups, soft and hard gelatin capsules, suppositories, sterilized injection fluid, and packaged powder.
The composition of the present invention may be delivered via any physiologically acceptable route, such as oral, parenteral (such as intramuscular, intravenous, subcutaneous, and intraperitoneal) , transdermal, suppository, and intranasal methods. Regarding parenteral administration, it is preferably used in the form of a sterile water solution, which may comprise other substances, such as salts or glucose sufficient to make the solution isotonic to blood. The water solution may be appropriately buffered (preferably with a pH value of 3 to 9) as needed. Preparation of an appropriate parenteral composition under sterile conditions may be accomplished with standard pharmacological techniques well known to persons skilled in the art, and no extra creative labor is required.
According to the present invention, ginsenoside M1 or compositions comprising ginsenoside M1 as the active ingredient may be used in treating or preventing renal fibrosis. Specifically, ginsenoside M1 or compositions comprising ginsenoside M1 as the active ingredient may be administered to individuals with renal fibrosis or individuals with the risk of acquiring renal fibrosis so as to prevent occurrence of the disease or improve the symptoms or delay deterioration of the symptoms. Specifically, the ginsenoside M1 can be administered prior to or at the  time of occurrence of renal fibrosis, or the ginsenoside M1 can be administered after occurrence of renal fibrosis.
In addition, according to the present invention, ginsenoside M1 or compositions comprising ginsenoside M1 as the active ingredient may be used in combination with existing therapeutic methods or medicaments, such as pharmaceutical treatment, including but not limited to corticosteroids (such as prednisolone) , non-steriodal anti-inflammatory drugs (NSAIDs) , cytotoxic drugs (such as cyclophosphamide, chlorambucil, and azathioprine) , immunosuppressants (such as cyclosporine and Mycophenolate Mofetil) , and vasodilators (such as angiotensin-converting-enzyme inhibitors (ACE inhibitors) ) . In one embodiment, the medicament or therapeutic method used in combination may be used simultaneously (parallel) or sequentially. When medicaments are used in combination, the medicaments may be mixed in the same formula or put in different formulas separately, such as separate capsules, pills, tablets, and injections.
The present invention is further illustrated by the following examples, which are provided for the purpose of demonstration rather than limitation.
Example
1. Materials and Methods
1.1 Animal model and experimental protocol
UUO (Unilateral ureteral obstruction) animal model
Male 8-week-old C57BL/6 mice were purchased form National Laboratory Animal Breeding and Research Center (Taipei, Taiwan) and maintained at the animal center of the National Defense Medical Center (Taipei, Taiwan) . All animal experiments were performed with the approval of the Institutional Animal Care and Use Committee of The National Defense Medical Center, Taiwan, and were consistent with the National Institutes of Health Guide for the Care and Use of Laboratory Animals.
Mice were first anesthetized with sodium pentobarbital (50 mg/kg, i.p. ) . After a left flank incision was taken, the left ureter was exposed, ligated with 6–0 silk sutures at two points, and cut between the two ligatures. Lastly, the peritoneal membrane and skin were sutured. Sham surgery was performed as control by following all steps of UUO-procedure except ligation and cut of ureter. (Xiao et al. Adenosine A2A receptor: a target for regulating renal interstitial fibrosis in obstructive nephropathy, PLoS One. 2013 Apr 9; 8 (4) : e60173) .
Briefly, all mice received preoperative analgesia (subcutaneous injection of 50 mg/kg buprenorphin (Temgesic, Shering-Plough) ) and the right ureter was subsequently ligated with 6.0 silk through a small abdominal incision under 2.0%isoflurane-induced anesthesia. The abdomen was closed in two layers and mice were allowed to recover from surgery for 12 hours at 28℃ in a ventilated stove. Mice were sacrificed 7 or 14 days after surgery (Pulskens, et al. Nlrp3 prevents early renal interstitial edema and vascular permeability in unilateral ureteral obstruction, PLoS One. 2014 Jan 15; 9 (1) : e85775) .
1.2 Ginsenoside M1 and administration
Ginsenoside M1, 20-O-β-D-glucopyranosyl-20 (S) -protopanaxadiol (named LCHK168 below) , was prepared by methods known in the art, such as those described in Taiwan Patent Application No. 094116005 (I280982) and U.S. Patent No. 7,932,057. LCHK168 was dissolved in 3%cremophore (Sigma-Aldrich) . Starting from one day before UUO operation, mice were given a daily dose of LCHK168 (30 mg/kg) by intraperitoneal injection till sacrifice.
1.3 Renal Histopathology
Paraffin-embedded kidney tissues were sectioned and stained with Masson’s trichrome and hematoxylin and eosin (H&E) using standard protocols. For histopathology, the tissues were fixed in 10%buffered formalin and embedded in paraffin, and then sections (3 μm) were prepared and stained with hematoxylin and eosin (H&E) .
Sections all are examed after mounting. The methods of semi-quantitative analysis are according to the degree of inflammation and fibrosis on renal tubulointerstitial whereas and randomly selected 20 fields randomly for scoring. “0” indicates normal morphology, “1” indicates influent area is less than 10%, “2” indicates influent area is around 10 to 30%, “3” indicates influent area is around 30 to 50%, and “4” indicates influent area is more than 50%.
1.4 Immunohistochemistry (IHC) and detection of apoptosis
For IHC, formalin-fixed and paraffin-embedded tissue sections were incubated overnight at 4℃ with anti-bodies against PCNA (Santa Cruz Biotechnology, Santa Cruz, CA, USA) , CD3 (DAKO, Glostrup, Denmark) , α-SMA (Thermo Fisher Scietific, Waltham, Massachusetts, USA) , Collagen Ⅲ, Collagen IV (both from SouthernBiotech, Birmingham, AL, USA) , F4/80 (monocytes/macrophages; Serotec,  Raleigh, NC, USA) , phospho-NF-κB p65 (pNF-κB p65; Cell Signaling Technology, Beverly, MA, USA) , diluted in DAKO antibody dilution buffer (DAKO, Glostrup, Denmark) , and then for 1h at room temperature with horseradish peroxidase (HRP) -conjugated second antibodies (DAKO) or anti-rabbit polymer-HRP (BioGenex, Fremont , CA, USA) in the same buffer; then DAB (BioGenex) was added. Hematoxylin was used to counterstain nuclei. For detection of apoptosis, terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling (TUNEL) was used. Formalin-fixed tissue sections were stained using an ApopTag Plus Peroxidase In Situ Apoptosis detection kit (Chemicon, Temecula, CA, USA) according to the manufacturer’s instructions. The abundance of α-SMA, Collagen Ⅲ, Collagen IV or the number of phosphorylated NF-κB p65+, PCNA+, F4/80+, CD3+ and apoptotic cells was counted at a magnification of x400 randomly selected fields of the tubulointerstitial compartment in the cortical area by Pax-It quantitative image analysis software (Paxcam) .
1.5 Flowcytometry
Splenocytes from the mice were treated with Tris-buffered ammonium chloride to eliminate erythrocytes, washed, and resuspended in RPMI1640 medium supplemented with 10%fetal calf serum, Hepes buffer, L-glutamine, and penicillin/streptomycin (all from Invitrogen/Life Technologies, Grand Island, NY, USA) . The cells were then stained for surface markers of T or B cells, using FITC-conjugated anti-mouse CD3 antibodies (17A2) for T cells, anti-mouse CD19 antibodies (1D3) for B cells, and phycoerythrin-conjugated anti-mouse CD69 antibodies (H1.2F3; all antibodies from BD Biosciences) . For bone marrow cells analysis, bone marrow from mice were isolated as described (Takeshita S et al., 2014) , and were stained with antibodies againt mouse Sca-1 (BD Biosciences) or CD34 (eBioscience, San Diego, CA, USA) . Flow cytometric analysis was carried out using a FACSCalibur (BD Biosciences) .
1.6 Masson’s trichrome staining
Three μm renal sections are incubated in 75℃ for 15minutes. Transfer the slides above into xyline and gradient alcohol. Mordant is mounted onto slides following stain with hematoxylin for 1 minute. After staining with 0.75%orange G solution, masson stain solution B, 2.5%phosphotungostic acid solution, and aniline blue stain solution, slides are washed by 1%acetic acid twice and mount with gum  Arabic. Air dry for few minutes and the stained sections are exam onto optical microscope.
1.7 Enzyme-linked immunosorbent assay
Urine samples are collected from calyx and pelvis by 29G insulin syringe. As normal control, urine is collected by metabolic cages. MCP-1, TNF-α, and IL-1βare then detected by ELISA following instruction manual (eBioscience and R&D) . Briefly, capture antibody is coated in 96well plates over night. After wash with PBST, blocking buffer, samples, detection antibody, avidin-HRP are loaded in order. Colors are developed after TMB substrate step, and then add 2N H2SO4 to stop reaction. Finally, O. D. 450 nm is measure.
1.8 Statistical analysis
Values are means ±SD. Comparison between two groups was performed using Student’s t-test. A P value <0.05 was considered statistically significant.
2. Results
2.1 Establishment of experimental UUO model
Experiments were performed on 8-10 weeks old C57BL/6 mice. Mice were randomly divided into sham control, disease control and LCHK168 therapy group and were sacrificed after UUO for 7 or 14 days. Retention of urine in the pelvis of UUO kidney was observed, indicated UUO model established successfully. See Fig. 1A and Fig. 1B.
2.2 Renal histopathology evaluation
Renal tissues were collected, fixed and underwent H&E staining. As shown in Fig. 2, on 7 days of UUO, disease control mice showed significant mononuclear leukocyte infiltration and tubular dilation in the interstitial area, and this histopathology was inhibited by LCHK168 administration. On 14 days of UUO, fibrosis and tubular atrophy are observed, which are significantly inhibited after LCHK168 administration.
2.3 Renal cell apoptosis and proliferation evaluation
In the UUO kidney, mechanic stress caused by the retention of urine, and cytokine and ROS release by renal cells as a response to the stress will cause damage to cells, which lead to cell self-renew or apoptosis reaction. TUNNEL staining of renal section for apoptotic cells showed the UUO renal interstitial area, and was decrease after LCHK168 administration. See Fig. 3A.
Immunohistochemical staining of renal section with proliferating cell nuclear antigen (PCNA) , a marker not only for the proliferative status of tubular epithelial cells but also activated myofibroblast and inflammatory cells, showed significant increased after UUO, particularly in dilated tubule. However, PCNA expression was decrease after administration of LCHK168. These results indicated LCHK168 might attenuated cell apoptosis and damage. See Fig. 3B.
2.4 Fibrosis expression evaluation
In UUO kidney, fibrosis is a maladaptive response to renal tissue damage and will develop to irreversible renal dysfunction if obstruction persisted. Histochemical staining of renal section with Masson’s trichrom staining showed a significant increase of fibrosis tissue in the UUO interstitial area compared to sham control, and was decreased after LCHK168 administration. See Fig. 4A. Further examination were performed by immunohistochemical staining of renal section for α-SMA, collagen Ⅲ and Ⅳ, which strongly expressed in UUO interstitial area and decreased after LCHK168 administration. See Fig. 4B-E. These data obtained showed that LCHK168 might ameliorates myofibroblasts activation and renal fibrosis in renal interstitial.
2.5 NF-κB modulated inflammatory response
2.5.1 Expression of NF-κB in renal tissue
NF-κB is an essential nuclear transcript factor for the modulation of inflammatory related factors for its stimulation of cytokine and cell-adhesion molecules. Previous studies indicated the expression level of NF-κB elevates in various inflammatory disorders, including kidney diseases. Immunohistochemical staining of renal section for NF-κB showed an increase of NF-κB expression, and was inhibited after LCHK168 administration. See Fig. 5. This data showed LCHK168 may inhibit NF-κB in UUO kidney.
2.5.2 Mononuclear leukocyte infiltration
Previous studies indicated that inflammatory cells play an important role in the pathogenesis of UUO. Infiltration of mononuclear leukocyte, especially macrophage and T lymphocyte, can be observed in the early stage of UUO. With the persistent of obstruction, the activated inflammatory cells stimulate tubular epithelial cells or other inflammatory cells to release various cytokines and chemokines, cause more inflammatory cells recruiting to the renal interstitial area and induced renal  inflammation and fibrosis. Evaluation of mononuclear leukocyte infiltration was carried out by immunohistochemical staining of renal section for F4/80 and CD3+, which are the markers for macrophage and T cells. The expression of F4/80 and CD3+increased on after UUO and were decreased after LCHK168 administration. See Fig. 6A, 6B. The data obtained showed LCHK168 might prevents macrophage and T cells infiltration in renal interstitial.
2.5.3 Evaluation of expression of MCP-1, TNF-α and IL-1β in urine
At initial stage, cytokine and chemokines are release focally in renal and could recruit immune cells. And the recruited immune cells then release more and more inflammatory factors. MCP-1, TNF-α, and IL-1β in renal pelvis are detected. Both UUO and UUO+LCHK group express higher MCP-1, TNF-α, and IL-1βcompare to normal control group at day 7. Although the level of these cytokine and chemokines are significant decreased at day 14 on UUO+LCHK group compare to UUO group. The figure shows: LCHK168 could significant reduce urine MCP-1, TNF-α, and IL-1β level.
2.6 Activation of cellular immunity and hematopoietic stem cell activation
2.6.1 Activation of B and T lymphocyte
To examine LCHK168 whether prevents local inflammation and fibrosis through inhibition of systemic cellular immunity, we examined B cells and T cells activation in splenocytes by flow cytometry. As shown in Fig. 8A and 8B, there was no significant change in the percentage of CD3+CD69+ (activated pan B) within three groups, nor was CD19+CD69+ (activated pan T) . The data obtained indicated nephropathy caused by UUO might have little effect on systemic cellular immunity, and LCHK168 may not prevent local inflammation and fibrosis through inhibition of systemic cellular immunity.
2.6.2 Activation of hematopoietic stem cell
To examine LCHK168 whether prevents local inflammation and fibrosis through inhibition of hematopoietic stem cell activation, we examined stem cells and hematopoietic stem cell activation in bone marrow cells by flow cytometry. As shown in Fig. 9A and 9B, there was no significant change in the percentage of Stem-cell antigen-1-positive cells (hematopoietic stem cell) and CD34+ (stem cell) cells within three groups. The data obtained indicated nephropathy caused by UUO might have  little effect on hematopoietic stem cell activity, and LCHK168 may not prevent local inflammation and fibrosis through inhibition of hematopoietic stem cell activity.
In summary, our study shows that ginsenoside M1 is effective in preventing development of renal fibrosis in the UUO model. In particular, the results shows (1) a reduction in cell apoptosis and tubular damage in renal interstitial tissues (2) prevention of fibrosis and collage deposition in renal interstitial tissues, (3) a reduction of NF-κB activation, (4) prevention of macrophage and T cell infiltration of the kidney, and (5) normal function of hematopoietic stem cell and systemic cellular immunity. All these findings suggest that ginsenoside M can be further developed to a candidate new drug for treatment or prevention of renal fibrosis.
It is believed that a person of ordinary knowledge in the art where the present invention belongs can utilize the present invention to its broadest scope based on the descriptions herein with no need of further illustration. Therefore, the descriptions and claims as provided should be understood as of demonstrative purpose instead of limitative in any way to the scope of the present invention.

Claims (9)

  1. A method of inhibiting renal fibrosis in a subject in need thereof comprising administering to an effective amount of ginsenoside M1 to the subject.
  2. The method of claim 1, wherein the subject is a patient with an obstructive nephropathy.
  3. The method of claim 1, wherein the ginsenoside M1 is administrated in an amount effective in reducing or alleviating one or more symptoms or conditions, selected from the group consisting of mononuclear leukocyte inflammation, tubular dilation, tubular atrophy, proliferation of tubular epithelial cells, activation of fibroblast or myofibroblast and deposition of collagens, in the subject.
  4. The method of claim 1, wherein the ginsenoside M1 is administrated in an amount effective in reducing or alleviating mononuclear leukocyte infiltration or fibrosis in renal interstitial in the subject.
  5. The method of claim 1, comprising diagnosing or identifying the subject with an obstructive nephropathy prior to administration of ginsenoside M1.
  6. Use of ginsenoside M1 for manufacturing a medicament for inhibiting renal fibrosis in a subject in need thereof.
  7. Use of claim 6, wherein the subject is a patient with an obstructive nephropathy.
  8. Use of claim 6, wherein the medicament is effective in reducing or alleviating one or more symptoms or conditions, selected from the group consisting of mononuclear leukocyte inflammation, tubular dilation, tubular atrophy, proliferation of tubular epithelial cells, activation of fibroblast or myofibroblast and deposition of collagens, in the subject.
  9. Use of claim 6, wherein the medicament is effective in reducing or alleviating mononuclear leukocyte infiltration or fibrosis in renal interstitial in the subject.
PCT/CN2015/079156 2014-05-16 2015-05-18 Use of ginsenoside m1 for inhibiting renal fibrosis WO2015172746A1 (en)

Priority Applications (7)

Application Number Priority Date Filing Date Title
KR1020167035138A KR102360315B1 (en) 2014-05-16 2015-05-18 Use of ginsenoside m1 for inhibiting renal fibrosis
CN201580019639.7A CN106795546A (en) 2014-05-16 2015-05-18 Ginsenoside M1 is used to suppress the purposes of kidney fibrosis
DK15793585.9T DK3146062T3 (en) 2014-05-16 2015-05-18 USE OF GINSENOSIDE M1 FOR INHIBITION OF renal fibrosis
US15/311,683 US10357507B2 (en) 2014-05-16 2015-05-18 Use of ginsenoside M1 for inhibiting renal fibrosis
ES15793585T ES2739203T3 (en) 2014-05-16 2015-05-18 Use of ginsenoside M1 for the inhibition of renal fibrosis
JP2017512092A JP6626094B2 (en) 2014-05-16 2015-05-18 Use of ginsenoside M1 for inhibiting renal fibrosis
EP15793585.9A EP3146062B1 (en) 2014-05-16 2015-05-18 Use of ginsenoside m1 for inhibiting renal fibrosis

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201461994486P 2014-05-16 2014-05-16
US61/994,486 2014-05-16

Publications (1)

Publication Number Publication Date
WO2015172746A1 true WO2015172746A1 (en) 2015-11-19

Family

ID=54479353

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2015/079156 WO2015172746A1 (en) 2014-05-16 2015-05-18 Use of ginsenoside m1 for inhibiting renal fibrosis

Country Status (9)

Country Link
US (1) US10357507B2 (en)
EP (1) EP3146062B1 (en)
JP (1) JP6626094B2 (en)
KR (1) KR102360315B1 (en)
CN (1) CN106795546A (en)
DK (1) DK3146062T3 (en)
ES (1) ES2739203T3 (en)
TW (1) TWI670063B (en)
WO (1) WO2015172746A1 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3137090A4 (en) * 2014-11-03 2017-09-06 Lee, Sheau-Long Use of ginsenoside m1 for treating iga nephropathy
WO2021164723A1 (en) * 2020-02-20 2021-08-26 Lee Sheau Long Ginsenoside m1 as a modulator of angiotensin regulating enzymes and its use for treating diseases or conditions including symptoms caused by coronavirus

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR100381441B1 (en) * 2000-06-14 2003-04-26 기장 메디칼 주식회사 The percutaneous form using moxa and moxa mixed with medical plant, making method and using method thereof
JP7406496B2 (en) * 2018-04-23 2023-12-27 リー・シュー-ロン Use of ginsenoside M1 for the treatment of Huntington's disease

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
TW200930388A (en) * 2008-01-10 2009-07-16 Shih-Ming Chen Pharmaceutical composition for treating cisplatin induced nephropathy
US7932057B2 (en) * 2007-07-09 2011-04-26 Sheau-Long Lee Method for preparing 20-O-β-D-glucopyranosyl-20(S)-protopanaxadiol (ginsenoside M1) by using sanqi leaves and stems
CN102942610A (en) * 2012-11-26 2013-02-27 吉林农业大学 Preparation of ginsenoside M1 n-butyrate and application of antidiabetic medicines of ginsenoside M1 n-butyrate

Family Cites Families (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050102326A1 (en) * 2003-10-22 2005-05-12 Nitzan Peleg Method and apparatus for performing conflict resolution in database logging
CN100402038C (en) * 2004-03-09 2008-07-16 山东绿叶天然药物研究开发有限公司 Use of ginseng saponin C-K in preparing medicine for treating or preventing liver fibrosis
WO2005102326A2 (en) * 2004-04-23 2005-11-03 Ab Science Use of c-kit inhibitors for treating renal diseases
TR201901929T4 (en) * 2005-02-08 2019-03-21 Genzyme Corp Antibodies to TGFBeta.
KR20060114774A (en) * 2005-05-02 2006-11-08 에스케이케미칼주식회사 Cosmetic composition containing (20s)-ginsenoside rg3 for preventing and improving skin wrinkles
US8080568B1 (en) * 2010-06-29 2011-12-20 Ewha University - Industry Collaboration Foundation 2-pyridyl substituted imidazoles as therapeutic ALK5 and/or ALK4 inhibitors
DK3137089T3 (en) * 2014-05-02 2019-01-28 Lee Sheau Long USE OF GINSENOSIDE M1 FOR TREATMENT OF LUPUS NEPHRITIS
WO2016070795A1 (en) * 2014-11-03 2016-05-12 Sheau-Long Lee Use of ginsenoside m1 for treating iga nephropathy

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7932057B2 (en) * 2007-07-09 2011-04-26 Sheau-Long Lee Method for preparing 20-O-β-D-glucopyranosyl-20(S)-protopanaxadiol (ginsenoside M1) by using sanqi leaves and stems
TW200930388A (en) * 2008-01-10 2009-07-16 Shih-Ming Chen Pharmaceutical composition for treating cisplatin induced nephropathy
CN102942610A (en) * 2012-11-26 2013-02-27 吉林农业大学 Preparation of ginsenoside M1 n-butyrate and application of antidiabetic medicines of ginsenoside M1 n-butyrate

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
WANG DONG ET AL.: "The effect of utility monomer in ginseng to the growth and transforming growth factor-beta expression of renal fibroblast cell", JOURNAL OF ZHEJIANG CHINESE MEDICAL UNIVERSITY, vol. 36, no. 8, 31 August 2012 (2012-08-31), pages 917 - 920, XP008185680 *

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3137090A4 (en) * 2014-11-03 2017-09-06 Lee, Sheau-Long Use of ginsenoside m1 for treating iga nephropathy
WO2021164723A1 (en) * 2020-02-20 2021-08-26 Lee Sheau Long Ginsenoside m1 as a modulator of angiotensin regulating enzymes and its use for treating diseases or conditions including symptoms caused by coronavirus

Also Published As

Publication number Publication date
DK3146062T3 (en) 2019-08-12
JP6626094B2 (en) 2019-12-25
CN106795546A (en) 2017-05-31
KR20170007426A (en) 2017-01-18
TW201609111A (en) 2016-03-16
US20170087170A1 (en) 2017-03-30
ES2739203T3 (en) 2020-01-29
EP3146062A4 (en) 2017-11-29
TWI670063B (en) 2019-09-01
EP3146062A1 (en) 2017-03-29
EP3146062B1 (en) 2019-05-08
KR102360315B1 (en) 2022-02-08
US10357507B2 (en) 2019-07-23
JP2017515904A (en) 2017-06-15

Similar Documents

Publication Publication Date Title
Gan et al. Celastrol attenuates bone erosion in collagen-Induced arthritis mice and inhibits osteoclast differentiation and function in RANKL-induced RAW264. 7
Wu et al. Antifibrotic effects of Fraxetin on carbon tetrachloride-induced liver fibrosis by targeting NF-κB/IκBα, MAPKs and Bcl-2/Bax pathways
US10357507B2 (en) Use of ginsenoside M1 for inhibiting renal fibrosis
Shen et al. Anti-proliferation and anti-inflammation effects of corilagin in rheumatoid arthritis by downregulating NF-κB and MAPK signaling pathways
Zhu et al. Bixin protects mice against bronchial asthma though modulating PI3K/Akt pathway
US10172876B2 (en) Use of ginsenoside M1 for treating IgA nephropathy
KR102402139B1 (en) Use of ginsenoside m1 for treating lupus nephritis
Tan et al. BAY61‐3606 protects kidney from acute ischemia/reperfusion injury through inhibiting spleen tyrosine kinase and suppressing inflammatory macrophage response
Wang et al. Metformin ameliorates chronic colitis-related intestinal fibrosis via inhibiting TGF-β1/Smad3 signaling
Liu et al. Protection effect of curcumin for macrophage-involved polyethylene wear particle-induced inflammatory osteolysis by increasing the cholesterol efflux
Wu et al. Renoprotective effects of combination of angiotensin converting enzyme inhibitor with mycophenolate mofetil in diabetic rats
Ma et al. Biliary antibiotics irrigation for E. coli-induced chronic proliferative cholangitis and hepatolithiasis: A pathophysiological study in rabbits
WO2019201280A1 (en) Use of ginsenoside m1 for manufacturing medicament for treating oral cancer
Cheung et al. Therapeutic efficacy of Traditional Chinese Medicine 319 recipe on hepatic fibrosis induced by carbon tetrachloride in rats
Su et al. Anti-inflammatory effects of Abelmoschus manihot (L.) Medik. on LPS-induced cystitis in mice: Potential candidate for cystitis treatment based on classic use
CN109528738A (en) Glycyrrhizic acid promotes the application of Remyelination inhibition neuroinflamation drug in preparation
CN114191450B (en) Application of Prussian blue and analogues thereof in preparation of medicines for preventing, delaying or treating osteoporosis
Lin et al. Inhibition of soluble epoxide hydrolase relieves adipose inflammation via modulating M1/M2 macrophage polarization to alleviate airway inflammation and hyperresponsiveness in obese asthma
Long et al. Combined Epimedii Folium and Ligustri Lucidi Fructus with dexamethasone alleviate the proliferation of airway smooth muscle cells by regulating apoptosis/autophagy
Wei et al. Qiguiyishen decoction reduced the accumulation of extracellular matrix in the kidneys of rats with adriamycin-induced nephropathy
Chen et al. Attenuating Renal Interstitial Fibrosis by Shenqi Pill via Reducing Inflammation Response Regulated by NF-ΰ B Pathway In Vitro and In Vivo.
Zhao et al. The Protective Effects of Herbacetin Against 2, 4, 6-Trinitrobenzene Sulfonic Acid-Induced Inflammatory Bowel Disease via Inhibition of the NF-κB Signaling Pathway
Barrera-Chimal Tan Rui-Zhi1†, Diao Hui1, 2†, Li Jian-Chun1, Zhong Xia1, Wang Xiao-Jia1, Wen Dan1, 2, Fan Jun-Ming1, 3 and Wang Li1
KR20210149516A (en) Pharmaceutical composition for prevention or treatment of bone disease containing Pizotifen or pharmaceutically acceptable salts thereof as an active ingredient
Girbovan The effect of repeated resveratrol administration on global ischemia-induced hippocampal neurodegeneration, neurochemical effects and functional alterations

Legal Events

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

Ref document number: 15793585

Country of ref document: EP

Kind code of ref document: A1

REEP Request for entry into the european phase

Ref document number: 2015793585

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2015793585

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 2017512092

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 15311683

Country of ref document: US

ENP Entry into the national phase

Ref document number: 20167035138

Country of ref document: KR

Kind code of ref document: A