US20100183568A1 - Method for production of hepatic-lobule-like cell cluster from adipose-tissue-derived cell - Google Patents

Method for production of hepatic-lobule-like cell cluster from adipose-tissue-derived cell Download PDF

Info

Publication number
US20100183568A1
US20100183568A1 US12/664,344 US66434408A US2010183568A1 US 20100183568 A1 US20100183568 A1 US 20100183568A1 US 66434408 A US66434408 A US 66434408A US 2010183568 A1 US2010183568 A1 US 2010183568A1
Authority
US
United States
Prior art keywords
hepatic
lobule
cell
substance
derived
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US12/664,344
Other languages
English (en)
Inventor
Akifumi Matsuyama
Hiroshi Komoda
Yoshiki Sawa
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
FOUNDATION OF BIOMEDICAL RESEARCH AND INNOVATION
Foundation for Biomedical Research and Innovation at Kobe
Original Assignee
Foundation for Biomedical Research and Innovation at Kobe
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 Foundation for Biomedical Research and Innovation at Kobe filed Critical Foundation for Biomedical Research and Innovation at Kobe
Assigned to FOUNDATION OF BIOMEDICAL RESEARCH AND INNOVATION reassignment FOUNDATION OF BIOMEDICAL RESEARCH AND INNOVATION ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: KOMODA, HIROSHI, SAWA, YOSHIKI, MATSUYAMA, AKIFUMI
Publication of US20100183568A1 publication Critical patent/US20100183568A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0652Cells of skeletal and connective tissues; Mesenchyme
    • C12N5/0662Stem cells
    • C12N5/0667Adipose-derived stem cells [ADSC]; Adipose stromal 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system

Definitions

  • the present invention relates to a method for obtaining a hepatic lobule cell population from an adipose-tissue-derived cell, a hepatic lobule cell population obtainable thereby, a method of screening for a substance that promotes or inhibits formation of hepatic lobule and for a substance that causes the activity of a hepatic lobule to increase or decrease, and a kit therefor, and the like.
  • Japan has a high incidence of hepatitis C. That chronic hepatitis, hepatic cirrhosis and then liver cancer occur due to infection by hepatitis C virus is a known fact. Medical therapies mainly centered on interferon are considered to be effective in chronic hepatitis and hepatic cirrhosis states. Meanwhile, once liver cancer occurs, a complete cure is medically impossible, leaving surgical extraction of the tumor as the number one choice. However, in reality, since advanced hepatic cirrhosis is to be the stage where liver cancer will occur, only surgical treatments can be selected also for liver failure, and even therapies such as TAE or PEIT cannot be attempted if the case becomes serious.
  • Liver transplantation has been performed on such serious hepatic cirrhosis patients for a long time.
  • cerebrally dead donor liver transplantation the absolute insufficiency of donors, and regarding living donor liver transplantation, ethical issues such as safety for the donor, and insertion of a surgical knife into a healthy subject exist.
  • ethical issues such as safety for the donor, and insertion of a surgical knife into a healthy subject exist.
  • the idea of a treatment of liver failure by hepatocyte regeneration was born.
  • Non-patent Reference 1 Since collection of adipose tissue is simple, only little ethical issues are considered to exist if the collected adipose-tissue-derived mesenchymal stem cell were used in not only the subject it is derived from, but even in a patient other than such subject.
  • Non-patent Reference 2 a report exists, in which a subcutaneous tissue-derived stem cell was used for causing differentiation into hepatocyte (Non-patent Reference 2). However, there was no report that a hepatic lobule could be regenerated, which is the smallest functional unit of the liver, that can be used in a treatment.
  • An object of the present invention is to provide a method for causing a hepatic lobule cell population, which is the smallest functional unit of the liver, to be formed from an adipose-tissue-derived cell, and a hepatic lobule cell population obtainable thereby.
  • a hepatic-lobule-like cell cluster may be caused to form with a high probability by culturing, and in particular by culturing in a suspended state, an undifferentiated cell obtained from adipose-tissue-derived cells, and reached completion of the present invention.
  • the present invention provides
  • the step of obtaining a hepatic-lobule-like cell cluster from an undifferentiated cell is a step of producing a hepatic-lobule-like cell cluster by culturing an undifferentiated cell in a suspended state
  • (9) a method for treating or preventing a disease that occurs due to deterioration in a liver function, said method comprising: administering the hepatic-lobule-like cell cluster described in (5) and/or the hepatocyte described in (6),
  • (10) a method of screening for a substance that promotes formation of hepatic lobule, said method comprising: adding a candidate substance to a culture medium when culturing an adipose-tissue-derived cell to obtain a hepatic-lobule-like cell cluster, wherein the candidate substance is a substance that promotes formation of hepatic lobule when formation of hepatic-lobule-like cell cluster is promoted in comparison with formation in a system not containing the candidate substance,
  • a method of screening for a substance that inhibits formation of hepatic lobule comprising: adding a candidate substance to a culture medium when culturing an adipose-tissue-derived cell to obtain a hepatic-lobule-like cell cluster, wherein the candidate substance is a substance that inhibits formation of a hepatic lobule when formation of hepatic-lobule-like cell cluster is inhibited in comparison with formation in a system not containing the candidate substance,
  • a method of screening for a substance that causes an activity of a hepatic lobule to increase comprising: culturing, in a culture medium containing a candidate substance, a hepatic-lobule-like cell cluster that has been obtained by culturing an adipose-tissue-derived cell, wherein the candidate substance is a substance that causes the activity of a hepatic lobule to increase when an activity of the hepatic-lobule-like cell cluster has increased in comparison with an activity in a system not containing the candidate substance,
  • a method of screening for a substance that causes the activity of a hepatic lobule to decrease comprising: culturing, in a culture medium containing a candidate substance, a hepatic-lobule-like cell cluster that has been obtained by culturing an adipose-tissue-derived cell, wherein the candidate substance is a substance that causes an activity of a hepatic lobule to decrease when the activity of the hepatic-lobule-like cell cluster has decreased in comparison with an activity in a system not containing the candidate substance,
  • kits for screening for a substance which causes an activity of a hepatic lobule to increase or decrease said kit being used in the method according to (15) or (17).
  • a method for obtaining a hepatic-lobule-like cell cluster from an adipose-tissue-derived cell, and a hepatic-lobule-like cell cluster obtainable thereby a method of screening for a substance that promotes or inhibits formation of hepatic lobule and a substance obtainable thereby which promotes or inhibits formation of hepatic lobule, a method of screening for a substance that causes the activity of a hepatic lobule to increase or decrease, and a substance obtainable thereby which causes the activity of a hepatic lobule to increase or decrease, as well as kits for such screenings, and the like, are provided.
  • FIG. 1 is a micrograph of the obtained adipose tissue-derived cells.
  • FIG. 2 is a micrograph of the obtained adipospheres.
  • FIG. 3 is a micrograph of the obtained hepatic-lobule-like cell cluster.
  • FIG. 4 is a graph showing the results of quantitative RT-PCR for ⁇ -fetoprotein.
  • the vertical axis represents the ratio of the expression of ⁇ -fetoprotein with respect to the expression of GAPDH.
  • FIG. 5 is a graph showing the results of quantitative RT-PCR for albumin.
  • the vertical axis represents the ratio of the expression of albumin with respect to the expression of GAPDH.
  • FIG. 6 is a graph showing the results of quantitative RT-PCR for keratin 18.
  • the vertical axis represents the ratio of the expression of keratin 18 with respect to the expression of GAPDH.
  • FIG. 7 is a graph showing the results of quantitative RT-PCR for keratin 19.
  • the vertical axis represents the ratio of the expression of keratin 19 with respect to the expression of GAPDH.
  • FIG. 8 is a graph showing the results of quantitative RT-PCR for CYP1B1.
  • the vertical axis represents the ratio of the expression of CYP1B1 with respect to the expression of GAPDH.
  • FIG. 9 is a graph showing the results of quantitative RT-PCR for glutamine synthase.
  • the vertical axis represents the ratio of the expression of glutamine synthase with respect to the expression of GAPDH.
  • FIG. 10 is the result of western blot analysis for the ⁇ -fetoprotein produced by the hepatic-lobule-like cell cluster.
  • FIG. 11 is the result of western blot analysis for albumin produced by the hepatic-lobule-like cell cluster.
  • FIG. 12 is a micrograph by immunohistochemical staining showing the presence of ⁇ -fetoprotein in a hepatic-lobule-like cell cluster.
  • FIG. 13 is a micrograph by immunohistochemical staining showing the presence of albumin in a hepatic-lobule-like cell cluster.
  • FIG. 14 is a fluorescence micrograph showing the incorporation of DiI-LDL in a hepatic lobule cell population.
  • FIG. 15 is a micrograph showing the accumulation of glycogen in a hepatic lobule cell population by PAS staining.
  • FIG. 16 is a graph showing the amount of urea generated by a hepatic-lobule-like cell cluster.
  • FIG. 17 is a graph showing the effects from the grafting of a hepatic-lobule-like cell cluster in a hepatitis mouse model.
  • the vertical axis represents total bilirubin concentration (mg/dL).
  • FIG. 18 micrograph by HE staining showing a survival of hepatic-lobule-like cell cluster after grafting of the hepatic-lobule-like cell cluster under the renal capsule[2] of mouse.
  • FIG. 19 is a micrograph by immunohistological staining showing the expression of albumin in a hepatic-lobule-like cell cluster after grafting.
  • FIG. 20 is a micrograph by PAS staining showing the accumulation of glycogen in a hepatic-lobule-like cell cluster after grafting.
  • FIG. 21 is a micrograph of ADMPC obtained from an adipose tissue.
  • FIG. 22 shows the expression of a neural crest[1] cell-specific marker gene from the results of RT-PCR using ADMPC-derived RNA.
  • FIG. 23 is an electrophoretic image showing the results of RT-PCR using ADMPC-derived RNA.
  • FIG. 24 shows the results of RT-PCR when ADMPC was sub-cultured six times.
  • FIG. 25 is a graph showing the amount of expression by quantitative PCR of Sca-1 in ADMPC.
  • the vertical axis represents Sca-1/GAPDH.
  • FIG. 26 is a graph showing the amount of expression by quantitative PCR of ABCG2 in ADMPC.
  • the vertical axis represents ABCG2/GAPDH.
  • FIG. 27 is the result of FACS analysis showing the expression of SSEA-4, CD29, CD44, CD73, CD105 and CD166 in ADMPC.
  • FIG. 28 is the result of FACS analysis showing that fibroblastic contamination is small in ADMPC compared to ADSC from a conventional method.
  • FIG. 29 is a micrograph of pancreatic endocrine cells obtained by causing ADMPC to differentiate.
  • FIG. 30 is a micrograph of hepatocytes obtained by causing ADMPC to differentiate.
  • FIG. 31 is a graph showing the amount of expression by quantitative PCR of ⁇ -fetoprotein in hepatocytes obtained by causing ADMPC to differentiate.
  • the vertical axis represents AFP ( ⁇ -fetoprotein)/GAPDH.
  • FIG. 32 is a graph showing the amount of expression by quantitative PCR of albumin in hepatocytes obtained by causing ADMPC to differentiate.
  • the vertical axis represents albumin/GAPDH.
  • FIG. 33 is a graph showing the amount of expression by quantitative PCR of CYP1B1 in hepatocytes obtained by causing ADMPC to differentiate.
  • the vertical axis represents CYP1B1/GAPDH.
  • FIG. 34 is a graph showing the amount of expression by quantitative PCR of glutamine synthase in hepatocytes obtained by causing ADMPC to differentiate.
  • the vertical axis represents glutamine synthase/GAPDH.
  • FIG. 35 shows the results of RT-PCR in cells cultured in the presence of DMSO or OP9 culture supernatant.
  • FIG. 36 is a graph showing the expression by quantitative PCR of Nk ⁇ 2.5 in cells cultured in the presence of DMSO.
  • the vertical axis represents Nk ⁇ 2.5/GAPDH.
  • FIG. 37 is a graph showing the expression by quantitative PCR of GATA-4 in cells cultured in the presence of DMSO.
  • the vertical axis represents GATA-4/GAPDH.
  • FIG. 38 is a graph showing the expression by quantitative PCR of ⁇ -CA in cells cultured in the presence of DMSO.
  • the vertical axis represents ⁇ -CA/GAPDH.
  • FIG. 39 is a graph showing the expression by quantitative PCR of MLC in cells cultured in the presence of DMSO.
  • the vertical axis represents MLC/GAPDH.
  • FIG. 40 is a graph showing the expression by quantitative PCR of MHC in cells cultured in the presence of DMSO.
  • the vertical axis represents MHC/GAPDH.
  • FIG. 41 is a strong magnification image of a sheet containing ADMPC-derived cardiac myoblasts.
  • FIG. 42 is an echocardiograph two weeks before grafting, before grafting, and four and 16 weeks after grafting of a sheet containing ADMPC-derived cardiac myoblast.
  • the top row is from MI controls and the bottom row is from a heart grafted with a sheet containing ADMPC-derived cardiac myoblast.
  • FIG. 43 is an echocardiograph two weeks before grafting, before grafting, and four and 16 weeks after grafting of a sheet containing ADMPC.
  • the top row is from MI controls and the bottom row is from a heart grafted with a sheet containing ADMPC.
  • FIG. 44 is a graph (unit of the vertical axis: mm) showing improvement of LVDd of a heart grafted with a sheet containing ADMPC-derived cardiac myoblasts (circle) and a sheet containing ADMPC (square).
  • FIG. 45 is a graph (unit of the vertical axis: mm) showing improvement of LVDs of a heart grafted with a sheet containing ADMPC-derived cardiac myoblasts (circle) and a sheet containing ADMPC (square).
  • FIG. 46 is a graph showing improvement of % EF of a heart grafted with a sheet containing ADMPC-derived cardiac myoblasts (circle) and a sheet containing ADMPC (square).
  • FIG. 47 is a graph showing improvement of % FS of a heart grafted with a sheet containing ADMPC-derived cardiac myoblasts (circle) and a sheet containing ADMPC (square).
  • FIG. 48 is a graph showing improvement of LVDs of a heart grafted with a sheet containing ADMPC-derived cardiac myoblasts (triangle) in comparison to a sheet containing ADMPC (square).
  • FIG. 49 is a graph showing improvement of % EF of a heart grafted with a sheet containing ADMPC-derived cardiac myoblasts (triangle) in comparison to a sheet containing ADMPC (square).
  • FIG. 50 is a HE staining image (100 ⁇ ) of a heart grafted with a sheet containing cardiac myoblast.
  • FIG. 51 is a HE staining image (100 ⁇ ) of a heart grafted with a sheet containing ADMPC.
  • FIG. 52 is a micrograph (100 ⁇ ) of a heart grafted with a sheet containing cardiac myoblast when immunostained using an anti-human ⁇ -CA antibody.
  • FIG. 53 is a micrograph (100 ⁇ ) of a heart grafted with a sheet containing ADMPC when immunostained using an anti-human ⁇ -CA antibody.
  • FIG. 54 is a micrograph (100 ⁇ ) of a heart grafted with a sheet containing cardiac myoblast when immunostained using an anti-human MHC antibody.
  • FIG. 55 is a micrograph (100 ⁇ ) of a heart grafted with a sheet containing ADMPC when immunostained using an anti-human MHC antibody.
  • FIG. 56 is a figure showing micrographs of structures of hearts after grafting of sheets containing ADMPC-derived cardiac myoblast and ADMPC when immunostained using an anti-human ⁇ -CA antibody.
  • FIG. 57 is a figure showing micrographs of structures of hearts after grafting of sheets containing ADMPC-derived cardiac myoblast and ADMPC when immunostained using an anti-human MHC antibody.
  • FIG. 58 is a figure representing the thickness of anti-human ⁇ -CA antibody-positive regions after grafting sheets containing ADMPC-derived cardiac myoblast and ADMPC.
  • FIG. 59 is a figure graphed with the thickness of anti-human ⁇ -CA antibody-positive regions after grafting sheets containing ADMPC-derived cardiac myoblast and ADMPC as the index.
  • FIG. 60 is a micrograph (100 ⁇ ) of a heart grafted with a sheet containing cardiac myoblast when immunostained with an anti-human HLA-ABC antibody.
  • FIG. 61 is a micrograph (100 ⁇ ) of a heart grafted with a sheet containing ADMPC when immunostained with an anti-human HLA-ABC antibody.
  • FIG. 62 is a figure comparing the states of differentiation into adipocytes for ADMPC and ADSC by oil red 0 staining.
  • FIG. 63 is a graph showing the content in lipids contained in an adipocyte obtained by causing ADMPC to differentiate.
  • the vertical axis represents lipid content (oil red 0 content/well).
  • FIG. 64 is a figure comparing the states of differentiation into bones for ADMPC and ADSC by alizarin red staining.
  • FIG. 65 is a figure comparing the states of differentiation into bones for ADMPC and ADSC by alkaline phosphatase activity.
  • the present invention relates to a method for obtaining a hepatic-lobule-like cell cluster from an adipose tissue-derived cell, comprising culturing adipose tissue-derived cells.
  • the present invention is exceptionally excellent on the point that, a cell population similar to a hepatic lobule, which is the minimum functional unit of the liver, can be produced.
  • Adipose tissue-derived cells refers to cells or cell population obtained from a visceral adipose tissue or a subcutaneous adipose tissue, or to cells or cell population induced to differentiate from stem cells such as mesenchymal hepatocytes[4] and ES cells and similar to cells contained in an adipose tissue in an organism.
  • adipose tissue-derived cells refer to any or all of adipose tissue-derived stem cells, adipose tissue-derived interstitial cells, adipose tissue-derived multipotent progenitor cells which is described late, adipose progenitor cells or cells similar to these, or a cell population containing a mixture comprising all or a portion thereof.
  • Adipose tissue-derived cell can be obtained from adipose tissues and the like by means/method well known to those skilled in the art.
  • the obtained adipose tissue-derived cell may be grown using means/method well known to those skilled in the art, for instance, to stabilize the phenotype.
  • the adipose tissue-derived cells may be grown by culturing the adipose tissue-derived cells in a culture medium containing dexamethasone and ascorbic acid, for instance, in a 60% DMEM (low glucose) and 40% MCDB201 culture medium added with ITS (10.0 mg/L insulin, 5.5 mg/L transferrin, 6.7 ng sodium selenite), 1 nM dexamethasone, 0.1 mM ascorbic acid, 10 ng/mL rhEGF, and 5% FCS, in an incubator such as fibronectin-coated dish.
  • the animal species from which the adipose tissue-derived cell is derived are not limited in particular, and are preferably, for instance, mammals including mouse, rat, rabbit, dog, cat, cow, horse, monkey and the like, and more preferably human. It is more desirable to use an adipose tissue-derived cell from an identical animal species or an identical individual to the animal species in which a disease is to be prevented or treated using the obtained hepatic-lobule-like cell cluster. Since adipose tissues are present in sufficient quantities in organisms and are obtained relatively readily, the present invention is exceptionally excellent compared to, for instance, methods in which hepatic lobules are obtained from limited materials such as corpses, and the like.
  • Hepatic-lobule-like cell cluster refers to hepatic lobule inside an organism and to cell population having similar function/morphology thereto, including, for instance, hepatocyte, biliary tract epithelial cell, endothelial cell, Kupffer cell, hepatic stellate cell and the like.
  • the hepatic-lobule-like cell cluster of the present invention is exceptionally excellent on the point that a sufficient quantity of secretory protein can be produced, the point that it has high metabolic capability, on the point that it has high detoxification capability, and the like.
  • it also has the advantage of being easily used, for example, in purposes such as grafting or the like, by causing a cell population to be formed.
  • the acquisition method of hepatic-lobule-like cell cluster from adipose tissue-derived cell of the present invention comprises as an important step the step of causing a hepatic-lobule-like cell cluster to be formed from an undifferentiated cell. It is desirable that the acquisition method of hepatic-lobule-like cell cluster from adipose tissue-derived cell of the present invention further comprises as an important step the step of obtaining an undifferentiated cell from adipose tissue-derived cells. These steps may be carried out one after the other or may be carried out in parallel.
  • Undifferentiated cell refers to a cell that is capable of differentiating into diverse cells, for instance, hepatic progenitor cell, pancreatic progenitor cell, cardiac muscle progenitor cell, vascular endothelial progenitor cell, osteoblast, chondroblast and the like.
  • the step of obtaining an undifferentiated cell may further comprise the step of growing the obtained undifferentiated cell.
  • the step of obtaining the undifferentiated cell may be carried out using known methods such as, for instance, sorting, MACS, method by antigen-antibody reaction such as rosette forming method, density gradient method, method of selecting based on morphology and single cell cloning, or may be carried out by culturing the adipose tissue-derived cell in a suspended state, causing an adiposphere to be formed. Since it allows already differentiated cell to be killed and undifferentiated cell to survive/grow, culturing the adipose tissue-derived cell in a suspended state, causing an adiposphere to be formed, is preferred. Regarding culturing in a suspended state, a description will follow.
  • an adiposphere is defined as a spheroid containing undifferentiated cell as the principal constituent. Since formation of adiposphere and the subsequent differentiation into a hepatic-lobule-like cell cluster may occur sequentially or coincidentally, an adiposphere may contain in addition to undifferentiated cell, hepatocyte, biliary tract epithelial cell, endothelial cell, Kupffer cell, hepatic stellate cell and the like.
  • the step of obtaining hepatic-lobule-like cell cluster is carried out by culturing undifferentiated cell in a culture medium containing, for instance, fibroblast growth factor, hepatocyte growth factor, oncostatin M, epithelial growth factor, and dimethylsulfoxide; preferably, such as step is carried out by culturing the undifferentiated cell in a suspended state. Culturing in suspension allows a cell population having a similar morphology to a hepatic lobule inside an organism to be obtained more readily.
  • Culture of undifferentiated cell in suspended state means placing and culturing the cell in a freed state by preventing or suppressing adhesion to the culture container.
  • Suspension of a cell can be carried out by a variety of well known means/methods.
  • cells may be placed in a suspended state using a culture container or apparatus treated to prevent or suppress adhesion of cells or made with such materials that prevent or suppress adhesion of cells.
  • culture container or apparatus low binding culture containers or the like exist, such as siliconized culture container (for instance, siliconized flask) or low binding culture dish (for instance, HydroCell (CellSeed)).
  • cells may be cultured in a suspended state using the hanging drop culture method.
  • well known means/method may be used in combination suitably at suspension starting time point or to continue suspension.
  • a temperature responsive culture equipment[5] for cell recovery for instance, RepCell (CellSeed)
  • the present invention relates to a hepatic-lobule-like cell cluster obtainable by the above-mentioned method.
  • the hepatic-lobule-like cell cluster of the present invention contains a hepatocyte.
  • carrying out the above method using cells collected from the adipose tissue of a subject, or a similar to the subject, having a disease that occurs due to a decrease in liver function such as hepatic cirrhosis or predisposition therefor, and grafting the obtained hepatic-lobule-like cell cluster to the subject allow diseases that occur due to a decrease in liver function such as hepatic cirrhosis to be treated or prevented, and the like.
  • the present invention relates to hepatocyte contained in the hepatic-lobule-like cell cluster obtainable by the above method.
  • the present invention relates to a medicinal composition for preventing or treating a disease that occurs due to a decrease in liver function, containing a hepatic-lobule-like cell cluster that obtainable by the above method and/or a hepatocyte contained in such a hepatic-lobule-like cell cluster.
  • a disease that occurs due to a decrease in liver function includes, disease that occurs due not only to a decrease but also to insufficiency in liver function, for instance, hepatitis, hepatic cirrhosis, liver cancer, hepatic insufficiency, drug liver damage, alcoholic liver damage, congenital metabolic anomaly, cholestatic liver damage and the like.
  • the hepatic-lobule-like cell cluster or the hepatocyte may be suspended in a suitable solution such as PBS.
  • the medicinal composition of the present invention may contain, in addition to the hepatic-lobule-like cell cluster or hepatocyte, a substance that promotes grafting thereof to liver, liver function improvement drug, suitable additive, diluent and the like.
  • the present invention relates to use of hepatic-lobule-like cell cluster obtainable by the above method and/or hepatocyte contained in such a hepatic lobule cell population, for the preparation of a medicinal product for preventing or treating a disease that occurs due to a decrease in liver function.
  • the present invention relates to a method for the treatment or prevention of a disease that occurs due to a decrease in liver function comprising administering a subject with the hepatic-lobule-like cell cluster obtainable by the above-mentioned culture method and/or hepatocyte contained in such a hepatic-lobule-like cell cluster.
  • a hepatic-lobule-like cell cluster or hepatocyte obtainable from an identical species or autologous adipose tissue-derived cell is used preferably in the present invention.
  • Hepatic-lobule-like cell cluster or hepatocyte may be grafted or injected, for instance, under the renal capsule[6], via portal vein, inside the liver, inside the great omentum, side the peritoneal cavity, inside the spleen, under the skin and the like.
  • the subject may be any one; it may be a human subject, or it may be a subject other than human, for instance, a mammal such as mouse or monkey.
  • the administration quantity, administration frequency and the like of the hepatic-lobule-like cell cluster or hepatocyte are selected suitably according to a variety of factors such as, for instance, the state of the subject, and the degree of seriousness of the disease.
  • the present invention relates to a method for decreasing the blood concentration of bilirubin comprising administering a hepatic lobule cell population and/or a hepatocyte contained in such a hepatic-lobule-like cell cluster.
  • a method for decreasing the blood concentration of bilirubin comprising administering a hepatic lobule cell population and/or a hepatocyte contained in such a hepatic-lobule-like cell cluster.
  • Such a method may be performed either in vitro or in vivo.
  • the present invention relates to a method of screening for a substance that promotes formation of hepatic lobule, comprising adding a candidate substance to the culture medium when culturing an adipose tissue to obtain a hepatic-lobule-like cell cluster, and showing that the candidate substance is a substance that promotes formation of hepatic lobule when formation of hepatic-lobule-like cell cluster has been promoted compared to formation in a system not containing the candidate substance.
  • a method also comprises a method of screening for a substance that promotes differentiation into hepatocyte.
  • candidate substances many exist and, for instance, analogs or derivatives of basic fibroblast growth factor, hepatocyte growth factor or oncostatin M, and the like, may be cited, without being limited to these.
  • the addition of a candidate substance to the culture medium when obtaining a hepatic-lobule-like cell cluster from an adipose tissue may be carried out once or several times at either or both of the step of obtaining an undifferentiated cell from an adipose tissue-derived cell and the step of obtaining a hepatic-lobule-like cell cluster from an undifferentiated cell.
  • hepatic-lobule-like cell cluster can be checked, for instance, by measuring the number of formed hepatic-lobule-like cell clusteres by microscopic observation, by quantifying ⁇ -fetoprotein, albumin and the like secreted in the culture supernatant, for instance, using ELISA, by measuring the expression of genes such as of ⁇ -fetoprotein, albumin, CYP1B1, glutamine synthase, keratin 18 and keratin 19 by quantitative PCR, or by measuring a marker substance of which the expression is known to decrease [7] or increase accompanying the differentiation into/formation of hepatic lobule, for instance, transthyretin, ⁇ 1-anti-trypsin, tyrosine aminotransferase, glucose-6-phosphatase and the like, by quantitative PCR or ELISA or the like.
  • the present invention relates to a substance obtainable by the above-mentioned screening method, which promptes formation of hepatic lobule.
  • the number of the obtained hepatic-lobule-like cell clusteres may be increased, or the speed of formation of hepatic-lobule-like cell cluster may be increased, by using such a substance in the method for obtaining a hepatic-lobule-like cell cluster from an adipose tissue-derived cell of the present invention.
  • such a substance may be used in the treatment or prevention of a disease that occurs due to a decrease in liver function.
  • Another further aspect of the present invention relates to a method of screening for a substance that inhibits formation of hepatic lobule, comprising adding a candidate substance to the culture medium when culturing an adipose tissue-derived cell to obtain a hepatic-lobule-like cell cluster, and showing that the candidate substance is a substance that inhibits formation of hepatic lobule when formation of hepatic-lobule-like cell cluster has been inhibited compared to formation in a system not containing the candidate substance.
  • a method also comprises a method of screening for a substance that inhibits differentiation into hepatocyte.
  • candidate substances many exist and, for instance, analogs or derivatives of drugs having hepatotoxicity such as carbon tetrachloride and phenobarbital, and the like, may be cited, without being limited to these.
  • a substance obtainable by such a screening method is sought to be suited for the treatment or prevention of a disease occurring due to liver hyperfunction.
  • addition of the candidate substance to the culture medium and means/method for evaluating formation of hepatic-lobule-like cell cluster are as described above.
  • the present invention relates to a substance obtainable by the above-mentioned screening method, which inhibits formation of a hepatic lobule.
  • the present invention relates to a kit used in the above-mentioned method of screening for a substance that accelerates or suppresses the formation of a hepatic lobule.
  • the kit of the present invention may contain cell acquisition means from an adipose tissue, a culture medium, a culture container, as well as means for checking the formation of the hepatic-lobule-like cell cluster, and the like. Normally, handling instructions are included with the kit. Using such a kit allows the above-mentioned screening to be carried out rapidly and readily.
  • the present invention relates to a method of screening for a substance that elevates the activity of a hepatic lobule, comprising culturing in a medium containing a candidate substance a hepatic-lobule-like cell cluster obtained by culturing an adipose tissue-derived cell, and showing that the candidate substance is a substance that elevates the activity of a hepatic lobule when the activity of the hepatic-lobule-like cell cluster has been increased compared to the activity in a system not containing the candidate substance.
  • Activity of a hepatic lobule refers to detoxification action, protein synthesis capability, metabolic action, and the like, of the hepatic lobule.
  • Elevation of the activity of a hepatic-lobule-like cell cluster can be checked, for instance, by quantifying ⁇ -fetoprotein, albumin and the like secreted in the culture supernatant using ELISA or the like, and from the increase or decrease of such protein quantities, or by measuring the expression of genes such as of ⁇ -fetoprotein, albumin, CYP1B1, glutamine synthase, keratin 18 and keratin 19 by quantitative PCR, and from the increase or decrease of the expression of such genes.
  • candidate substances many exist and, for instance, analogs or derivatives of basic fibroblastic growth factor, hepatocyte growth factor, or oncostatin M, and the like, may be cited, without being limited to these. Addition of candidate substance into the culture medium may be carried out once of multiple times.
  • the present invention relates to a substance obtainable by the above-mentioned screening method, which elevates the activity of a hepatic lobule.
  • the activity of the obtained hepatic-lobule-like cell cluster may be increased by using such a substance in the method for obtaining a hepatic-lobule-like cell cluster from an adipose tissue-derived cell of the present invention.
  • such a substance may be used in the treatment or prevention of a disease that occurs due to a decrease in liver function.
  • the present invention relates to a method of screening for a substance that diminishes the activity of a hepatic lobule, comprising culturing in a medium containing a candidate substance a hepatic-lobule-like cell cluster obtained by culturing an adipose tissue-derived cell, and showing that the candidate substance is a substance that diminishes the activity of a hepatic lobule when the activity of the hepatic-lobule-like cell cluster has been diminished compared to the activity in a system not containing the candidate substance.
  • candidate substances many exist and, for instance, carbon tetrachloride an phenobarbital, and the like, may be cited, without being limited to these.
  • the candidate substance into the culture medium means/method for evaluating the activity of hepatic-lobule-like cell cluster, the descriptions are as above.
  • the present invention relates to a substance obtainable by the above-mentioned screening method, which diminishes the activity of a hepatic lobule.
  • the present invention relates to a kit used in the above-mentioned method of screening for a substance that elevates or diminishes the activity of a hepatic lobule.
  • the kit of the present invention may contain cell acquisition means from an adipose tissue, a culture medium, a culture container, as well as means for checking the activity of the hepatic-lobule-like cell cluster, and the like. Normally, handling instructions are included with the kit. Using such a kit allows the above-mentioned screening to be carried out rapidly and readily.
  • the present invention relates to a cell population containing an adipose tissue-derived multipotent progenitor cell.
  • Adipose tissue-derived multipotent progenitor cell refers to a cell, which is a cell that can differentiate into a variety of cell lines such as of the endoderm, mesoderm and ectoderm, and expressing Islet-1, a marker for the absence of differentiation.
  • An adipose tissue-derived multipotent progenitor cell can be obtained from an embryonic stem cell or the like by causing differentiation, in addition to an adipose tissue.
  • the animal species from which the adipose tissue-derived multipotent progenitor cell is derived are not limited in particular, and are preferably, for instance, mammals including human, mouse, rat, rabbit, dog, cat, cow, horse, monkey and the like, and more preferably human. Or, the species similar to or identical to a subject to be treated by a regenerative medical therapy using such a cell population is desirable.
  • the cell population of the present invention has a low proportion of undesired contaminants, for instance, cells other than the adipose tissue-derived multipotent progenitor cell, such as erythrocytes and vascular endothelial cells, it has advantages such as ease of culture and high differentiation efficiency.
  • adipose tissue-derived multipotent progenitor cell for instance, the density method
  • means/methods using the difference with the adhesiveness of the adipose tissue-derived multipotent progenitor cell methods using for instance a chelator such as EDTA or an enzyme such as trypsin, antigen-antibody method such as sorting and MACS, methods that select based on morphology, single cell cloning, hemolysis method, and the like, may be cited.
  • the decrease in contaminants within the cell population may be verified, for instance, by quantifying a marker that the contaminant has using methods such as RT-PCR and ELISA, visually under a microscope, or by flow cytometry or immunohistological staining.
  • the cell population of the present invention preferably contains at least 20%, 30%, 40%, 50%, 60%, 70%, 75%, 80%, 85%, 90%, 93%, 96% or 99% adipose tissue-derived multipotent progenitor cell.
  • adipose tissue-derived multipotent progenitor cells in such proportions gives the cell population of the present invention advantages such as ease of maintenance of the adipose tissue-derived multipotent progenitor cells and high efficiency when [they are] caused to differentiate.
  • the cell population of the present invention may contain cells that are effective for the maintenance or differentiation of the adipose tissue-derived multipotent progenitor cells, such as, feeder cells, vascular endothelial cells, fibroblasts and the like. Inclusion of such cells may enhance the above advantages.
  • the present invention relates to a method, which is a method for obtaining an adipose tissue-derived multipotent progenitor cell from an adipose tissue, comprising the steps of (a) removing erythrocytes from an adipose tissue-derived cell population to obtain a preadipose tissue-derived multipotent progenitor cell population, and next, (b) removing cells other than an adipose tissue-derived multipotent progenitor cell from the preadipose tissue-derived multipotent progenitor cell population to obtain an adipose tissue-derived multipotent progenitor cell.
  • the present invention allows adipose tissue-derived cells other than the adipose tissue-derived multipotent progenitor cells to be decreased, and the adipose tissue-derived multipotent progenitor cells to be obtained at high yield and high purity.
  • the above steps may be carried out one after the other, or may be carried out in parallel.
  • the adipose tissue used in this aspect of the present invention may be either of a subcutaneous adipose tissue and a visceral adipose tissue from an organism.
  • the animal species from which the adipose tissue is derived are not limited in particular, and are preferably, for instance, mammals including human, mouse, rat, rabbit, dog, cat, cow, horse, monkey and the like, and more preferably human. Or, the species similar to or identical to the subject to be treated by a regenerative medical therapy using the adipose tissue-derived multipotent progenitor cell obtainable by the method of the present invention is desirable.
  • the adipose tissue-derived cell population used herein refers to a cell population containing at least an adipose tissue-derived multipotent progenitor cell.
  • the adipose tissue-derived cell population may contain, in addition to an adipose tissue-derived multipotent progenitor cell, erythrocytes, vascular endothelial cells, fibroblasts and the like.
  • the adipose tissue-derived cell population is obtained by treating an adipose tissue, for instance, with an enzyme such as collagenase, or by physical means/method, and/or eliminating lipids and the like, for instance, by centrifugal separation and filtration.
  • Erythrocytes have the properties of adsorbing adipose tissue-derived multipotent progenitor cells, whereby the yield of adipose tissue-derived multipotent progenitor cell may be decreased. Consequently, it is necessary to eliminate erythrocytes from the adipose tissue-derived cell population. Elimination of erythrocytes from the adipose tissue-derived cell population may be carried out by any means/method, for instance, it may be one carried out by a means/method other than one based on the difference in adhesiveness between the erythrocytes and cells other than these.
  • the density method is carried out by the density method, the hemolysis method or the filtration method, and more preferably, by the density method.
  • the density method may be carried out using a density solution with an adequate density, for instance, a commercially available density solution such as Lymphoprep. It suffices that the density of the density solution used is one between the densities of erythrocytes and cells other than these, preferably 1.063 to 1.119, more preferably 1.070 to 1.110 and most preferably 1.077.
  • the preadipose tissue-derived multipotent progenitor cell population used herein refers to a cell population containing at least an adipose tissue-derived multipotent progenitor cell.
  • the preadipose tissue-derived multipotent progenitor cell population may contain, in addition to the adipose tissue-derived multipotent progenitor cell, vascular endothelial cells, fibroblasts and the like.
  • the preadipose tissue-derived multipotent progenitor cell population in substance, does not contain erythrocytes.
  • the subsequent elimination of cells other than the adipose tissue-derived multipotent progenitor cell may be carried out readily and efficiently.
  • the cells other than the adipose tissue-derived multipotent progenitor cell used herein refer to adherent cells, or the like, such as vascular endothelial cells and fibroblasts.
  • elimination of cells other than the adipose tissue-derived multipotent progenitor cells from the preadipose tissue-derived multipotent progenitor cell population may be carried out by any means/methods, it is carried out using preferably a substance other than trypsin, more preferably a chelator such as EDTA or EGTA and most preferably EDTA.
  • such elimination is one that is carried out based on the difference in adhesiveness between the adipose tissue-derived multipotent progenitor cells and cells other than these.
  • cells other than adipose tissue-derived multipotent progenitor cells can be eliminated, for instance, by filter filtration[3] or the like. the purity and yield of the obtained adipose tissue-derived multipotent progenitor cell population increases by eliminating these cells.
  • the present invention relate to an adipose tissue-derived multipotent progenitor cell obtainable by the method for obtaining an adipose tissue-derived multipotent progenitor cell from an adipose tissue described above.
  • Such an adipose tissue-derived multipotent progenitor cell expresses Islet-1, as described above.
  • the present invention relates to a cell population containing an adipose tissue-derived multipotent progenitor cell obtainable by the method for obtaining an adipose tissue-derived multipotent progenitor cell from an adipose tissue described above.
  • a cell population in substance, does not contain undesirable adipose tissue-derived cells such as erythrocytes and vascular endothelial cells, but may contain cells that are effective for the maintenance/differentiation and the like of the adipose tissue-derived multipotent progenitor cells, such as, feeder cells.
  • cardiac myoblasts are provided by the present invention in sufficient amounts to constitute a sheet. Consequently, in one aspect, the present invention provides a sheet that contains cardiac myoblasts.
  • a cardiac myoblast refers to a cell that has been directed to differentiate into a cardiac myocyte, which is a cell expressing ⁇ -cardiac actin ( ⁇ -CA) and Myosin Light Chain (MLC).
  • the animal species from which the cardiac myoblast is derived are not limited in particular, and are preferably, for instance, mammals including human, mouse, rat, rabbit, dog, cat, cow, horse, monkey and the like, and more preferably human.
  • the species similar to or identical to the subject in which such sheet containing cardiac myoblasts is applied is desirable. For instance, by grafting a subject with a sheet containing cardiac myoblasts derived from the same species or the same animal as the subject, treating severe cardiac failure, or the like, becomes possible, without fearing a rejection reaction.
  • a sheet containing cardiac myoblasts refers to a cell population containing cardiac myoblasts as essential constituent.
  • the cardiac myoblast may be contained in either form of a mono-layer or a multi-layer. Having the morphology of a sheet allows for ease of handling when used for grafting or the like.
  • Constituents of the sheet other than cardiac myoblasts may be any, for instance, adipose tissue-derived stem cells, cardiac myocytes, cell scaffolds, vascular endothelium, matrix, and the like, may be cited.
  • the size and thickness of the sheet may be selected suitably according to a variety of conditions such as the extent of the injured area.
  • the proportion of cardiac myoblasts contained in a sheet is not limited in particular and may be selected suitably according to a variety of conditions such as, for instance, the state of the subject in which the sheet is applied.
  • the proportion is, for instance, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 100% and the like.
  • the proportion of cardiac myoblasts may be determined by quantifying ⁇ -CA or MLC, which are cardiac myoblast marker genes, using means known to those skilled in the art, for instance, quantitative RT-PCR.
  • the function of the sheet of the present invention can be checked by well known methods, for instance, by grafting a sheet to a subject, and by echography of the cardiac function of the grafted subject, or by measuring the diameter at end-diastole (LVDd), the diameter at end-systole (LVDs), the left ventricular ejection fraction (% EF) or the left ventricular internal diameter shortening fraction (% FS) and the like.
  • the present invention provides a method for obtaining cardiac myoblasts, comprising culturing an adipose tissue-derived stem cell.
  • cardiac myoblasts can be obtained in large amounts from an adipose tissue-derived stem cell.
  • the method may comprise the step of obtaining an adipose tissue-derived stem cell from an adipose tissue-derived cells.
  • An adipose tissue-derived stem cell refers to a cell, which is a cell that can differentiate into a variety of cell lines such as of the endoderm, mesoderm and ectoderm, and includes adipose tissue-derived multipotent progenitor cell (ADMPC), which expresses Islet-1, a marker for the absence of differentiation.
  • ADMPC adipose tissue-derived multipotent progenitor cell
  • the animal species from which the adipose tissue-derived stem cell is derived are not limited in particular, and are preferably, for instance, mammals including human, mouse, rat, rabbit, dog, cat, cow, horse, monkey and the like, and more preferably human. Or, the species similar to or identical to a subject to be treated with cardiac myoblasts obtained from such adipose tissue-derived stem cell is desirable.
  • the method for obtaining cardiac myoblasts of the present invention is one that comprises the step of culturing an adipose tissue-derived stem cell in the presence of a DMSO or OP9 culture supernatant.
  • the cell may differentiate and/or be induced into a cardiac myoblast.
  • the culture medium used in such culture may be selected suitably.
  • Such a culture medium may be one containing a variety of factors such as, for instance, retinoic acid, BMP2, BMP4, TGF ⁇ 2, HGF, bFGF, thyroxine, oxytocin or fatty acid concentrate.
  • Differentiation into cardiac myoblast can be checked by measuring the expression of a cardiac myoblast marker such as, for instance, ⁇ -CA or MLC by RT-PCR.
  • the present invention provides cardiac myoblast obtainable by the method for obtaining cardiac myoblasts described above.
  • cardiac myoblasts can be used in treatment of cardiac diseases such as myocardial infarction and cardiac myopathy, or can be used as materials for a sheet containing cardiac myoblasts.
  • the present invention provides a method, which is a method for obtaining a sheet containing cardiac myoblasts, comprising the following steps: (a) causing an adipose tissue-derived stem cell to differentiate into a cardiac myoblast, then, (b) causing a sheet containing cardiac myoblasts to form. These steps may be carried out one after the other or may be carried out in parallel. According to such a method, a sheet containing cardiac myoblasts can be obtained readily and efficiently.
  • the step of causing an adipose tissue-derived stem cell to differentiate into a cardiac myoblast is as described above.
  • the step of causing a sheet containing cardiac myoblasts to form from a cardiac myoblast may be achieved by means or method known by those skilled in the art.
  • the step may be achieved by causing a cardiac myoblast to multiply in an attached state to form a cell population and then peeling the formed cell population.
  • the number of cardiac myoblasts used and culture time may be selected suitably according to a variety of conditions, such as, the extent of the obtained sheet and the number of cardiac myoblasts contained in the sheet.
  • a sheet may be obtained by culturing 10 5 to 10 6 cardiac myoblasts for 24 to 72 hours. Peeling of the cell population may be carried out by a variety of means, for instance, physical stimulation.
  • the cardiac myoblasts may be cultured in a temperature-sensitive culture dish and incubated for instance at 20° C. or below to peel the cell population.
  • the present invention relates to a sheet containing cardiac myoblasts, obtainable by the method for obtaining a sheet containing cardiac myoblasts described above.
  • a sheet containing cardiac myoblasts can be used in the treatment of cardiac diseases such as myocardial infarction and cardiac myopathy.
  • the present invention relates to a method for treating and/or preventing a disease occurring due to a decline in the function of cardiac muscle, comprising grafting a sheet containing cardiac myoblasts or cardiac myoblasts to a subject.
  • Diseases occurring due to a decline in the function of cardiac muscle are, for instance, acute myocardial infarction, old myocardial infarction, ischemic myocardial infarction, dilated cardiac myopathy, congenital cardiac disease and the like. Grafting to a subject may be carried out by methods known to those skilled in the art.
  • It may be carried out, for instance, by grafting a sheet containing cardiac myoblasts to a region where function of cardiac muscle decreased, or by grafting cardiac myoblasts via the coronary artery.
  • the size and thickness of the sheet to be grafted, the number of cardiac myoblasts contained in the sheet, and the number of cardiac myoblasts may be selected suitably according to a variety of conditions such as the state of the subject and the extent of the injured region.
  • the present invention provides a composition containing a sheet containing cardiac myoblasts or cardiac myoblasts for treating and/or preventing disease caused by a decrease in cardiac function.
  • a composition may contain, in addition to the sheet or the cardiac myoblasts, for instance, PBS, a culture medium, a substance that promotes grafting, a cardiac function improver, a growth factor, a proliferation factor and the like.
  • the present invention relates to a method, which is a method of screening for a substance that promotes differentiation into cardiac myoblast, comprising the following steps:
  • candidate substances many exist and analogs and derivatives of, for instance, retinoic acid, BMP2, BMP4, TGF ⁇ 2, HGF, bFGF, thyroxine, or oxyton[8], and the like, may be cited, without limiting to these.
  • Differentiation of adipose tissue-derived stem cell into cardiac myoblast is as described above. Differentiation into cardiac myoblast can be checked by a variety of methods, for instance, by measuring the expression of a cardiac myoblast marker such as ⁇ -CA or MLC by RT-PCR.
  • the present invention relates to a substance that promotes differentiation into cardiac myoblast, which can be obtained by the method of screening for a substance that promotes differentiation into cardiac myoblast described above.
  • the substance may be used in the production method for the cardiac myoblast and sheet containing cardiac myoblasts described above to increase the number of the obtained cardiac myoblasts and sheets. Or, such a substance may be used in the treatment or prevention of a disease caused by a decrease in cardiac function.
  • the present invention relates to a method, which is a method of screening for a substance that inhibits differentiation into cardiac myoblast, comprising the following steps:
  • candidate substances many exist and analogs and derivatives of, for instance, noggin, and the like, may be cited, without limiting to these.
  • the present invention relates to a substance that inhibits differentiation into cardiac myoblast, which can be obtained by the method of screening for a substance that inhibits differentiation into cardiac myoblast described above.
  • the substance may be used in the treatment or prevention of a disease caused by an elevation in cardiac function.
  • the present invention relates to a kit for screening for a substance that promotes or inhibits differentiation into cardiac myoblast.
  • the kit of the present invention may contain an adipose tissue-derived stem cell, a culture medium, a culture container, as well as means for checking differentiation into cardiac myoblast and the like. Normally, handling instructions are included with the kit. Using such a kit allows the above-mentioned screening to be carried out rapidly and readily.
  • the present invention provides a method, which is a method of screening for a substance that promotes or inhibits formation of a sheet containing cardiac myoblasts, comprising the following steps:
  • the candidate substance is a substance that promotes or inhibits formation of a sheet containing cardiac myoblasts when the formation has been promoted or inhibited compared to that formed in a system not containing the candidate substance.
  • the method for checking the formation of sheet is as described above.
  • the addition of candidate substance may be carried out in both steps (a) and (b) described above, or may be carried out in either one.
  • the present invention relates to a substance that promotes or inhibits formation of a sheet containing cardiac myoblasts, which can be obtained by the screening method for a substance that promotes or inhibits formation of a sheet containing cardiac myoblasts described above.
  • the present invention relates to a kit for screening for a substance for promoting or inhibiting formation of a sheet containing cardiac myoblasts described above.
  • a human adipose tissue was sliced into 2 to 3 mm 2 -large fragments and digested using collagenase I.
  • the digestate was cultured for 24 to 36 hours in DMEM containing 10% FBS and antibiotics and treated with 0.02% EDTA to obtain adipose tissue-derived cells.
  • the obtained adipose tissue-derived cells were amplified by 3 to 5 passage cultures in a culture medium containing 60% DMEM (low glucose), 40% MCDB201, 1 ⁇ ITS (10.0 mg/L insulin, 5.5 mg/L transferrin, 6.7 ng sodium selenite), 10 ng/mL rhEGF, 1 nM dexamethasone, 0.1 mM ascorbic acid and 5% FCS (Hyclone), in a fibronectin-coated dish.
  • a micrograph of adipose tissue-derived cells is shown in FIG. 1 .
  • the obtained adiposphere was washed 2 to 3 times in PBS (centrifugation at 1000 to 1200 rpm) and cultured for 3 to 4 weeks in a culture medium containing 60% DMEM (low glucose), 40% MCDB201, 1 ⁇ ITS, 1 nM dexamethasone, 10011M ascorbic acid, 10 ng/mL rhEGF, bFGF, HGF and OSM (oncostatin M), in a low binding culture dish (Hydrocell; CellSeed). From day 10 of the beginning of the culture, 0.1% DMSO was added to obtain a hepatic-lobule-like cell cluster. A micrograph of the obtained hepatic-lobule-like cell cluster is shown in FIG. 3 .
  • RNA from the hepatic-lobule-like cell cluster was carried out using RNeasy Protect Mini Kit (QIAGEN), as follows.
  • the hepatic-lobule-like cell cluster was recovered, and the buffer RLT containing 10 ⁇ l/ml 2-mercaptoethanol (Naacalai Tesqu[13]) was added at a proportion of 600 ⁇ l/10 7 cell.
  • Cells were homogenized by pipetting with a 20G needle and then 600 ⁇ l of 70% ethanol was added. Transferred onto an RNeay[14] Mini column inside a 2 ml collection tube were 700 ⁇ l of the obtained mixed solution, which was centrifuged at 1000 rpm for 15 seconds.
  • buffer RW1 350 ⁇ l of buffer RW1 was added onto the column and centrifuged at 1000 rpm for 15 seconds.
  • Added to 70 ⁇ l of buffer RDD were 10 ⁇ l of DNase I stock solution (QIAGEN), which were tumble-mixed, added to the RNeasy silica gel membrane inside the RNeasy Mini column, and incubated at room temperature for 15 minutes.
  • the 2 ml collection tube was replaced with a new one.
  • Added onto the column was 500 ⁇ l of buffer RPE, and centrifugation was performed at 1000 rpm for 15 seconds.
  • RNA solution 0.5 ⁇ l of 0.5 mM Random Primer (Invitorogen[15]) and 1 ⁇ l of 10 mM dNTPmix (Invitorogen[16]) were added, reacted at 65° C. for 5 minutes, and then cooled on ice.
  • 4 ⁇ l of 5 ⁇ First-Strand buffer (Invitorogen[17]), 1 ⁇ l of 0.1M DTT (Invitorogen[18]), 1 ⁇ l of RNaseOUT (Invitorogen[19]), and 1 ⁇ l of SuperScript III RT (Invitorogen[20]) were added, reacted at 25° C. for 5 minutes, at 50° C. for 60 minutes and at 70° C. for 15 minutes to prepare a single-stranded cDNA.
  • the obtained cDNA was stored at 4° C. until use.
  • Hepatic-lobule-like cell cluster was washed three times with PBS (Nacalai Tesque), and then M-PER (PIERCE) was added. Cells were lysed by ultrasonication, centrifuged at 14000 g for 15 minutes to eliminate insoluble cell constituents. Sample buffer (Nacalai Tesque) was added in the same amounts as the sample, boiled at 100° C. for 5 minutes and ice-cooled. The protein concentration in the obtained sample was measured using BCA Protein Assay Reagent (PIERCE).
  • Gel mini plate for electrophoresis (PAG mini “Daiichi”; Daiichi Pure Chemicals Co.) and running buffer were used to perform SDS-PAGE.
  • the amount of protein used was 5 ⁇ g.
  • the electrophoresis conditions were 10 mA in the stacking gel and 40 mA in the running gel.
  • the electrophoresed gel above was washed in blotting buffer for 10 minutes. Next, the proteins in the gel were copied onto a nitrocellulose membrane by wet blotting (100 mA, overnight).
  • the membrane was washed for 10 minutes in PBS containing 0.1% Tween20.
  • Blocking One Nacalai Tesque
  • BETHYL Human Albumin antibody
  • Alpha Fetoprotein Ab-2 LAB Vision
  • Washing was performed for 15 minutes in PBS containing 0.1% Tween20 (three times).
  • Reaction was performed for one hour with a 1000-fold diluted solution of polyclonal pig anti-rabbit immunoglobulin/HRP or polyclonal rabbit anti-goat immunoglobulin/HRP as secondary antibody. Washing was performed for 15 minutes in PBS containing 0.1% Tween20 (three times).
  • the bands were detected using ECL Plus Western Blotting Detection Reagents. The results are shown in FIGS. 10 and 11 .
  • the hepatic-lobule-like cell cluster obtained by methods of the present invention was found to produce sufficient amounts of ⁇ -fetoprotein and albumin.
  • the hepatic-lobule-like cell cluster was washed three times was PBS (Nacali[22] Tesque) and centrifuged. The obtained pellet was embedded in Tissue-Tek OCT-compound (Sakura Fineteck Inc.) and conserved at ⁇ 30° C. Using a cryostat, 7 ⁇ m sections were prepared, pasted on glass and conserved at ⁇ 30° C.
  • Human LDL (density: 1.019 to 1.063 g/ml) was isolated. The isolation was carried out from a donor having normal lipoproteins by subjecting the same [23] to ultracentrifugation sequentially, dialysis with saline-EDTA, and then sterilization by filtration with a 0.2 ⁇ m filter. Next, the above LDL was incubated with 1,1′-dioctadecyl-3,3,3′,3′-tetramethylindocarbocyanine (DiI; Molecular Probes) (3 mg/ml) in 100 ml of DMSO at 37° C. for 8 hours in 0.5% bovine serum albumin (BSA)/PBS to label the lipoprotein (LDL) with DiI. Thereafter, this lipoprotein was dialyzed with PBS and filtered before use.
  • BSA bovine serum albumin
  • DiI-LDL a hepatic lobule cell population, which was differentiated from ADMPC, was incubated in serum-free DMEM containing 10 ⁇ g/ml DiI-LDL at 37° C. for 3 hours. Next, the cells were washed three times and mounted over Permaflur[24]. This slide was checked using a confocal laser scanning microscope (Floview FV1000, Olympus). The results are shown in FIG. 14 . It is clear from FIG. 14 that DiI-LDL has been incorporated massively in the cytoplasmic regions of the hepatic lobule cell population. From this, it could be confirmed that the hepatic-lobule-like cell cluster obtained by the present invention had a function for incorporating LDL.
  • a differentiated hepatic-lobule-like cell cluster was fixed with 4% paraformaldehyde and embedded in paraffin.
  • the sample was sliced in a thickness of 5 ⁇ m to prepare a section. Thereafter, this section was oxidized with 1% periodic acid for 5 minutes and rinsed three times with deionized water (dH 2 O). Next, treatment with a Schiff reagent for 15 minutes and rinsing for 5 to 10 minutes with dH 2 O were performed. Furthermore, this section was counterstained for 1 minute with Mayer's haematoxylin, rinsed with dH 2 O and observed with a light microscope. The results are shown in FIG. 15 . From FIG.
  • the hepatic-lobule-like cell cluster obtained by the present invention was shown to have a function for accumulating glycogen.
  • a hepatic-lobule-like cell cluster was incubated for two hours in 5 ml of Hank's balanced salt solution (Gibco) containing 5 mM NH 4 Cl.
  • the urea concentration in 0.5 ml of supernatant was measured using QuantiChrom Urea Assay Kit (Bioassay Systems). The obtained concentration was multiplied by the total volume of supernatant to calculate the total amount of urea generated.
  • HepG2 was used as control.
  • the obtained total amount of urea generated was divided by the overall DNA amount to calculate the overall amount of urea generated.
  • the results are shown in FIG. 16 .
  • the hepatic-lobule-like cell cluster was found to generate a sufficient amount of urea compared to HepG2. From this, it could be confirmed that the hepatic-lobule-like cell cluster obtained by the present invention had sufficient detoxification action.
  • a mouse hepatitis model was prepared by intraperitoneal injection into NOD-SCID mouse of carbon tetrachloride (CCl 4 ) at 300 ⁇ l/kg, twice weekly and for 12 weeks.
  • a hepatic-lobule-like cell cluster was washed with Hank's balanced salt solution, centrifuged and pelleted.
  • the above mouse was anaesthetized with sevofluene[25].
  • a celiotomy [26] was performed by left paramedian incision, the left kidney was exposed, the renal capsule[27] separated to create a pocket.
  • the pelletized cell population was injected and grafted inside the created pocket.
  • the abdominal wall was closed in two layers.
  • kidney grafted with hepatic-lobule-like cell cluster was also used to perform immunofluorescence staining for albumin.
  • the tissue was placed in OCT-compound (Sakura Fineteck Inc.) and frozen immediately.
  • a 7 ⁇ m section was prepared and fixed in 4% paraformaldehyde/PBS (WAKO) for 30 minutes. This fixed section was incubated with a blocking solution (Blocking One; Nacalai Tesque).
  • Excessive adipose tissue was extracted during gastric cancer operation from ten subjects (four males and six females) from whom informed consents were received.
  • the protocol was according to Osaka University graduate School of Medicine Review Boards for Human Research. All subjects were fasted for at least 10 hours. The age of the subjects was 55 ⁇ 5 years (average ⁇ SE; 40 to 60 years range). There were no subject being administered with a steroidal agent or TZD. From the subjects, 1 to 10 g of abdominal subcutaneous (outside of fascial surface) adipose tissue and dorsal mesogastrium adipose tissue.
  • adipose tissue was sliced, and then digested in Hank's buffered saline solution (HBSS) containing 0.075% collagenase (Sigma Chemical Co.) for one hour while shaking in a 37° C. water bath.
  • HBSS Hank's buffered saline solution
  • ADMPCs were plated in Culture Medium I: 60% DMEM-low glucose, 40% MCDB201, 10 ⁇ g/mL EGF, 1 nM dexamethasone, 100 ⁇ M ascorbic acid, and 5% FBS, at a density of 10,000 cell/cm 2 over human fibronectin coated dishes, subcultured 3 to 5 times, and used in the experiments.
  • the micrograph of ADMPC cultured for 10 days is shown in FIG. 21 .
  • ADSC adipose tissue-derived stem cell
  • ADMPC was shown to express a marker of absence of differentiation, in particular Islet-1, known as a marker for cardiac, hepatic and pancreatic progenitor cells.
  • ADSC did not express Islet-1, which confirms that the obtained ADMPC is a different cell from ADSC.
  • ADMPC was checked for the expression of Sca-1 and ABCG2, which are markers for the absence of differentiation, by as thus described carrying out real time PCR with Applied Byosystems[11] 7900 Fast Real-Time PCR system.
  • the TaqMan probes used are shown in Table 3.
  • the results are shown in FIGS. 25 and 26 .
  • ADMPC was confirmed to express Sca-1 and ABCG2.
  • ADMPCs isolated from adipocytes were subjected to FACS.
  • ADMPCs were separated from a culture dish with a 0.5 g/L-trypsin/0.53 mM-EDTA solution and suspended in a Dulbecco's Phosphate-buffered Saline (DPBS, Nacalai Tesque) containing 0.1% FBS.
  • DPBS Dulbecco's Phosphate-buffered Saline
  • FITC fluorescein isothiocyanate
  • CD105 Ancell
  • CD133 R&D
  • PE phycoerythrin
  • the incubated cells were incubated with mouse monoclonal antibodies against human SSEA-4, TRA-1-60, TRA-1-81 (Chemicon), ABCG-2, CD117 (BD PharMingen), or fibroblast/epithelial cell (AbD Serotec), as well as non-specific mouse antibody used as negative control, at 4° C. for 30 minutes.
  • the cells were incubated with PE-labeled goat anti-mouse Ig antibody (BD PharMingen), at 4° C. for 30 minutes. After washing three times, the cells were resuspended with DPBS, analysis was carried out by flow cytometry using FACSCalibur flow cytometer and CellQuest Pro software (BD Biosciences).
  • FIG. 27 and FIG. 28 The results are shown in FIG. 27 and FIG. 28 .
  • the expression did almost not vary in ADMPC for the markers of hematopoietic system cell and hematopoietic stem cell (CD45, ABCG-2, CD34, CD133) (Mitchell et al., Stem Cells, 24, 376-85 (2006)) and endothelial cell (CD31), surface antigen c-Kit (CD117), as well as surface markers for given ES cell and EG (embryonic germ) cell (TRA-1-60 and TRA-1-81) (James et al., 1998 and Shamblott et al., Proc. Natl. Acad. Sci. USA 95, 1372613731 (1998)).
  • ADMPC according to the present invention has high differentiation capability, and furthermore, the method of the present invention allows high purity ADMPC to be obtained.
  • ADMPCs were differentiated into pancreatic endocrine cells by the methods described in WO 2007/039986.
  • the obtained pancreatic endocrine cells are shown in FIG. 29 .
  • the ADMPCs were verified to be capable of differentiating into pancreatic endocrine cells, that is to say, to have functions as pancreatic progenitor cells.
  • the efficiency of differentiation from such ADMPCs into pancreatic endocrine cells was higher than that from ADSCs (data not shown).
  • DMSO 0.1%), HGF (10 ng/mL), bFGF (10 ng/mL), and oncostatin M (10 ng/mL), which are known to cause a hepatic progenitor cell to differentiate into a hepatoblast/hepatocyte were added to a Culture Medium I to prepare Culture Medium II.
  • ADMPCs were cultured for 14 days in Culture Medium II to obtain hepatocytes. The obtained hepatocytes are shown in FIG. 30 .
  • the ADMPCs were verified to be capable of differentiating into hepatocytes, that is to say, to have functions as hepatic progenitor cell.
  • the efficiency of differentiation from the ADMPCs into hepatocytes was higher than that from ADSCs (data not shown).
  • ADMPCs were cultured in Culture Medium I containing DMSO (Group 1) or OP9 culture supernatant (Group 2) for 14 days and the expression of genes in the obtained cells was checked by RT-PCR as described above. As control, ADMPCs cultured in Culture Medium I were used. The results are shown in FIG. 35 . It could be verified that ⁇ -CA and MLC, which are markers of cardiac myoblast, were expressed in the cells from Groups 1 and 2, that is to say, ADMPCs were differentiated/induced into cardiac myoblasts.
  • ADMPCs were cultured in the presence of DMSO for 1, 2, 3, 4, 5, 7, 10 and 14 days to check the differentiation into cardiac myoblasts.
  • ADMPC and cardiac myocyte were used. The results are shown in FIGS. 36 to 40 . It was found that culturing in the presence DMSO allowed cardiac myoblasts to be obtained.
  • the obtained cardiac myoblast were cultured in Culture Medium I containing DMSO in a thermosensitive culture dish (CellSeed Inc.) at 37° C. to form a cell population.
  • the cell population was detached by incubating at 20° C. or lower for 30 minutes to obtain a sheet containing cardiac myoblasts (refer to FIG. 41 ).
  • the obtained sheet was used in the following grafting experiment.
  • a myocardial infarction model rat was prepared by ligating the coronary artery of a nude rat. Then, an ADMPC-derived sheet containing cardiac myoblasts was grafted to the injured region. Cardiac function was evaluated at two weeks before grafting, before grafting, and two weeks, four weeks and 16 weeks after grafting by measuring the diameter at end-diastole (LVDd), the diameter at end-systole (LVDs), the left ventricular ejection fraction (% EF) and the left ventricular internal diameter shortening fraction (% FS) with echo. As a control, a sheet containing ADMPCs formed as described above was used. The results are shown in FIGS. 42 to 49 .
  • rats grafted with a sheet containing ADMPCs although wall motion was observed after 2 weeks, it was not observed after 10 weeks.
  • the LVDd started to extend and the EF decreased at week 8 and later with the sheet containing cardiac myoblast; in contrast, LVDd and EF were both maintained with the sheet containing ADMPC-derived cardiac myoblast (ADMPC-derived cardiac myoblast is obtained by treating ADMPC in 0.1% DMSO for 48 hours), showing an improvement in cardiac function.
  • rats were sacrificed and the hearts were extracted.
  • the extracted hearts were fixed with a 4% paraformaldehyde solution, and then substituted with ethanol at 70%.
  • the fixed hearts were cut into a width of a few millimeters and solidified with paraffin to prepare blocks.
  • the obtained paraffin blocks were thin-sectioned to 2 ⁇ m using a microtome, pasted onto a slide glass and dried.
  • the obtained thin sections were subjected to haematoxylin-eosin staining and immunohistological staining as follows.
  • Simple Stain Rat MAX-PG (Nichirei Bioscience Corp.) was added drop-wise and reacted at room temperature for 30 minutes.
  • Simple Stain AEC Solution (Nichirei Bioscience Corp.) was added drop-wise and colored while examining under microscope.
  • nuclear staining was carried out by staining with haematoxylin solution for 3 minutes.
  • non-water soluble mounting agent (Nichirei Bioscience Corp.) was added drop-wise, mounting was performed with a cover glass, and observation was carried out with a microscope and a fluorescence microscope.
  • the thickness of ⁇ -CA antibody-positive region was measured. The results are shown in FIGS. 51 to 61 . It could be verified that the site where the sheet containing cardiac myoblasts was grafted was HLA-ABC-positive, that is to say, human-derived, expressed ⁇ -CA and MHC, was differentiated into cardiac muscle, and that the expression of ⁇ -CA was also maintained in the remaining cardiac muscle. Consequently, it was found that the grafted cardiac myoblasts transdifferentiated into cardiac myocytes and that the remaining cardiac muscle was protected.
  • ADMPCs were cultured using the adipocyte differentiation agent PPAR-y agonist to be differentiated into adipocytes.
  • the obtained adipocytes were oil red O-stained to measure the lipid content.
  • ADSCs were used as control. The results are shown in FIGS. 62 and 63 .
  • ADMPCs were found to be capable of differentiating into adipocytes, that is to say, to have functions as adipose progenitor cells. In addition, the efficiency of such differentiation was high compared to ADSCs. From the above it was verified that ADMPCs were cells that have the capability of differentiating into multipotent cells.
  • ADMPCs were cultured for seven days in DMEM containing 10 nM dexamethasone, 50 mg/dl ascorbic acid 2-phosphate, 10 mM ⁇ -glycerophosphate (Sigma), and 10% FBS to induce differentiation into bone tissue.
  • the differentiation state was verified by alizarin red staining and alkaline phosphatase (ALPase) activity.
  • Alizarin red staining the obtained cells were washed three times and fixed with anhydrous ethanol. After fixation, the cells were stained with 1% alizarin red S in 0.1% NH 4 OH (pH 6.5) for 5 minutes and washed with H 2 O.
  • ALPase activity well known methods were followed (Bessey, O. A.
  • the reaction mixture was incubated at 37° C. for 30 minutes and the reaction was stopped by the addition of 0.25 volume of 1N NaOH.
  • the hydrolysis of pNP was monitored as the change in the value of the absorbance at 410 nm of the spectrophotometer.
  • p-nitrophenol was used as a standard.
  • One activity unit was defined as the amount that hydrolyzes 1 nmol of p-NP in 1 minute.
  • the ALPase activity per cell was calculated based on the DNA amount.
  • the DNA content was measured by an improvement of a generic method (Labarca, C. et al., Biochem. 102, 344-352 (1980)). After washing, the cells were homogenized at 0 to 4° C.
  • the ADMPCs according to the present invention showed strong positivity in alizarin red staining and AP activity, verifying that they were easily induced to differentiate into bone tissue. From the above, ADMPCs were shown to have the capabilities to differentiate into tissues cells from multiple lineages including from the pancreas, the liver, the heart, fat, bone and the like.
  • a method for obtaining a hepatic lobule cell population from an adipose-tissue-derived cell, a hepatic lobule cell population obtainable thereby, a method of screening for a substance that promotes or inhibits formation of hepatic lobule and for a substance that causes the activity of a hepatic lobule to increase or decrease, and a kit therefor, and the like can be obtained, all of which are useful, for example, in the fields of preventing and treating cirrhosis and hepatic cancer, and the fields of, for example, research into hepatic regeneration.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Biomedical Technology (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Zoology (AREA)
  • Genetics & Genomics (AREA)
  • Wood Science & Technology (AREA)
  • Biotechnology (AREA)
  • Developmental Biology & Embryology (AREA)
  • General Health & Medical Sciences (AREA)
  • General Engineering & Computer Science (AREA)
  • Biochemistry (AREA)
  • Rheumatology (AREA)
  • Cell Biology (AREA)
  • Microbiology (AREA)
  • Medicinal Chemistry (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Cardiology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Materials For Medical Uses (AREA)
  • Investigating Or Analysing Biological Materials (AREA)
US12/664,344 2007-06-14 2008-06-16 Method for production of hepatic-lobule-like cell cluster from adipose-tissue-derived cell Abandoned US20100183568A1 (en)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
JP2007-157388 2007-06-14
JP2007157388 2007-06-14
JP2007-157387 2007-06-14
JP2007157387 2007-06-14
PCT/JP2008/060980 WO2008153180A1 (ja) 2007-06-14 2008-06-16 脂肪組織由来細胞から肝小葉様細胞塊を得る方法

Publications (1)

Publication Number Publication Date
US20100183568A1 true US20100183568A1 (en) 2010-07-22

Family

ID=40129780

Family Applications (4)

Application Number Title Priority Date Filing Date
US12/664,343 Active 2029-07-20 US9845458B2 (en) 2007-06-14 2008-06-16 Multipotent progenitor cell derived from adipose tissue
US12/664,344 Abandoned US20100183568A1 (en) 2007-06-14 2008-06-16 Method for production of hepatic-lobule-like cell cluster from adipose-tissue-derived cell
US15/825,245 Abandoned US20180072994A1 (en) 2007-06-14 2017-11-29 Multipotent progenitor cell derived from adipose tissue
US15/827,778 Abandoned US20180100141A1 (en) 2007-06-14 2017-11-30 Multipotent progenitor cell derived from adipose tissue

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US12/664,343 Active 2029-07-20 US9845458B2 (en) 2007-06-14 2008-06-16 Multipotent progenitor cell derived from adipose tissue

Family Applications After (2)

Application Number Title Priority Date Filing Date
US15/825,245 Abandoned US20180072994A1 (en) 2007-06-14 2017-11-29 Multipotent progenitor cell derived from adipose tissue
US15/827,778 Abandoned US20180100141A1 (en) 2007-06-14 2017-11-30 Multipotent progenitor cell derived from adipose tissue

Country Status (4)

Country Link
US (4) US9845458B2 (ja)
EP (2) EP2166084A4 (ja)
JP (2) JPWO2008153179A1 (ja)
WO (2) WO2008153179A1 (ja)

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8784803B2 (en) 2009-08-28 2014-07-22 Osaka Air Machine Service, Ltd. Therapeutic agent for liver-related diseases
US8883210B1 (en) 2010-05-14 2014-11-11 Musculoskeletal Transplant Foundation Tissue-derived tissuegenic implants, and methods of fabricating and using same
US9352003B1 (en) 2010-05-14 2016-05-31 Musculoskeletal Transplant Foundation Tissue-derived tissuegenic implants, and methods of fabricating and using same
CN107580617A (zh) * 2015-02-13 2018-01-12 拉姆拉特有限公司 印模膏
US10092600B2 (en) 2013-07-30 2018-10-09 Musculoskeletal Transplant Foundation Method of preparing an adipose tissue derived matrix
US10130736B1 (en) 2010-05-14 2018-11-20 Musculoskeletal Transplant Foundation Tissue-derived tissuegenic implants, and methods of fabricating and using same
US10531957B2 (en) 2015-05-21 2020-01-14 Musculoskeletal Transplant Foundation Modified demineralized cortical bone fibers
US10912864B2 (en) 2015-07-24 2021-02-09 Musculoskeletal Transplant Foundation Acellular soft tissue-derived matrices and methods for preparing same
US11052175B2 (en) 2015-08-19 2021-07-06 Musculoskeletal Transplant Foundation Cartilage-derived implants and methods of making and using same

Families Citing this family (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP5804342B2 (ja) * 2010-09-11 2015-11-04 国立大学法人金沢大学 脂肪組織由来間質細胞群を含む肝炎治療剤
US9644181B2 (en) 2012-04-25 2017-05-09 Riken Cell preparation containing myocardium-committed cell
JP6209377B2 (ja) * 2012-07-11 2017-10-04 学校法人近畿大学 脂肪組織由来体性幹細胞の製造方法
WO2016013352A1 (ja) * 2014-07-23 2016-01-28 学校法人日本大学 脱分化脂肪細胞の製造方法
US20210187031A1 (en) * 2016-01-19 2021-06-24 Osaka University Transplant material for treatment of heart disease
JP7237301B2 (ja) 2016-12-16 2023-03-13 有限会社大阪空気機械サービス 組織治癒剤
JP6923137B2 (ja) 2018-06-15 2021-08-18 有限会社大阪空気機械サービス 組織治癒剤
CN110628713A (zh) * 2019-11-07 2019-12-31 安徽科门生物科技有限公司 一种脂肪源干细胞提取制备方法
JP2023516484A (ja) 2020-03-11 2023-04-19 ビット バイオ リミテッド 肝細胞作製方法

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060154235A1 (en) * 2005-01-07 2006-07-13 Takahiro Ochiya Human hepatocyte-like cells and uses thereof
US20070148766A1 (en) * 2003-11-04 2007-06-28 Biomaster, Inc. Method and system for preparing stem cells from fat tissue

Family Cites Families (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
IL145002A0 (en) 1999-03-10 2002-06-30 Univ Pittsburgh Adipose-derived stem cells and lattices
US20030082152A1 (en) 1999-03-10 2003-05-01 Hedrick Marc H. Adipose-derived stem cells and lattices
RU2351648C2 (ru) 2001-11-09 2009-04-10 Артесел Сайенсиз, Инк. Дифференцировка стромальных клеток, полученных из жировой ткани, в эндокринные клетки поджелудочной железы и их использование
JP2006254896A (ja) * 2005-01-07 2006-09-28 Effector Cell Institute Inc ヒト肝細胞様細胞およびその利用
CN101233227A (zh) 2005-02-03 2008-07-30 国立大学法人冈山大学 由胚胎干细胞向肝细胞的分化诱导方法和由该方法诱导的肝细胞
WO2006134951A1 (ja) * 2005-06-13 2006-12-21 Biomaster, Inc. 脂肪由来幹細胞の長期培養増殖法
JP5131833B2 (ja) * 2005-10-05 2013-01-30 晃文 松山 脂肪組織由来細胞から膵内分泌細胞を得る方法

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070148766A1 (en) * 2003-11-04 2007-06-28 Biomaster, Inc. Method and system for preparing stem cells from fat tissue
US20060154235A1 (en) * 2005-01-07 2006-07-13 Takahiro Ochiya Human hepatocyte-like cells and uses thereof

Cited By (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8784803B2 (en) 2009-08-28 2014-07-22 Osaka Air Machine Service, Ltd. Therapeutic agent for liver-related diseases
US8883210B1 (en) 2010-05-14 2014-11-11 Musculoskeletal Transplant Foundation Tissue-derived tissuegenic implants, and methods of fabricating and using same
US9352003B1 (en) 2010-05-14 2016-05-31 Musculoskeletal Transplant Foundation Tissue-derived tissuegenic implants, and methods of fabricating and using same
US10130736B1 (en) 2010-05-14 2018-11-20 Musculoskeletal Transplant Foundation Tissue-derived tissuegenic implants, and methods of fabricating and using same
US11305035B2 (en) 2010-05-14 2022-04-19 Musculoskeletal Transplant Foundatiaon Tissue-derived tissuegenic implants, and methods of fabricating and using same
US10092600B2 (en) 2013-07-30 2018-10-09 Musculoskeletal Transplant Foundation Method of preparing an adipose tissue derived matrix
US10596201B2 (en) 2013-07-30 2020-03-24 Musculoskeletal Transplant Foundation Delipidated, decellularized adipose tissue matrix
US11779610B2 (en) 2013-07-30 2023-10-10 Musculoskeletal Transplant Foundation Acellular soft tissue-derived matrices and methods for using same
US11191788B2 (en) 2013-07-30 2021-12-07 Musculoskeletal Transplant Foundation Acellular soft tissue-derived matrices and methods for preparing same
CN107580617A (zh) * 2015-02-13 2018-01-12 拉姆拉特有限公司 印模膏
US11596517B2 (en) 2015-05-21 2023-03-07 Musculoskeletal Transplant Foundation Modified demineralized cortical bone fibers
US10531957B2 (en) 2015-05-21 2020-01-14 Musculoskeletal Transplant Foundation Modified demineralized cortical bone fibers
US10912864B2 (en) 2015-07-24 2021-02-09 Musculoskeletal Transplant Foundation Acellular soft tissue-derived matrices and methods for preparing same
US11524093B2 (en) 2015-07-24 2022-12-13 Musculoskeletal Transplant Foundation Acellular soft tissue-derived matrices and methods for preparing same
US11052175B2 (en) 2015-08-19 2021-07-06 Musculoskeletal Transplant Foundation Cartilage-derived implants and methods of making and using same
US11806443B2 (en) 2015-08-19 2023-11-07 Musculoskeletal Transplant Foundation Cartilage-derived implants and methods of making and using same
US11938245B2 (en) 2015-08-19 2024-03-26 Musculoskeletal Transplant Foundation Cartilage-derived implants and methods of making and using same

Also Published As

Publication number Publication date
JPWO2008153180A1 (ja) 2010-08-26
EP2166084A4 (en) 2010-06-23
EP2172541A4 (en) 2010-06-30
WO2008153179A1 (ja) 2008-12-18
JPWO2008153179A1 (ja) 2010-08-26
US9845458B2 (en) 2017-12-19
WO2008153180A1 (ja) 2008-12-18
EP2166084A1 (en) 2010-03-24
US20100151574A1 (en) 2010-06-17
EP2172541A1 (en) 2010-04-07
US20180072994A1 (en) 2018-03-15
US20180100141A1 (en) 2018-04-12

Similar Documents

Publication Publication Date Title
US20180072994A1 (en) Multipotent progenitor cell derived from adipose tissue
JP5791111B2 (ja) 馴化培地、および馴化培地を作る方法
US20110171726A1 (en) Multipotent stem cells derived from human adipose tissue and cellular therapeutic agents comprising the same
US20070264239A1 (en) Isolation of pericytes
CA2921948C (en) Method for preparing pluripotent stem cells
JP6555691B2 (ja) 間葉系幹細胞の賦活化剤、賦活化された間葉系幹細胞およびその製造方法
CN107828722B (zh) 特异性表达pd-1的干细胞、其鉴定和分离方法及用途
KR20080081088A (ko) 태반 줄기세포와 제2 세포원으로부터 얻은 줄기세포의 공동배양
JP7286651B2 (ja) 間葉系間質細胞及び臍帯から間葉系間質細胞を得るための方法
JP2011519574A5 (ja)
JP5388297B2 (ja) アディポクラスター
Nakatsuka et al. Identification and characterization of lineage− CD45− Sca-1+ VSEL phenotypic cells residing in adult mouse bone tissue
Sharifiaghdas et al. Isolation of human adult stem cells from muscle biopsy for future treatment of urinary incontinence
WO2020066991A1 (ja) アカルボース又はスタキオースを含む哺乳動物細胞保存用液
KR20120006386A (ko) 1기 태반조직 유래 줄기세포 및 이를 함유하는 세포치료제
KR101834800B1 (ko) 심장 조직-유래 세포
JP5230969B2 (ja) 心筋芽細胞を含むシート
US11920180B2 (en) Method for inducing differentiation of pluripotent stem cells in vitro
US9687512B2 (en) Isolated cardiac stem cells and methods of their use
EP3680324A1 (en) Stem cells derived from young pig and preparation method therefor
Chen et al. Heterogeneity of stem cells in human amniotic fluid
RU2663118C1 (ru) Клеточный продукт инсулин-продуцирующих клеток млекопитающих и его использование для терапии сахарного диабета
RU2793467C2 (ru) Мезенхимальные стромальные клетки и способы получения мезенхимальных стромальных клеток из пуповины
Truong et al. Phenotypic and cytogenetic characterization of expanded adipose derived stem cells
JP2022513475A (ja) Hla-eを発現する肝臓前駆細胞を含む細胞組成物

Legal Events

Date Code Title Description
AS Assignment

Owner name: FOUNDATION OF BIOMEDICAL RESEARCH AND INNOVATION,

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:MATSUYAMA, AKIFUMI;KOMODA, HIROSHI;SAWA, YOSHIKI;SIGNING DATES FROM 20091210 TO 20091217;REEL/FRAME:023711/0874

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION