WO2008140141A1 - Pharmaceutical composition for preventing and treating liver fibrosis or hepatic cirrhosis comprising mesenchymal stem cell - Google Patents

Pharmaceutical composition for preventing and treating liver fibrosis or hepatic cirrhosis comprising mesenchymal stem cell Download PDF

Info

Publication number
WO2008140141A1
WO2008140141A1 PCT/KR2007/002397 KR2007002397W WO2008140141A1 WO 2008140141 A1 WO2008140141 A1 WO 2008140141A1 KR 2007002397 W KR2007002397 W KR 2007002397W WO 2008140141 A1 WO2008140141 A1 WO 2008140141A1
Authority
WO
WIPO (PCT)
Prior art keywords
liver
group
liver fibrosis
mscs
dmn
Prior art date
Application number
PCT/KR2007/002397
Other languages
French (fr)
Inventor
Kyung Sun Kang
Jung Chan Ra
Il Seob Shin
Ju Yeon Seo
Original Assignee
Rnl Bio Co., Ltd.
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 Rnl Bio Co., Ltd. filed Critical Rnl Bio Co., Ltd.
Priority to PCT/KR2007/002397 priority Critical patent/WO2008140141A1/en
Publication of WO2008140141A1 publication Critical patent/WO2008140141A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/28Bone marrow; Haematopoietic stem cells; Mesenchymal stem cells of any origin, e.g. adipose-derived stem cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/48Reproductive organs
    • A61K35/51Umbilical cord; Umbilical cord blood; Umbilical stem cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics

Definitions

  • the present invention relates to a pharmaceutical composition for preventing and treating liver fibrosis or hepatic cirrhosis comprising a mesenchymal stem cell.
  • hepatic disease is one of the major causes of death in many countries (Corrao G et al., Int J Epidemiol. 1997 Feb;26(l):100-109).
  • hepatic cirrhosis is a primary cause of death from liver diseases (Schlichting P et al., Scand J Gastroenterol. 1983 Oct;18(7): 881-888).
  • liver fibrosis is central and essential (Hu QW et al., Life Sci. 2006 M 4;79(6): 606-612).
  • Liver fibrosis is a part of a wound healing process related to various types of liver damage, which is characterized by continuous deposition of collagen fibrils into an extra-cellular matrix (ECM) (Du WD et al., World J Gastroenterol. 1999 Oct; 5(5):397-403).
  • ECM extra-cellular matrix
  • liver fibrosis is reversible, which is different from hepatic cirrhosis, and has thin fibrils and no tuberculation.
  • the liver can recover from liver fibrosis.
  • liver fibrosis may lead to irreversible hepatic cirrhosis in which crosslinking between connective tissues increases to accumulate thick fibrils, and a liver lobule loses its normal structure to cause tuberculation.
  • a chronic liver disease may have various causes, it can eventually lead to liver fibrosis or hepatic cirrhosis irrespective of the cause.
  • a liver disease may be asymptomatic in an initial state and thus be difficult to diagnose early.
  • the liver disease is discovered in a chronic state, it may be difficult to treat and have a high mortality rate. Also, effective medication has not yet been developed.
  • mesenchymal stem cells are derived from adult tissues including bone marrow (BM) (Cohnheim J. Ueber Entz W ndung und Eittation, 1867; Ftedenstein AJ et al., J Embryol. Exp. Morphol, Dec 16 1996, 3:381-390; Pittenger MF et al., Science 1999 Apr 2, 284(5411):143-147), adipose Tissue (AD) (ZuJc PA et al., Tissue Eng. 2001 Apr; 7(2):211-228) and umbilical cord blood (UCB) (E ⁇ ces A et al., Br J Haematol.
  • BM bone marrow
  • AD adipose Tissue
  • UB umbilical cord blood
  • Human BM-derived multipotent adult progenitor cells can be differentiated into hematocyte- like cells acquiring functional characteristics of hematocyte in vitro ⁇ .Schwartz RE et al., J Clin. Invest. 2002 May;109(10):1291-1302).
  • the present invention is directed to a pharmaceutical composition for preventing and treating liver fibrosis including a mesenchymal stem cell.
  • the present invention is further directed to a pharmaceutical composition for preventing and treating hepatic cirrhosis including a mesenchymal stem cell.
  • a pharmaceutical composition for preventing and treating liver fibrosis including a mesenchymal stem cell is provided.
  • a pharmaceutical composition for preventing and treating hepatic cirrhosis including a mesenchymal stem cell is provided.
  • liver fibrosis is a disease in which a thin fibril is abnormally proliferated in the liver, and may be caused by a variety of chronic toxic damage, including: chronic alcohol abuse; chronic exposure to drugs including acetaminophen, amiodarone, aspirin, azathioprine, isoniazid, methyldopa, methotrexate, nitrofurantoin, propylthiouracil and sulfonamide; chronic exposure to chemical agents including CCU, dimethyl nitrosamine, vinyl chloride, polychlorinated biphenyl, aflatoxin and insecticides; autoimmune diseases including viral infection, diabetes, primary sclerosing cholangitis, primary bile duct liver cirrhosis, autoimmune hepatitis, lupus hepatitis and inflammatory bowel disease; hemochromatosis; alpha-1-antotrypsin deficiency; chronic cholesta
  • hepatic cirrhosis is a disease caused by a hardened liver due to lesions of a parenchymal cell of the liver and increase of connective tissues.
  • MSC mesenchymal stem cell
  • 75 cell which is a pluripotent cell having a potential to be differentiated into all kinds of cytotypes with self -restoring ability.
  • the present invention is the first to report that a damaged liver cell can be replaced by an MSC, thereby recovering liver function and reducing collagen fibrils deposited into the liver.
  • liver fibrosis which was induced by dimethylnitrosamine (DMN).
  • the animal models had advanced liver fibrosis, low mortality and stable persistency, which thus had great potential as screen models for liver fibrosis medication ⁇ George J et al., Toxicology 2001 Jan. 2; 156(2-)
  • DMN was administered to 6-week-old male SD rats for 8 weeks to induce liver fibrosis.
  • ⁇ l-2> which will be described below, an accumulative survival rate of the DMN-induced group began to
  • liver weight per body weight LW/BW
  • 100 fibrosis may be effectively induced by DMN in rats. Accordingly, in Exemplary Embodiment 2, the inventors checked the effect of the MSCs on liver fibrosis in the DMN-induced liver fibrosis rats which were observed from the moment mortality began to decrease.
  • FIGS. 7 and 8 show that the administration of MSCs may significantly lower collagen fibrils.
  • the MSCs may substitute for damaged liver cells of a recipient, and decompose collagen fibrils around the damaged liver cell, thereby preventing and treating liver fibrosis and
  • the present invention provides a pharmaceutical composition for preventing and treating liver fibrosis including MSCs.
  • the present invention also provides a pharmaceutical composition for preventing and treating hepatic cirrhosis including MSCs.
  • the MSC the active ingredient of the pharmaceutical composition of the present invention, is a cell having an unexpressed histocompatibility antigen (HLA- DR; class II), the most critical cause of tissue or organ rejection.
  • HLA- DR histocompatibility antigen
  • the MSC which may be autologous or allogeneic, may not cause or may minimize the chance of immune reaction such as rejection, which can be a problem in 125 conventional transplant operations.
  • the MSC may be isolated from cord blood or adipose tissue of mammals including human.
  • cord blood refers to blood collected from the umbilical vein which connects a placenta to a fetus of a mammal.
  • adipose tissue refers to tissue including plural 130 cytotypes such as an adipocyte, a microvascular cell and the like. Also, the adipose tissue includes connective tissue storing adipose.
  • Cord blood may be easily collected from the umbilical vein of a donor after delivery. More particularly, in spontaneous vaginal delivery (SVD), cord blood may be collected from the umbilical vein which is completely out of a birth 135 canal, but the placenta still remains in a womb after delivery. Also, in Caesarean delivery, cord blood may be collected from the umbilical vein while the placenta is out of the womb after delivery.
  • SMD spontaneous vaginal delivery
  • cord blood may be collected from the umbilical vein while the placenta is out of the womb after delivery.
  • cord blood is collected from the umbilical vein connecting the placenta to a fetus after delivery by aseptic treatment.
  • the 140 cord blood may be collected before or after placental abruption in or out of the womb after the delivery.
  • the cord blood may be collected outside the womb after the placental abruption.
  • the cord blood is extracted from a cord blood collection bag containing warfarin using a probe 145
  • the adipose tissue may be obtained by a predetermined method well known to those of ordinary skill in the art.
  • the adipose tissue may be obtained by suction-assisted liposuction, ultrasonic-assisted liposuction, adipose tissue removal or combinations thereof.
  • suction-assisted liposuction the adipose tissue is collected by inserting a cannula in or around an adipose tissue storage
  • Adipose tissue removal includes the steps of incidentally collecting tissue containing adipose tissue (e.g., skin), that is, target tissue for an operation (e.g., skin in lipectomy or cosmetic surgery) together with the adipose tissue.
  • tissue containing adipose tissue e.g., skin
  • target tissue for an operation e.g., skin in lipectomy or cosmetic surgery
  • the MSCs may be isolated from the cord blood or the adipose tissue
  • the MSCs may be isolated by any conventional method well known to those of ordinary skill in the art.
  • the methods may include density gradient fractionation, immunoselection and differential adhesion separation.
  • Isolating and culturing MSCs from cord blood may be performed by any conventional method including one disclosed in Korean Patent application No. 10- 2002-0008639 (Pittinger MF, Mackay AM et al., Science 1999, 284:143-7; Lazarus HM, Haynesworth SE et al., Bone Marrow Transplant 1995, 16: 557-64).
  • the isolation of MSCs from cord blood may be performed
  • the MSC may be isolated by the steps of isolating cord blood cells into low-density mononuclear fractions, culturing the mononuclear cells to result in a confluency of 80% to 90% in appropriate conditions and subculturing the cells after being treated with trypsin.
  • the isolation and culture of the MSCs from the adipose tissue may be
  • the MSCs may be isolated by the steps of treating collagenase to the adipose tissue at a sufficient concentration, culturing the adipose tissue in appropriate conditions (temperature and time), isolating floating fat cells by centrifugation or another method well known to those of ordinary skill in the art, and tissue-culturing precipitating stromal fractions.
  • the isolated MSCs may be cultured in a cell culture medium well known to those of ordinary skill in the art, which may include DMEM medium, McCoys 5 A medium (Gibco), Eagle's basal medium, CMRL media, Glasgow minimal essential medium, Ham's F- 12 medium, Iscove's modified Dulbecco's medium, Liebovitz's L- 15 medium, and RPMI 1640 medium, but the present invention is not limited thereto.
  • At least one auxiliary element may be added when required, which may include: serum of calf, horse and human; antibiotics such as streptomycin sulfate and penicillin G for preventing contamination of microorganisms; and antifungal agents such as amphotericin B, gentamicin and nystatin.
  • the isolated MSCs may be stored by a method well known to those of ordinary skill in the art before use.
  • the MSCs may be cold-stored after cyroprotection treatment.
  • the cyroprotection treatment may be performed using a cyroprotective agent such as dimethyl sulfoxide (DMSO), glycerol, polyvinylpyrrolidine, polyethylene glycol, albumin, dextran, sucrose, ethylene glycol, i-erythritol, D-ribitol, D-mannitol, D-sorbitol, i-inositol, D-lactose or choline chloride.
  • DMSO dimethyl sulfoxide
  • glycerol polyvinylpyrrolidine
  • polyethylene glycol albumin
  • dextran sucrose
  • ethylene glycol i-erythritol
  • D-ribitol D-ribitol
  • D-mannitol D-mannitol
  • the pharmaceutical composition of the present invention including the MSC as an active ingredient may further include a pharmaceutically available carrier and diluent.
  • the pharmaceutically available carrier and diluent may be biologically and physiologically friendly to MSCs and a recipient.
  • the pharmaceutically available diluents may include saline water, soluble buffer, solvent and/or dispersing agent, but the present invention is not limited thereto.
  • the pharmaceutical composition of the present invention may be suitable
  • the present invention is not limited thereto.
  • the pharmaceutical composition of the present invention may be applied using a catheter.
  • cells may be injected through a catheter in a single lump or several smaller amounts. The injection of
  • the cells using a catheter may include delivery into a vein through, for example, a standard peripheral vein catheter, a central vein catheter or a pulmonary catheter.
  • the effective administration amount of the pharmaceutical composition of 210 the present invention may be determined by one skilled in the art according to the specific purpose described above in consideration of a variety of factors such as administration route, the number of treatments, and age, weight, health condition, gender, seriousness of the illness, diet and an evacuation rate of a patient.
  • a composition including a mesenchymal stem cell according to the present 220 invention can substitute for a damaged liver cell to recover liver function and reduce collagen fibrils deposited to the liver, and thus can be used for prevention and treatment of liver fibrosis or hepatic cirrhosis.
  • FIG. 1 illustrates an experimental method designed to induce liver fibrosis 225 in rats using dimethylnitrosamine (DMN).
  • FIG. 2 illustrates a measurement result of levels of total protein (A), albumin (B), alanine transaminase (ALT) (C) and aspartic acid transaminase (AST) (D) which are measured in second and fifth weeks after administration of DMN to rats.
  • A total protein
  • B albumin
  • ALT alanine transaminase
  • AST aspartic acid transaminase
  • FIG. 3 is a graph (D) illustrating per-area % (C) of collagen fibrils and liver weight per body weight in a DMN-induced group after the livers of 5 week 235 old DMN-induced rats are stained by Masson's trichrome staining (A: negative control, B: DMN-induced group).
  • FIG. 4 illustrates an experimental method designed to measure an effect of administration of mesenchymal stem cells (MSC) to DMN-induced liver fibrosis rats.
  • MSC mesenchymal stem cells
  • FIG. 5 is a graph illustrating an accumulated survival rate according to administration of MSCs into DMN-induced liver fibrosis rats.
  • FIG. 6 illustrates a measurement result of levels of total protein (A), albumin (B), ALT (C) and AST (D) according to injection of MSCs into DMN- induced liver fibrosis rats.
  • FIG. 7 illustrates a measurement result of liver weight per body weight according to administration of MSCs into DMN-induced liver fibrosis rats.
  • FIG. 8 shows a photograph of a liver stained by Masson's trichrome staining in one week after administration of MSCs into a DMN-induced liver fibrosis rat.
  • A Liver of normal rat
  • FIG. 9 shows a photograph of a liver stained by Masson's trichrome staining in 4 weeks after administration of MSCs into a DMN-induced liver fibrosis rat.
  • A Liver of normal rat
  • FIG. 10 illustrates image analysis results of livers stained by Masson's 270 trichrome staining in 1 week and 4 weeks after administration of MSCs into DMN- induced liver fibrosis rats. * denotes a significant difference from a value of the NC (P ⁇ 0.05).
  • FIG. 11 illustrates hybridization results in tissues in one week after administration of cord blood-derived MSCs to DMN-induced liver fibrosis rats. 275 A: H&E slide of liver tissue of cord blood-derived MSCs administered group in one week after the cell injection
  • Group II was a group having dimethylnitrosamine (DMN)-induced liver fibrosis.
  • the DMN diluted with 1% saline was applied to Group II at a concentration of l(l/g by abdominal injection for the first three days in each of 8 weeks (Hu QW et al., Life Sci. 2006 M 4;79(6):606- 300 612). Referring to FlG. 1, the DMN induction was terminated in the 8 th week when all rats were dead.
  • ⁇ l-2> General Test and Accumulated Survival Rate
  • the primary characteristics, 305 including ascites, jaundice, epistaxis, bilirubin urea and weight loss were checked every day, and survival rate was calculated.
  • autopsies were performed on animals within 24 hours after death. The animals that received an autopsy were not included in the measurement of accumulated survival rate.
  • the accumulated survival rate of the DMN- 310 induced liver fibrosis group began to decrease in the 5th week and abruptly decreased in the 6th week, and all of the remaining rats died in the 8th week.
  • liver fibrosis SD rats exhibited general clinical symptoms of low total
  • 335 DMN-induced group was decreased by approximately 50% of the negative control in the 5th week after the DMN induction.
  • Liver tissue of the rats used in Embodiment ⁇ 1-1> was immobilized with 10% neutral formalin, embedded and fragmented to stain it using H&E staining 340 and Masson's trichrome staining. To measure the area of collagen fibrils with respect to other tissues, the collagen fibrils were analyzed using Image-Pro Plus software (Media, Cybernetics, LP). Per-area % indicates a percentage of the area selected as a collagen fibril with respect to the total area.
  • the H&E staining result shows dilatations and thicknesses of portal triads, 345 arterioles and bile ducts.
  • a damaged liver cell of the DMN-induced group was replaced by a thick collagen fibril (not illustrated).
  • the Masson's trichrome staining shows that in the 5- week-old DMN- induced group collagen fibrils increased by a large amount over the negative
  • 365 MSCs (AD-MSCs group) were administrated, respectively.
  • the MSCs were injected into a tail vein at a concentration of IxIO 6 cell/100(l PBS per rat in the first week. Further, in the VC group, the same volume of vehicles (100(1 PBS) was injected into the same route.
  • the UCB-MSCs were treated by a method which has been disclosed by
  • the cells were cultured and proliferated for 4 to 6 weeks after the initial culture, and subcultured once or twice before use for a subsequent clinical experiment.
  • the human AD-MSCs were isolated from subcutaneous adipose tissue.
  • the adipose tissue was obtained from tissue removed after selective
  • the post-liposuction disposal tissue was periodically agitated for an hour at 37 ⁇ to be decomposed using lmg collagenase type I per ml of Dulbecco's modified Eagle's medium (DMEM; Gibco). Floating adipose cells were isolated from precipitating stromal fractions by centrifugation. The stromal cells were cultured in a tissue culture flask containing DMEM (Gibco) having 10% fetal
  • AD-MSC group were lower than that of the VC group to which only cell vehicles are administrated
  • the survival rates of the VC group, the UCB-MSC group and the AD-MSC group in the 4th week after the cell injection were 66.7%, 33.3% and 22.2%, respectively.
  • the rats to which human MSCs were transplanted lived for 4 weeks after the cell injection. It has been reported that a xenotransplanted recipient can have a variety of complications and may not live for more than one week. Thus, the reason for low survival rates of the UCB-MSC group and the AD-MSC group is graft versus host rejection due to the transplantation of the MSCs.
  • liver weight per body weight (LW/BW) of each group was measured.
  • LW/BW increased only in the AD-MSC group in the first week after the cell injection.
  • LW/BW decreased in the AD-MSC group in the fourth week after the cell injection, and highly increased in the VC group
  • the H&E staining result shows that the collagen fibril of the MSCs- administered group was thinner than that of the VC group.
  • the Masson's trichrome staining shows that the collagen fibrils in the UCB-MSCs group and the AD-MSCs group significantly decreased in number in the 450 first week after the cell injection (C and D of FIG. 8), however the collagen fibrils in the VC group slightly decreased in number (B of FIG. 8). In the UCB- MSC group and the AD-MSC group, the significant decrease of collagen fibrils was maintained until the fourth week after the cell injection (C and D of FIG. 9).
  • FIG. 10 shows that the per-area % of the collagen fibril in the UCB-MSC 455 group and the AD-MSC group is decreased 6% and 2% in the first week after the cell injection, respectively.
  • a DNA probe suitable for the human-specific AIu gene was formed by a
  • Genome DNAs of human UCB-MSCs were extracted using a DNAeasy tissue kit (Qiagen, Hilden, Germany).
  • a PCR primer was located at the most compatible area of the human AIu sequence, and produced a 224 bp PCR product ⁇ Kim J et al., Cell 1998 Aug 7;94(3):353-362; Cho
  • AIu sense primer (sequence No. 1) - Id ⁇
  • the PCR was performed under the following conditions: 25 cycles of 10 min at 95 0 C , 30 sec at 95V , 45 sec at 58r and 45 sec at 72 0 C , and 10 min at 72 1 C .
  • the PCR product was electrophoresed in a 2% Agaros gel, and stained 480 with ethidium bromide (lOng/ml). A 224bp DNA band was extracted using a QIAquick gel extraction kit (Qiagen). The PCR product was marked with DIG using a DIG probe synthesis kit (Roche Diagnostics, Basel, Switzerland). The PCR was performed on the 50ng extracted DNA using the same method as the above- described PCR protocol. The marked probe was purified by ethanol precipitation 485 according to the PCR protocol for the DIG probe synthesis.
  • tissue fragments were deparaffinated in xylene and then rehydrated with PBS. Subsequently, the rehydrated slide was cultured together with PBS 490 containing 0.3% tryptone X-100, and further cultured together with TE buffer containing 2mg/ml proteinase K for 30 min at 37 1 C , and then was washed three times for 5 min. The slide was acetylated two times for 5 min using TEA buffer containing 0.25% (v/v) acetic acid dehydrate (Sigma-Aldrich, St. Louis) in order to reduce non-specific background staining.
  • a composition including a mesenchymal stem cell according to the present 525 invention can substitute for a damaged liver cell to recover liver function and reduce collagen fibrils deposited to the liver, and thus can be used for prevention and treatment of liver fibrosis or hepatic cirrhosis.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Cell Biology (AREA)
  • Developmental Biology & Embryology (AREA)
  • Engineering & Computer Science (AREA)
  • Chemical & Material Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Immunology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Hematology (AREA)
  • Zoology (AREA)
  • Epidemiology (AREA)
  • Virology (AREA)
  • Biotechnology (AREA)
  • Biomedical Technology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Reproductive Health (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)

Abstract

Provided is a pharmaceutical composition for preventing and treating liver fibrosis and hepatic cirrhosis including a mesenchymal stem cell. The composition substitutes for a damaged liver cell, thereby recovering liver functions and reducing collagen fibrils deposited on the liver, and thus may be used for preventing and treating liver fibrosis or hepatic cirrhosis.

Description

PHARMACEUΉCAL COMPOSITION FOR PREVENTING AND TREATING LIVER FIBROSIS OR HEPATIC CIRRHOSIS COMPRISING MESENCHYMAL
STEM CELL
[Technical Held] The present invention relates to a pharmaceutical composition for preventing and treating liver fibrosis or hepatic cirrhosis comprising a mesenchymal stem cell. [Background Art]
A hepatic disease is one of the major causes of death in many countries (Corrao G et al., Int J Epidemiol. 1997 Feb;26(l):100-109). Generally, hepatic cirrhosis is a primary cause of death from liver diseases (Schlichting P et al., Scand J Gastroenterol. 1983 Oct;18(7): 881-888). In progression of hepatic cirrhosis, liver fibrosis is central and essential (Hu QW et al., Life Sci. 2006 M 4;79(6): 606-612). Liver fibrosis is a part of a wound healing process related to various types of liver damage, which is characterized by continuous deposition of collagen fibrils into an extra-cellular matrix (ECM) (Du WD et al., World J Gastroenterol. 1999 Oct; 5(5):397-403). Generally, liver fibrosis is reversible, which is different from hepatic cirrhosis, and has thin fibrils and no tuberculation. Thus, when the cause of the liver damage disappears, the liver can recover from liver fibrosis. However, chronically repetitive mechanisms of liver fibrosis may lead to irreversible hepatic cirrhosis in which crosslinking between connective tissues increases to accumulate thick fibrils, and a liver lobule loses its normal structure to cause tuberculation. Although a chronic liver disease may have various causes, it can eventually lead to liver fibrosis or hepatic cirrhosis irrespective of the cause. A liver disease may be asymptomatic in an initial state and thus be difficult to diagnose early. Furthermore, if the liver disease is discovered in a chronic state, it may be difficult to treat and have a high mortality rate. Also, effective medication has not yet been developed.
It has been noted that mesenchymal stem cells (MSCs) are derived from adult tissues including bone marrow (BM) (Cohnheim J. Ueber Entz W ndung und Eiterung, 1867; Ftedenstein AJ et al., J Embryol. Exp. Morphol, Dec 16 1996, 3:381-390; Pittenger MF et al., Science 1999 Apr 2, 284(5411):143-147), adipose Tissue (AD) (ZuJc PA et al., Tissue Eng. 2001 Apr; 7(2):211-228) and umbilical cord blood (UCB) (Eήces A et al., Br J Haematol. 2000 Apr; 109(l):235-242; Goodwin HS et al., Biol. Blood Marrow Transplant 2001;7(ll):581-588; Bieback K et al., Stem Cells 2004;22(4):625-634; Kogler G et al., J Exp. Med. 2004 M 19;200(2):l23-135; Lee OK et al., Blood 2004 Mar l;103(5):1669-1675). Human BM-derived multipotent adult progenitor cells (MAPCs) can be differentiated into hematocyte- like cells acquiring functional characteristics of hematocyte in vitro {.Schwartz RE et al., J Clin. Invest. 2002 May;109(10):1291-1302). Also, human MSCs directly xenotransplanted into a liver of a rat in vivo can be differentiated into human hematocytes without fusion (Sato Y. et al., Blood 2005 M 15; 106(2): 756-763). [Disclosure] [Technical Problem]
Studies which have been conducted for several years to develop medication for liver fibrosis and hepatic cirrhosis have found that a mesenchymal stem cell can substitute for a damaged liver cell, thereby recovering liver function and decreasing collagen fibrils deposited in the liver. Thus, the present invention is directed to a pharmaceutical composition for preventing and treating liver fibrosis including a mesenchymal stem cell.
The present invention is further directed to a pharmaceutical composition for preventing and treating hepatic cirrhosis including a mesenchymal stem cell. [Technical Solution] In one aspect, a pharmaceutical composition for preventing and treating liver fibrosis including a mesenchymal stem cell is provided.
In another aspect, a pharmaceutical composition for preventing and treating hepatic cirrhosis including a mesenchymal stem cell is provided.
Hereinafter, the present invention will be described in detail. The term "liver fibrosis" is a disease in which a thin fibril is abnormally proliferated in the liver, and may be caused by a variety of chronic toxic damage, including: chronic alcohol abuse; chronic exposure to drugs including acetaminophen, amiodarone, aspirin, azathioprine, isoniazid, methyldopa, methotrexate, nitrofurantoin, propylthiouracil and sulfonamide; chronic exposure to chemical agents including CCU, dimethyl nitrosamine, vinyl chloride, polychlorinated biphenyl, aflatoxin and insecticides; autoimmune diseases including viral infection, diabetes, primary sclerosing cholangitis, primary bile duct liver cirrhosis, autoimmune hepatitis, lupus hepatitis and inflammatory bowel disease; hemochromatosis; alpha-1-antotrypsin deficiency; chronic cholestasis; non-alcoholic steatohepatitis (NASH); chronic biliary atresia; Wilson's disease; and other states which have been known to cause cirrhosis.
The term "hepatic cirrhosis" is a disease caused by a hardened liver due to lesions of a parenchymal cell of the liver and increase of connective tissues. The term "mesenchymal stem cell (MSC)" refers to a type of adult stem
75 cell, which is a pluripotent cell having a potential to be differentiated into all kinds of cytotypes with self -restoring ability.
The present invention is the first to report that a damaged liver cell can be replaced by an MSC, thereby recovering liver function and reducing collagen fibrils deposited into the liver.
80 In order to demonstrate whether the MSC can prevent and treat liver fibrosis, the inventors used animal models having liver fibrosis which was induced by dimethylnitrosamine (DMN). The animal models had advanced liver fibrosis, low mortality and stable persistency, which thus had great potential as screen models for liver fibrosis medication {George J et al., Toxicology 2001 Jan. 2; 156(2-
85 3);129-138; Jezequel AM et al., J Hepatol. 1989 Jan;8(l):42-52).
In one exemplary embodiment of the present invention, DMN was administered to 6-week-old male SD rats for 8 weeks to induce liver fibrosis. Referring to Exemplary Embodiment <l-2> which will be described below, an accumulative survival rate of the DMN-induced group began to
90 decrease in the 5th week, and drastically fell from the 6th week, and eventually all rats were dead in the 8th week. Further, as can be seen in FIGS. 2A to 2D, the DMN-induced group exhibited common clinical symptoms commonly shown in rats having liver fibrosis, in which total protein and albumin levels are low and AST and ALT levels are high. Furthermore, it may be shown in D of FIG. 3 that
95 the DMN-induced group exhibits an approximately 50% decrease in liver weight per body weight (LW/BW), replacement of the damaged liver cell by a thick collagen fibril, and increase of collagen fibrils in number, as compared with the negative control. From the results described above, the inventors could confirm that liver
100 fibrosis may be effectively induced by DMN in rats. Accordingly, in Exemplary Embodiment 2, the inventors checked the effect of the MSCs on liver fibrosis in the DMN-induced liver fibrosis rats which were observed from the moment mortality began to decrease.
As can be seen in FIG. 4, the rats xenotransplanted with human MSCs
105 without administration of an immune inhibitor lived until the 4th week after the cell injection. Also, as can be seen in FIG. 5, total protein and albumin levels indicating synthesis ability of a liver in the liver fibrosis rats increased due to the administration of MSCs, and ALT and AST levels indicating a degree of necrosis of liver cells decreased.
110 Further, FIGS. 7 and 8 show that the administration of MSCs may significantly lower collagen fibrils.
Accordingly, the above experimental results showed that the MSCs may substitute for damaged liver cells of a recipient, and decompose collagen fibrils around the damaged liver cell, thereby preventing and treating liver fibrosis and
115 hepatic cirrhosis.
Consequently, the present invention provides a pharmaceutical composition for preventing and treating liver fibrosis including MSCs.
The present invention also provides a pharmaceutical composition for preventing and treating hepatic cirrhosis including MSCs.
120 The MSC, the active ingredient of the pharmaceutical composition of the present invention, is a cell having an unexpressed histocompatibility antigen (HLA- DR; class II), the most critical cause of tissue or organ rejection. Thus, the MSC, which may be autologous or allogeneic, may not cause or may minimize the chance of immune reaction such as rejection, which can be a problem in 125 conventional transplant operations. Preferably, the MSC may be isolated from cord blood or adipose tissue of mammals including human.
The term "cord blood" used herein refers to blood collected from the umbilical vein which connects a placenta to a fetus of a mammal.
The term "adipose tissue" used herein refers to tissue including plural 130 cytotypes such as an adipocyte, a microvascular cell and the like. Also, the adipose tissue includes connective tissue storing adipose.
Cord blood may be easily collected from the umbilical vein of a donor after delivery. More particularly, in spontaneous vaginal delivery (SVD), cord blood may be collected from the umbilical vein which is completely out of a birth 135 canal, but the placenta still remains in a womb after delivery. Also, in Caesarean delivery, cord blood may be collected from the umbilical vein while the placenta is out of the womb after delivery.
In the present invention, cord blood is collected from the umbilical vein connecting the placenta to a fetus after delivery by aseptic treatment. Here, the 140 cord blood may be collected before or after placental abruption in or out of the womb after the delivery. In a Caesarean delivery, the cord blood may be collected outside the womb after the placental abruption. After securing the umbilical vein, the cord blood is extracted from a cord blood collection bag containing warfarin using a probe 145 The adipose tissue may be obtained by a predetermined method well known to those of ordinary skill in the art. For example, the adipose tissue may be obtained by suction-assisted liposuction, ultrasonic-assisted liposuction, adipose tissue removal or combinations thereof. In suction-assisted liposuction, the adipose tissue is collected by inserting a cannula in or around an adipose tissue storage
150 existing in a patient and sucking out lipids into suction equipment. Adipose tissue removal includes the steps of incidentally collecting tissue containing adipose tissue (e.g., skin), that is, target tissue for an operation (e.g., skin in lipectomy or cosmetic surgery) together with the adipose tissue.
The MSCs may be isolated from the cord blood or the adipose tissue
155 which are obtained according to the method well known to those of ordinary skill in the art. The MSCs may be isolated by any conventional method well known to those of ordinary skill in the art.
For example, the methods may include density gradient fractionation, immunoselection and differential adhesion separation.
160 Isolating and culturing MSCs from cord blood may be performed by any conventional method including one disclosed in Korean Patent application No. 10- 2002-0008639 (Pittinger MF, Mackay AM et al., Science 1999, 284:143-7; Lazarus HM, Haynesworth SE et al., Bone Marrow Transplant 1995, 16: 557-64).
In one aspect, the isolation of MSCs from cord blood may be performed
165 by a method to be described below. The MSC may be isolated by the steps of isolating cord blood cells into low-density mononuclear fractions, culturing the mononuclear cells to result in a confluency of 80% to 90% in appropriate conditions and subculturing the cells after being treated with trypsin.
The isolation and culture of the MSCs from the adipose tissue may be
170 performed by any method well known to those of ordinary skill in the art.
In another aspect, the MSCs may be isolated by the steps of treating collagenase to the adipose tissue at a sufficient concentration, culturing the adipose tissue in appropriate conditions (temperature and time), isolating floating fat cells by centrifugation or another method well known to those of ordinary skill in the art, and tissue-culturing precipitating stromal fractions.
The isolated MSCs may be cultured in a cell culture medium well known to those of ordinary skill in the art, which may include DMEM medium, McCoys 5 A medium (Gibco), Eagle's basal medium, CMRL media, Glasgow minimal essential medium, Ham's F- 12 medium, Iscove's modified Dulbecco's medium, Liebovitz's L- 15 medium, and RPMI 1640 medium, but the present invention is not limited thereto. Also, in the present invention, at least one auxiliary element may be added when required, which may include: serum of calf, horse and human; antibiotics such as streptomycin sulfate and penicillin G for preventing contamination of microorganisms; and antifungal agents such as amphotericin B, gentamicin and nystatin.
The isolated MSCs may be stored by a method well known to those of ordinary skill in the art before use. Generally, the MSCs may be cold-stored after cyroprotection treatment. The cyroprotection treatment may be performed using a cyroprotective agent such as dimethyl sulfoxide (DMSO), glycerol, polyvinylpyrrolidine, polyethylene glycol, albumin, dextran, sucrose, ethylene glycol, i-erythritol, D-ribitol, D-mannitol, D-sorbitol, i-inositol, D-lactose or choline chloride.
Also, the pharmaceutical composition of the present invention including the MSC as an active ingredient may further include a pharmaceutically available carrier and diluent. The pharmaceutically available carrier and diluent may be biologically and physiologically friendly to MSCs and a recipient. The pharmaceutically available diluents may include saline water, soluble buffer, solvent and/or dispersing agent, but the present invention is not limited thereto. The pharmaceutical composition of the present invention may be suitable
200 for transdermal, muscular, abdominal, vascular, hypodermic, nasal, spinal or oral administration, but the present invention is not limited thereto. Also, the pharmaceutical composition of the present invention may be applied using a catheter. For example, in a peripheral vein method, cells may be injected through a catheter in a single lump or several smaller amounts. The injection of
205 the cells using a catheter may include delivery into a vein through, for example, a standard peripheral vein catheter, a central vein catheter or a pulmonary catheter.
The effective administration amount of the pharmaceutical composition of 210 the present invention may be determined by one skilled in the art according to the specific purpose described above in consideration of a variety of factors such as administration route, the number of treatments, and age, weight, health condition, gender, seriousness of the illness, diet and an evacuation rate of a patient.
215 Hereinafter, particular methods of the present invention will be described in detail with reference to exemplary embodiments, but the scope of the present invention is not limited to the embodiments described herein. [Advantageous Effects]
A composition including a mesenchymal stem cell according to the present 220 invention can substitute for a damaged liver cell to recover liver function and reduce collagen fibrils deposited to the liver, and thus can be used for prevention and treatment of liver fibrosis or hepatic cirrhosis. [Description of Drawings J FIG. 1 illustrates an experimental method designed to induce liver fibrosis 225 in rats using dimethylnitrosamine (DMN).
FIG. 2 illustrates a measurement result of levels of total protein (A), albumin (B), alanine transaminase (ALT) (C) and aspartic acid transaminase (AST) (D) which are measured in second and fifth weeks after administration of DMN to rats. 230 NC: Negative control
DMN-induced: DMN-induced liver fibrosis group
* denotes a significant difference from a value of the NC (P<0.05).
FIG. 3 is a graph (D) illustrating per-area % (C) of collagen fibrils and liver weight per body weight in a DMN-induced group after the livers of 5 week 235 old DMN-induced rats are stained by Masson's trichrome staining (A: negative control, B: DMN-induced group).
NC: Negative control
DMN-induced: DMN-induced liver fibrosis group
* denotes a significant difference from a value of the NC (P<0.05).
240 FIG. 4 illustrates an experimental method designed to measure an effect of administration of mesenchymal stem cells (MSC) to DMN-induced liver fibrosis rats.
FIG. 5 is a graph illustrating an accumulated survival rate according to administration of MSCs into DMN-induced liver fibrosis rats.
245 FIG. 6 illustrates a measurement result of levels of total protein (A), albumin (B), ALT (C) and AST (D) according to injection of MSCs into DMN- induced liver fibrosis rats. FIG. 7 illustrates a measurement result of liver weight per body weight according to administration of MSCs into DMN-induced liver fibrosis rats. 250 FIG. 8 shows a photograph of a liver stained by Masson's trichrome staining in one week after administration of MSCs into a DMN-induced liver fibrosis rat.
A: Liver of normal rat
B: Group into which only cell carriers are administered, Arrow: Collagen 255 fibril deposition and bridge necrosis
C: Cord blood-derived MSC administered group, Arrow: Decreased collagen fibril deposition and bridge necrosis
D: Adipose tissue-derived MSC administered group, Arrow: Decreased collagen fibril deposition and bridge necrosis
260 FIG. 9 shows a photograph of a liver stained by Masson's trichrome staining in 4 weeks after administration of MSCs into a DMN-induced liver fibrosis rat.
A: Liver of normal rat
B: Group into which only cell carriers are administered
265 C: Cord blood-derived MSC administered group, Black arrow: Decreased collagen fibril deposition and bridge necrosis
D: Adipose tissue-derived MSC administered group, White arrow: Decreased collagen fibril deposition and bridge necrosis
FIG. 10 illustrates image analysis results of livers stained by Masson's 270 trichrome staining in 1 week and 4 weeks after administration of MSCs into DMN- induced liver fibrosis rats. * denotes a significant difference from a value of the NC (P<0.05). FIG. 11 illustrates hybridization results in tissues in one week after administration of cord blood-derived MSCs to DMN-induced liver fibrosis rats. 275 A: H&E slide of liver tissue of cord blood-derived MSCs administered group in one week after the cell injection
B: Positive reaction (arrow) in the liver tissue of the cord blood-derived- MSCs injected group in one week after the cell injection [Mode for Invention]
280 Hereinafter, exemplary embodiments of the present invention will be described in detail. However, the present invention is not limited to the exemplary embodiments disclosed below, but can be implemented in various types. Therefore, the present exemplary embodiments are provided for complete disclosure of the present invention and to fully inform the scope of the present 285 invention to those of ordinary skill in the art. <Exemplary Embodiment 1> Induction of liver fibrosis using DMN <1-1> Animals and Experiment Design
Sprague-Dawley (SD) rats (5-weeks old) were purchased (SLC INC.,
290 Shizuoka-ken, Japan) and adapted to circumstances for 5 days. All rats were bred at a temperature of 22 + 311C and a humidity of 50 ±20%, and in a 12-hour lighting cycle. Water and feed were supplied to the rats ad libitum. Animals and experimental procedures were approved by the Korean Association for Laboratory
Animal Science (KALAS; Approval No. SNU-051227-2, Seoul, Korea).
295 After the 5-day adaptation, 69 rats were separated into 2 groups. Group
I (n=15) was a negative control (NC), and Group II was a group having dimethylnitrosamine (DMN)-induced liver fibrosis. The DMN diluted with 1% saline was applied to Group II at a concentration of l(l/g by abdominal injection for the first three days in each of 8 weeks (Hu QW et al., Life Sci. 2006 M 4;79(6):606- 300 612). Referring to FlG. 1, the DMN induction was terminated in the 8th week when all rats were dead.
<l-2> General Test and Accumulated Survival Rate To check whether liver fibrosis was induced in the rats by the DMN in Embodiment <1-1>, clinical symptoms of liver fibrosis, the primary characteristics, 305 including ascites, jaundice, epistaxis, bilirubin urea and weight loss were checked every day, and survival rate was calculated. Meanwhile, autopsies were performed on animals within 24 hours after death. The animals that received an autopsy were not included in the measurement of accumulated survival rate.
As a result of the test, the accumulated survival rate of the DMN- 310 induced liver fibrosis group began to decrease in the 5th week and abruptly decreased in the 6th week, and all of the remaining rats died in the 8th week.
<l-3> Measurement of Total Protein, Albumin, aspartic acid transaminase (AST) and alanine transminase (ALT) Levels
The liver fibrosis SD rats exhibited general clinical symptoms of low total
315 protein and albumin levels and high AST and ALT levels. Here, in the 2nd and
5th weeks after the DMN injection described in Embodiment <1-1>, blood was collected from ophthalmic venous plexus of three rats in Groups I and II, and their total protein, albumin, AST and ALT levels were measured.
The procedures described above were repeated three times separately, 320 and the results obtained therefrom are shown as average ± standard deviations. Statistical comparison was performed using a Turkey test after an ANOVA test. It is considered herein that P<0.05 is significant. As can be seen in A and B of FIG. 2, the total protein and albumin levels indicating synthesis ability of a liver were significantly lowered in the DMN-
325 induced group until the 5th week. In contrast, as can be seen in C and D of FIG.
2, the AST and ALT levels indicating a hepatocellular necrosis level were increased in the DMN-induced group.
<l-4> Measurement of Organ Weight
Autopsies were performed on the laboratory animals used in Embodiment 330 <1-1> within 24 hours after death, and then their weights of liver, spleen and kidney were measured.
In the results, the weights of spleens and kidneys in the DMN-induced group did not show significant differences from those in the negative control (the result is not illustrated). Meanwhile, liver weight per body weight (LWIEW) in the
335 DMN-induced group was decreased by approximately 50% of the negative control in the 5th week after the DMN induction.
<l-5> Histopathology and Image Analysis
Liver tissue of the rats used in Embodiment <1-1> was immobilized with 10% neutral formalin, embedded and fragmented to stain it using H&E staining 340 and Masson's trichrome staining. To measure the area of collagen fibrils with respect to other tissues, the collagen fibrils were analyzed using Image-Pro Plus software (Media, Cybernetics, LP). Per-area % indicates a percentage of the area selected as a collagen fibril with respect to the total area.
The H&E staining result shows dilatations and thicknesses of portal triads, 345 arterioles and bile ducts. As shown in the results of the test, a damaged liver cell of the DMN-induced group was replaced by a thick collagen fibril (not illustrated). The Masson's trichrome staining shows that in the 5- week-old DMN- induced group collagen fibrils increased by a large amount over the negative
350 control (A and B of FIG. 3). It also shows that per-area % of the collagen fibrils in the DMN-induced group reached approximately 9% in the 5th week (C of FIG.3).
<Exemplary Embodiment 2>
Effects of Administration of MSCs to Liver Fibrosis-hiduced Rats
355 <2-l> Animals and Experiment Design
In order to demonstrate an effect of mesenchymal stem cells (MSCs) in liver fibrosis rats, by the same method described in Embodiment <1-1>, 36 rats were treated with DMN to induce liver fibrosis. Referring to Embodiment <2-2> which will be described below, survival rate of the DMN-induced group began to
360 decrease in the 5th week, and drastically decreased in the 6th week, and thus the DMN induction was terminated in the 8th week. The rats with liver fibrosis induced by DMN were separated into three groups. Into Group III (n=12), only cell-vehicles (VC group) were administrated, and into Groups IV (n=12) and V (n=12), cord blood-derived MSCs (UCB-MSCs group) and adipose tissue-derived
365 MSCs (AD-MSCs group) were administrated, respectively. The MSCs were injected into a tail vein at a concentration of IxIO6 cell/100(l PBS per rat in the first week. Further, in the VC group, the same volume of vehicles (100(1 PBS) was injected into the same route.
The UCB-MSCs were treated by a method which has been disclosed by
370 the present inventors (Kim SW et al., Stem Cells 2006 Jun;24(6):1620-1626). A UCB sample was donated from a woman undergoing full-termed delivery at Boramae Hospital, Seoul. This experiment was approved by Boramae Hospital and the Seoul National University Institutional Review Board (IRB) (SNUIRB No. 0603/001-002, Seoul, Korea). Cord blood was defrosted at 37^C and separated into
375 low-density mononuclear fractions using Ficoll-Paque Plus (GE Healthcare AB, Uppsala, Sweden, www. amersham.com). After that, the mononuclear cells were washed and suspended with Dulbecco's modified Eagle's medium low glucose (Gibco, Grand Island, NY, www.invitrogen.com) containing 20% fetal bovine serum (Gibco), and then inoculated to a T-25 flask at a concentration ranging from
380 4xlO6 to 5xlOδ cells/cm2. This remained at 37"C in a humidified atmosphere containing 5% CO2. Three days later, the suspended cells were moved to a new flask. Every seven days, the medium was changed. The cells reaching a confluency of 80 to 90% were trypsinized with 0.005% trypsin/ethylene diamine tetra-acetic acid (Gibco) and subcultured, and then replated at 5xlO4
385 cells/cm2. The cells were cultured and proliferated for 4 to 6 weeks after the initial culture, and subcultured once or twice before use for a subsequent clinical experiment.
The human AD-MSCs were isolated from subcutaneous adipose tissue. The adipose tissue was obtained from tissue removed after selective
390 cosmetic liposuction. The post-liposuction disposal tissue was periodically agitated for an hour at 37 ± to be decomposed using lmg collagenase type I per ml of Dulbecco's modified Eagle's medium (DMEM; Gibco). Floating adipose cells were isolated from precipitating stromal fractions by centrifugation. The stromal cells were cultured in a tissue culture flask containing DMEM (Gibco) having 10% fetal
395 bovine serum (Gibco). Twelve hours later, the culture medium was substituted by defined Keratinocyte-SFM (Gibco) containing 5% fetal bovine serum. Every other day, the medium was exchanged with new one, and the cells were cultured until reaching a predetermined confluency. One week later, a sufficient number of cells were generated for trypsinization, and then stored in liquid nitrogen or
400 subcultured before use for a subsequent clinical experiment. <2-2> General Test and Accumulated Survival Rate In each group in Embodiment <2-l>, as primary characteristics of liver fibrosis, clinical symptoms including ascites, jaundice, epistaxis, bilirubin, urea, and weight loss were checked every day, and a survival rate was calculated.
405 As can be seen in FIG. 5, survival rates of the UCB-MSC group and the
AD-MSC group were lower than that of the VC group to which only cell vehicles are administrated The survival rates of the VC group, the UCB-MSC group and the AD-MSC group in the 4th week after the cell injection were 66.7%, 33.3% and 22.2%, respectively.
410 The rats to which human MSCs were transplanted lived for 4 weeks after the cell injection. It has been reported that a xenotransplanted recipient can have a variety of complications and may not live for more than one week. Thus, the reason for low survival rates of the UCB-MSC group and the AD-MSC group is graft versus host rejection due to the transplantation of the MSCs.
415 <2-3> Measurement of Total Protein, Albumin, AST and ALT Levels
In order to demonstrate whether liver function is recovered by administration of MSCs, blood samples were collected from an abdominal vein, and a clinical and biochemical experiment was performed by the same method as described in Embodiment <l-3>. The blood was collected from the abdominal
420 vein in the first to fourth weeks after the cell injection.
In the results, total protein and albumin levels in all groups began to increase after the cell injection. As can be seen in A and B of FIG. 6, the total protein and albumin levels of the UCB-MSC group in the fourth week after the cell injection were higher than those of the AD-MSC group and the VC 425 group. As can be seen in C and D of FIG. 6, the AST and ALT levels begin to decrease after the cell injection. The UCB-MSC group exhibited the most excellent recovery characteristic in the fourth week after the cell injection.
<2-4> Measurement of Organ Weight
In the first and fourth weeks after the cell injection described in 430 Embodiment <2-l>, liver weight per body weight (LW/BW) of each group was measured.
As a result of the test, LW/BW increased only in the AD-MSC group in the first week after the cell injection. LW/BW decreased in the AD-MSC group in the fourth week after the cell injection, and highly increased in the VC group
435 and the UCB-MSC group (refer to FIG. 7). The MSC-administrated group and the
VC group did not show any significant difference.
<2-5> Histopathology and Image Analysis
The samples obtained from the respective groups in Embodiment <2-l> were immobilized with 10% neutral formalin to be used for histopathological
440 analysis. The histopathology and image analysis were performed by the same method as described in Embodiment <l-5>. In order to measure the area of collagen fibrils with respect to another tissue, three slide-based images from the respective groups were analyzed using Image-Pro Plus software. Per-area % indicates a percentage of the area selected as collagen fibrils with respect to the
445 total area.
The H&E staining result shows that the collagen fibril of the MSCs- administered group was thinner than that of the VC group. The Masson's trichrome staining shows that the collagen fibrils in the UCB-MSCs group and the AD-MSCs group significantly decreased in number in the 450 first week after the cell injection (C and D of FIG. 8), however the collagen fibrils in the VC group slightly decreased in number (B of FIG. 8). In the UCB- MSC group and the AD-MSC group, the significant decrease of collagen fibrils was maintained until the fourth week after the cell injection (C and D of FIG. 9).
FIG. 10 shows that the per-area % of the collagen fibril in the UCB-MSC 455 group and the AD-MSC group is decreased 6% and 2% in the first week after the cell injection, respectively.
It can be seen from the results described above that MSCs can substitute for damaged liver cells of a recipient, and decompose collage fibers existing around the damaged liver cell. 460 <2-6> In situ Hybridization
In order to demonstrate homing of the administrated MSCs toward the damaged liver tissue, a series of tissue fragments were stained by H&E staining, and hybridized with a human-specific AIu gene in the tissue.
A DNA probe suitable for the human-specific AIu gene was formed by a
465 method which will be described below. Genome DNAs of human UCB-MSCs were extracted using a DNAeasy tissue kit (Qiagen, Hilden, Germany). A PCR primer was located at the most compatible area of the human AIu sequence, and produced a 224 bp PCR product {Kim J et al., Cell 1998 Aug 7;94(3):353-362; Cho
JJ et a/., Nat. Med. 2002 Sep;8(9):1033-1036; Walker JA et al., Anal. Biochem. 2003
470 Apr 1;315(1):122-128). In order to amplify the AIu sequence using PCR, primers to be listed below were used.
AIu sense primer (sequence No. 1) - Id ¬
s' ACG CCT GTA ATC CCA GCA CTT-3' AIu anti-sense primer (sequence No. 2) 475 5'-TCG CCC AGG CTG GAG TGCA-3'
The PCR was performed under the following conditions: 25 cycles of 10 min at 950C , 30 sec at 95V , 45 sec at 58r and 45 sec at 720C , and 10 min at 721C .
The PCR product was electrophoresed in a 2% Agaros gel, and stained 480 with ethidium bromide (lOng/ml). A 224bp DNA band was extracted using a QIAquick gel extraction kit (Qiagen). The PCR product was marked with DIG using a DIG probe synthesis kit (Roche Diagnostics, Basel, Switzerland). The PCR was performed on the 50ng extracted DNA using the same method as the above- described PCR protocol. The marked probe was purified by ethanol precipitation 485 according to the PCR protocol for the DIG probe synthesis.
The hybridization in tissue was performed by a method which will be described below. Right before performing the hybridization in tissue, tissue fragments were deparaffinated in xylene and then rehydrated with PBS. Subsequently, the rehydrated slide was cultured together with PBS 490 containing 0.3% tryptone X-100, and further cultured together with TE buffer containing 2mg/ml proteinase K for 30 min at 371C , and then was washed three times for 5 min. The slide was acetylated two times for 5 min using TEA buffer containing 0.25% (v/v) acetic acid dehydrate (Sigma-Aldrich, St. Louis) in order to reduce non-specific background staining. After pre-hybridization for 3 hours at 495 850C using a hybridization buffer in which formamide (Sigma-Aldrich), 0.1% sodium lauryl sarcosine (Sigma-Aldrich), 0.02% SDS (Sigma-Aldrich) and 2% blocking agent (Roche) are dissolved in 50% 5xSSC, the slide was further cultured for 10 min at 9AV together with a new hybridization buffer containing denaturalized DIG- marked DNA probe (10-200ng/ml). Then, the slide remained in ice for 10 min
500 and overnight-cultured at A2V . The pre-hybridization and hybridization steps were performed in a moist chamber containing 50% formamide. After the hybridization, the slide was simply washed with 2xSSC at room temperature, and washed three times with 0. IxSSC for 15 min at 42V . Visualization of the DIG- marked DNA probe was performed according to the DIG hexane detection kit
505 (Roche) protocol. The slide was blocked for 30 min with blocking buffer [1% blocking agent (Roche) dissolved in maleic acid buffer (0.1M maleic acid, 0.15M NaCl, pH 7.5)], and cultured for an hour together with an antibody solution coupled with alkaline phosphate [both (alkali and antibody) are dissolved in a blocking buffer containing 0.1% tryptone X-IOO at a ratio of 1:2000]. Then, the
510 slide was washed four times for 15 min with a maleic acid buffer, and stabilized for 5 min in a Tris buffer (pH 9.5). Color development was performed using a substrate solution [(nitroblue tetrazolium salt and 5-bromo-4-chloro-3-indolyl phosphate (Roche; pH 9.5) which were dissolved in a Tris buffer). The reaction was terminated with termination buffer (1OmM Tris-HCl, pH 8.0, ImM EDTA) after
515 enzyme reaction for 2 to 6 hours. The slide was washed three times for 15 min, and counterstained with 0.1% nuclear fast red (Sigma- Aldrich). The slide was washed three times with PBS.
As can be seen in the result shown in FIG. 11, in the first week after the cell injection, a positive signal (black arrow) was detected from the UCB-MSC
520 group. The signal was not detected or was weakly detected from the AD-MSC group in the first week, and the UCB-MSC group and AD-MSC group in the fourth week after the cell injection. [Industrial Applicability]
A composition including a mesenchymal stem cell according to the present 525 invention can substitute for a damaged liver cell to recover liver function and reduce collagen fibrils deposited to the liver, and thus can be used for prevention and treatment of liver fibrosis or hepatic cirrhosis.
While the invention has been shown and described with reference to certain exemplary embodiments thereof, it will be understood by those of ordinary
530 skill in the art that various changes in form and details may be made therein without departing from the spirit and scope of the invention as defined by the appended claims.

Claims

[CLAIMS] [Claim 1]
535 A pharmaceutical composition for preventing and treating liver fibrosis comprising a mesenchymal stem cell. [Claim 2]
The composition according to claim 1, wherein the mesenchymal stem cell is derived from cord blood or adipose tissue. 540 [Claim 3]
The composition according to claim 2, wherein the cord blood or the adipose tissue is isolated from a human. [Claim 4]
A pharmaceutical composition for preventing and treating hepatic cirrhosis 545 comprising a mesenchymal stem cell. [Claim 5]
The composition according to claim 4, wherein the mesenchymal stem cell is derived from cord blood or adipose tissue. [Claim 6]
550 The composition according to claim 5, wherein the cord blood or the adipose tissue is isolated from a human.
555
PCT/KR2007/002397 2007-05-16 2007-05-16 Pharmaceutical composition for preventing and treating liver fibrosis or hepatic cirrhosis comprising mesenchymal stem cell WO2008140141A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
PCT/KR2007/002397 WO2008140141A1 (en) 2007-05-16 2007-05-16 Pharmaceutical composition for preventing and treating liver fibrosis or hepatic cirrhosis comprising mesenchymal stem cell

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
PCT/KR2007/002397 WO2008140141A1 (en) 2007-05-16 2007-05-16 Pharmaceutical composition for preventing and treating liver fibrosis or hepatic cirrhosis comprising mesenchymal stem cell

Publications (1)

Publication Number Publication Date
WO2008140141A1 true WO2008140141A1 (en) 2008-11-20

Family

ID=40002328

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/KR2007/002397 WO2008140141A1 (en) 2007-05-16 2007-05-16 Pharmaceutical composition for preventing and treating liver fibrosis or hepatic cirrhosis comprising mesenchymal stem cell

Country Status (1)

Country Link
WO (1) WO2008140141A1 (en)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2018003997A1 (en) * 2016-07-01 2018-01-04 国立大学法人東北大学 Prophylactic or therapeutic agent for organ fibrosis
EP3381458A4 (en) * 2015-11-24 2019-07-24 Rohto Pharmaceutical Co., Ltd. Therapeutic agent for liver disease including adipose-tissue-derived stromal cells, and method for producing said therapeutic agent
WO2022042703A1 (en) * 2020-08-28 2022-03-03 广东乾晖生物科技有限公司 Method for preparing liver-like organs
CN115925920A (en) * 2022-08-04 2023-04-07 北京顺元天生物制品有限公司 Method for treating liver cirrhosis by gene-enhanced immune cells

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050014254A1 (en) * 2003-06-23 2005-01-20 Fraunhofer Gesellschaft Zur Forderung Der Angewandten Forschung E. V. Isolated adult pluripotent stem cells and methods for isolating and cultivating thereof
WO2005073366A1 (en) * 2004-01-30 2005-08-11 Lifecord Inc. Method for isolating and culturing multipotent progenitor/stem cells from umbilical cord blood and method for inducing differentiation thereof

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050014254A1 (en) * 2003-06-23 2005-01-20 Fraunhofer Gesellschaft Zur Forderung Der Angewandten Forschung E. V. Isolated adult pluripotent stem cells and methods for isolating and cultivating thereof
WO2005073366A1 (en) * 2004-01-30 2005-08-11 Lifecord Inc. Method for isolating and culturing multipotent progenitor/stem cells from umbilical cord blood and method for inducing differentiation thereof

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
MORIYA K. ET AL.: "Embryonic stem cells develop into hepatocytes after intrasplenic transplantation in CCl4-treated mice", WORLD JOURNAL OF GASTROENTEROLOGY, vol. 13, no. 6, February 2007 (2007-02-01), pages 866 - 873 *
OYAGI S. ET AL.: "Therapeutic effect of transplanting HGF-treated bone marrow mesenchymal cells into CCl4-injured rats", JOURNAL OF HEPATOLOGY, vol. 44, no. 4, April 2006 (2006-04-01), pages 742 - 748, XP025054428 *
ZHAN Y. ET AL.: "Differentiation of hematopoietic stem cells into hepatocytes in liver fibrosis in rats", TRANSPLANTATION PROCEEDINGS, vol. 38, no. 9, November 2006 (2006-11-01), pages 3082 - 3085, XP025008241 *

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3381458A4 (en) * 2015-11-24 2019-07-24 Rohto Pharmaceutical Co., Ltd. Therapeutic agent for liver disease including adipose-tissue-derived stromal cells, and method for producing said therapeutic agent
WO2018003997A1 (en) * 2016-07-01 2018-01-04 国立大学法人東北大学 Prophylactic or therapeutic agent for organ fibrosis
JPWO2018003997A1 (en) * 2016-07-01 2019-04-25 国立大学法人東北大学 Preventive or therapeutic agent for organ fibrosis
AU2017287529B2 (en) * 2016-07-01 2022-11-10 Life Science Institute, Inc. Prophylactic or therapeutic agent for organ fibrosis
WO2022042703A1 (en) * 2020-08-28 2022-03-03 广东乾晖生物科技有限公司 Method for preparing liver-like organs
CN115925920A (en) * 2022-08-04 2023-04-07 北京顺元天生物制品有限公司 Method for treating liver cirrhosis by gene-enhanced immune cells

Similar Documents

Publication Publication Date Title
KR100821128B1 (en) Method and system for preparing stem cells from fat tissue
Tsuji et al. Xenografted human amniotic membrane–derived mesenchymal stem cells are immunologically tolerated and transdifferentiated into cardiomyocytes
KR101176146B1 (en) Embryonic-like stem cells derived from post-partum mammalian placenta and uses and methods of treatment using said cells
US9498497B2 (en) Method of treating lung diseases using cells separated or proliferated from umbilical cord blood
CN109674819B (en) Placenta mesenchymal stem cell preparation and use thereof for treating sclerotic disease
Reinders et al. Bone marrow-derived mesenchymal stromal cells from patients with end-stage renal disease are suitable for autologous therapy
KR20060036933A (en) Cell differentiation of adipose-derived precursor cells
KR20090038494A (en) Post-partum mammalian placenta, its use and placental stem cells therefrom
WO2009103818A1 (en) Methods for obtaining progenitor cells and uses thereof in the treatment of tissue or organ damage
CN112111448B (en) Improved mesenchymal stem cell culture medium, bone marrow mesenchymal stem cell, and culture method and application thereof
JP2020534344A (en) Methods and compositions for treating epidermolysis bullosa
CN109646458B (en) Method for treating scleropathy using placental mesenchymal stem cell preparation
KR20090033328A (en) Immune privileged and modulatory progenitor cells
WO2008140141A1 (en) Pharmaceutical composition for preventing and treating liver fibrosis or hepatic cirrhosis comprising mesenchymal stem cell
EP1637589A1 (en) Novel stem cell derived from adult tissue and use thereof
CN113512525A (en) Mesenchymal stem cell preparation and application thereof
KR101843952B1 (en) Methods for isolation adipose-derived stromal vascular fraction from fat tissues
EP3494979A1 (en) Alleviation and treatment of ischemia reperfusion-induced lung injury using pluripotent stem cells
EP1669441A1 (en) Method of differentiating mesenchymal stem cell into liver cell and artificial human liver cell
Xu et al. Implantation of BM cells transfected with phVEGF 165 enhances functional improvement of the infarcted heart
KR20080101140A (en) Pharmaceutical composition for preventing and treating liver fibrosis or hepatic cirrhosis comprising mesenchymal stem cell
CN109749981B (en) Hepatocyte-like cells derived from human adipose-derived stem cells, and preparation method and application thereof
CA3168330A1 (en) Method for treating chronic graft versus host disease
EP3556850A1 (en) Treatment of diseases of endothelial dysfunction and inflammation
EP3591037A1 (en) Mesenchymal stem cells and therapeutic agent for liver disease

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: 07746545

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 07746545

Country of ref document: EP

Kind code of ref document: A1