WO2003047607A1 - Medicinal compositions containing human amnion-origin cells - Google Patents

Medicinal compositions containing human amnion-origin cells Download PDF

Info

Publication number
WO2003047607A1
WO2003047607A1 PCT/JP2002/012761 JP0212761W WO03047607A1 WO 2003047607 A1 WO2003047607 A1 WO 2003047607A1 JP 0212761 W JP0212761 W JP 0212761W WO 03047607 A1 WO03047607 A1 WO 03047607A1
Authority
WO
WIPO (PCT)
Prior art keywords
cells
amniotic membrane
human
mesenchymal cells
active ingredient
Prior art date
Application number
PCT/JP2002/012761
Other languages
French (fr)
Japanese (ja)
Inventor
Toshio Nikaido
Original Assignee
Sankyo Company, Limited
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
Priority claimed from JP2002262265A external-priority patent/JP2005139067A/en
Application filed by Sankyo Company, Limited filed Critical Sankyo Company, Limited
Priority to AU2002354101A priority Critical patent/AU2002354101A1/en
Publication of WO2003047607A1 publication Critical patent/WO2003047607A1/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
    • A61K35/48Reproductive organs
    • A61K35/50Placenta; Placental stem cells; Amniotic fluid; Amnion; Amniotic stem cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • 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 therapeutic agent for diabetes comprising human amniotic membrane-derived epithelial cells as an active ingredient and a method for treating diabetes comprising administering human amniotic membrane-derived epithelial cells to a diabetic patient.
  • the present invention relates to a method for treating bone metabolism disorders, which comprises administering a therapeutic agent for bone metabolism disorder as an active ingredient and human amniotic membrane-derived mesenchymal cells to a patient with bone metabolism disorders.
  • the present invention relates to a method for treating a heart disease, which comprises administering a therapeutic agent for a heart disease and a human amniotic membrane-derived mesenchymal cell to a patient with a heart disease.
  • the present inventors have been searching for cells that can be substituted for stem cells derived from human fetuses.However, epithelial cells or mesenchymal cells derived from human amniotic membrane were administered to an organism under certain conditions. Then, they found that the pathology of living organisms could be improved, and completed the present invention.
  • the present invention relates to a pharmaceutical composition containing human amniotic membrane-derived epithelial cells as an active ingredient.
  • the present invention relates to a therapeutic agent for diabetes containing human amniotic membrane-derived epithelial cells as an active ingredient.
  • the present invention relates to a method for treating diabetes mellitus, which comprises administering human amniotic membrane-derived epithelial cells to a diabetic patient.
  • the present invention relates to a pharmaceutical composition containing human amniotic membrane-derived mesenchymal cells as an active ingredient.
  • the present invention relates to a therapeutic agent for bone metabolism disorder containing human amniotic membrane-derived mesenchymal cells as an active ingredient.
  • the present invention relates to a method for treating bone metabolism disorder, which comprises administering human amniotic membrane-derived mesenchymal cells to a patient with bone metabolism disorder.
  • the present invention relates to a therapeutic agent for heart disease containing human amniotic membrane-derived mesenchymal cells as an active ingredient.
  • the present invention relates to a method for treating heart disease, which comprises administering human amniotic membrane-derived mesenchymal cells to a heart disease patient.
  • human amniotic cells can be obtained by cesarean section of a human pregnant woman who has obtained informal consent and by a usual surgical technique.
  • Epithelial cells can be obtained from amniotic membrane by the following method. The obtained amniotic membrane is fragmented using sterilized scissors or the like, and is placed in a culture medium such as Dulbecco's modified Figur medium used for culturing normal animal cells, and shaken.
  • a protease such as trypsin is used. It is desirable that elements coexist. Suitable concentrations of proteolytic enzymes are between 0.1% and 0.5%.
  • the temperature for shaking is preferably between 25 and 40 ° C.
  • the shaking interval is preferably from 50 rpm to 200 rpm, and the shaking time is preferably from 20 minutes to 60 minutes. After collecting the cells by centrifugation and removing the supernatant, the cells are shaken again in the culture medium containing the protease under the same conditions as described above. However, the shaking interval is preferably from 400 rpm to 600 rpm, and the shaking time is preferably from 20 minutes to 60 minutes.
  • the culture solution is passed through a mesh such as sterile gauze, and the epithelial cells are separated.
  • Epithelial cells were collected by centrifugation, in a culture medium containing 10% serum, C0 2 incubator base - cultured in the evening.
  • Culture temperature is 370C
  • C0 2 concentration is preferably 5%.
  • the serum is preferably calf serum or fetal calf serum.
  • an antibiotic having a concentration of about 1% (for example, penicillin G, 100 units Zml: streptomycin, 100 g Zml: amphotericin B, 0.25 g / ml) is co-present in the culture solution.
  • the amniotic membrane remaining on the gauze can be re-fragmented and further dispersed in cells in the same manner as described above.
  • Epithelial cells obtained are collected by centrifugation, as above, in a culture medium containing 10% serum, cultured in C0 2 incubator.
  • the cell concentration of the culture is preferably 1 ⁇ 10 5 to 1 ⁇ 10 6 Zml.
  • 5 XK 7 epithelial cells can be isolated from one amniotic membrane.
  • Amniotic mesenchymal cells can be isolated from amniotic membrane from which epithelial cells have been completely removed.
  • the amniotic membrane remaining on the mesh such as gauze is treated with a protease such as 0.1-1.0 mgZml of collagenase (manufactured by Wako Pure Chemical Industries, Ltd.) and a nuclease such as 0.01-0.1 mgZml of DNAase (Sigma). Shake and digest.
  • the shaking temperature is preferably 20 to 40 ° C, and the shaking time is preferably 30 to 120 minutes. Preferably, the shaking interval is 100 to: LOOO rpm.
  • the digested amniotic membrane can be passed through a mesh such as gauze to separate mesenchymal cells.
  • the pharmaceutical composition of the present invention is characterized by containing human amniotic epithelial cells or mesenchymal cells obtained by the above-mentioned method.
  • the administration site is not particularly limited. Depending on the disease, a site such as a spleen, a spleen, or a site of a rheumatic disease or a variety of blood vessels is selected. The total number of cells to be administered depends on the patient's condition, weight, and the like.
  • the diseases that can be treated by the pharmaceutical composition or the therapeutic method of the present invention are as follows. Hyperlipidemia, hyperglycemia, impaired glucose tolerance (i-immediate ai red glucose tolerance: IGT), diabetic complications (eg retinopathy, nephropathy, cataract, coronary artery disease, etc.), arteriosclerosis, heart Vascular disease (eg, ischemic heart disease, etc.).
  • IGT impaired glucose tolerance
  • diabetic complications eg retinopathy, nephropathy, cataract, coronary artery disease, etc.
  • arteriosclerosis eg, ischemic heart disease, etc.
  • Osteoporosis due to di syndrome and limb fixation. Bone Paget's disease in adolescents and boys and girls (osteoarthritis).
  • Hypercalcemia and hematological malignancies solid myeloma, lymphoma and leukemia
  • solid tumors solid tumors (breast, lung and kidney)
  • idiopathic hypercalcemia and hyperthyroidism and renal function
  • Hypercalcemia associated with insufficiency Post-surgical steroid-induced osteopenia associated with small and large bowel disorders, and chronic hepatitis and kidney disease.
  • Ischemic heart disease eg, angina, myocardial infarction, Kawasaki disease, etc.
  • valvular disease eg, monk
  • Mitral stenosis mitral regurgitation, mitral valve prolapse syndrome, aortic stenosis, aortic regurgitation, etc.
  • arrhythmia eg, atrial fibrillation, atrial flutter, paroxysmal supraventricular tachycardia
  • WPW syndrome atrioventricular nodal recurrent tachycardia, ventricular extrasystole, ventricular tachycardia, ventricular fibrillation, bradyarrhythmia, sinus dysfunction syndrome, etc.
  • aortic and vascular diseases eg, ascending aortic aneurysm, arch Bilateral aor
  • FIG. 1 shows an amniotic membrane-derived epithelial cell and mesenchymal cell diagram.
  • FIG. 2 shows a blood glucose level fluctuation diagram in mice administered with amniotic epithelial cells and mesenchymal cells.
  • M represents the change in blood glucose level in mice administered with mesenchymal cells
  • HAE represents changes in blood glucose levels in mice administered with epithelial cells.
  • FIG. 3 shows immunohistochemical staining showing the expression of human ⁇ 2-microglobulin and human insulin in epithelial cell transplanted spleen.
  • Figure 4 shows the expression of human collagen type II gene in cultured amnion-derived mesenchymal cells.
  • the left figure is a phase contrast micrograph, in which cells that have undergone morphological changes were observed.
  • the right figure is an immunostaining diagram using an anti-human collagen type II antibody. Expression of human collagen type II was observed in cells that had undergone morphological changes.
  • Figure 5 shows that the isolated mesenchymal cells were cultured for about one month in the presence of 100 ng / ml FGF or 5 x M 5-azacitidine, and immunofluorescent staining was performed using a myocardial-specific antibody against NKX2.5.
  • FIG. 3 is a diagram in which RT was confirmed by the RT-PCR method.
  • Example 1 Isolation of human epithelial cells and mesenchymal cells
  • a pregnant woman with informed consent was subjected to cesarean section to obtain an amniotic membrane.
  • the resulting amniotic fragmented with surgical scissors sterile, cell culture medium containing 0.2% Toribushin (Dulbecco's modified Eagle's medium:. Hereinafter referred to as "DMEM") with placed in 50ml Falcon tubes, shaker of 37 Q C Shake at 100 rpm for 30 minutes. Removing the supernatant after centrifugation, fragmented with surgical scissors were sterilized again, shaking at 400-600rpm at 37 Q C Chez one force one 30 minutes, and digested with 0.2% trypsin. The solution was passed through sterile gauze to separate epithelial cells.
  • Epithelial cells are collected by centrifugation, and the culture medium is 370C in DMEM containing 10% fetal calf serum and antibiotics (benicillin G, 100 units Zml: streptomycin, 100 g / ml: amphotericin B, 0.25 g / ml), 5% C0 2 and cultured under.
  • antibiotics antibiotics (benicillin G, 100 units Zml: streptomycin, 100 g / ml: amphotericin B, 0.25 g / ml), 5% C0 2 and cultured under.
  • the amniotic membrane remaining on the gauze was fragmented with re-sterilized surgical scissors and similarly digested with 0.2% trypsin for 30 minutes.
  • epithelial cells were collected by centrifugation at a cell density of 3 ⁇ 10 5 cells / ml, 370C in DMEM containing 10% fetal calf serum and the same antibiotics as above, 5% C0 2 were cultured under.
  • epithelial cells usually 5 ⁇ 10 7 cells can be separated from one amniotic membrane. Amniotic mesenchymal cells were isolated from amniotic membrane from which epithelial cells had been completely removed.
  • the amniotic membrane left on the gauze was digested with collagenase (0.75 mgZml, manufactured by Wako Pure Chemical Industries, Ltd.) and DNAse (0.075 mgZml, manufactured by Sigma) at 37 ° C. for 60 minutes at 600 rpm for digestion.
  • the digested amniotic membrane was passed through gauze to separate mesenchymal cells.
  • FIG. 1 shows photographs of the separated amniotic membrane-derived epithelial cells and mesenchymal cells.
  • Test Example 1 Therapeutic effect of diabetes by human amniotic membrane-derived epithelial cells
  • mice Eight to 12 weeks of immunodeficient SCID (sever combined immunodeficient) mice can be treated with human amnion-derived epithelial cells (hereinafter simply referred to as “epithelial cells”) or human amniotic-derived mesenchymal cells (hereinafter simply referred to as “mesenchymal cells”). ).
  • epithelial cells human amnion-derived epithelial cells
  • mesenchymal cells human amniotic-derived mesenchymal cells
  • mice One week after STZ administration, under anesthesia, 1 ⁇ 10 6 cells / 0.1 ml of epithelial cells or mesenchymal cells were transplanted into the mouse spleen.
  • the blood glucose level of the transplanted mice was measured every 9 days between 9 and 11 am using the Aventis Pharma Co., Ltd. diasensor according to the experimental protocol of serum collected from the tail.
  • mice spleen and tentum obtained in (1) were cut into 10% formalin-fixed paraffin slices, sliced to 4 to 6 microns, attached to a slide glass, and used.
  • the following mouse-derived monoclonal antibodies were used as primary antibodies for immunostaining or fluorescence immunization.
  • Anti-human insulin antibody manufactured by Oncogene: Ab-lZl: 400 dilution
  • anti-human ⁇ 2-microglobulin antibody manufactured by Pharmaluminogen: Col ⁇ ZlOO dilution
  • anti-human collagen type II antibody manufactured by Sigma
  • a heron anti-mouse antibody was used as a secondary antibody, and a streptavidin-biotin method (SAB-PO kit: manufactured by Nichirei Co., Ltd.) was used as an antigen detection system. However, only for the detection of human collagen type II, FITC labeling as a secondary antibody ⁇ A heron anti-mouse antibody was used. Heat treatment in a microwave oven for 15 minutes in 0.01M-pH6.0 citrate buffer, deparaffinization, and endogenous peroxidase inhibition (rest in methanol with 0.3% hydrogen peroxide for 30 minutes) .
  • the primary antibody binding reaction is performed at room temperature for 1 hour, the biotin-labeled secondary antibody binding reaction is performed at room temperature for 30 minutes, and the peroxidase-labeled streptavidin (enzyme reagent) is allowed to bind at room temperature for 30 minutes, followed by DAB Aminobenzidine) The color was developed.
  • DNA was extracted from the mouse spleen and spleen obtained in (2) using DNAzol from Molecular Research Senyuichi according to the company's experimental method.
  • a primer designed based on the human-specific nucleotide sequence in the human ⁇ 2-microglobulin gene was obtained from the mouse spleen and spleen obtained in (2) using DNAzol from Molecular Research Senyuichi according to the company's experimental method.
  • the isolated mesenchymal cells were cultured in vitro in the presence of 100 ng / ml Bone Moropogenie Protein (BMP) for about 1 month with high molecular lactate-polyethylene glycol (molecular weight: about 9500) and 10% fetal calf serum. in containing DMEM 370C, 5% C0 2 with antibiotics (Bae two cylindrical G, 100 units ZML: streptomycin, 100 ⁇ g Zml: Anfoteri Singh B, 0.25 g / ml) after co-cultured in the presence of, cartilage cell-specific Analysis was performed by immunofluorescent staining with a collagen type II antibody as described below.
  • BMP Bone Moropogenie Protein
  • the above cultured mesenchymal cells were washed with phosphate buffered saline (PBS), and fixed by treatment with 100% methanol at ⁇ 20 for 10 minutes and 100% acetone at ⁇ 20 ° C. for 1 minute. Then react with an anti-human collagen type II antibody (Sigma: Tu-99 / l: diluted 1000) for 1 hour at room temperature, wash with PBS, and use FITC-labeled heron anti-mouse antibody as a secondary antibody at room temperature for 30 hours. After reacting for 1 minute, the cells were washed with PBS and observed with a fluorescence microscope. As a result, the expression of the human collagen type II gene was specifically observed only in the cells whose morphology had changed (Fig. 4). The results showed that chondrocytes could be induced from human amniotic mesenchymal cells. Test Example 3. Differentiation of human amniotic membrane-derived mesenchymal cells into cardiomyocytes
  • FIG. 5 The results showed that cardiomyocytes could be induced from human amniotic mesenchymal cells.
  • RNAzol Molecular Research Center

Abstract

It is intended to provide medicinal compositions containing, as the active ingredient, epithelial cells or mesenchymal cells originating in human amnion which are usable as a substitute for human fetal stem cells because of having regeneration ability and can be relatively easily obtained by using usual surgical techniques from maternal humans giving informed consent. More specifically, medicinal compositions such as remedies for diabetes which contain the above epithelial cells as the active ingredient, or medicinal compositions such as remedies for bone metabolic errors or heart diseases which contain human amnion-origin mesenchymal cells as the active ingredient. Thus, it becomes possible to provide medicinal compositions such as remedies for diabetes by using regeneration therapy without causing any ethical problems.

Description

明細書  Specification
ヒト羊膜由来細胞含有医薬組成物  Human amniotic membrane-derived cell-containing pharmaceutical composition
[技術分野] [Technical field]
本発明は、 ヒト羊膜由来上皮細胞を有効成分とする糖尿病治療剤及びヒト羊 膜由来上皮細胞を糖尿病患者に投与することを特徴とする糖尿病治療法に関し、 また、 ヒト羊膜由来間葉系細胞を有効成分とする骨代謝異常症治療剤及びヒト 羊膜由来間葉系細胞を骨代謝異常症患者に投与することを特徴とする骨代謝異 常症治療法に関し、 さらに、 ヒト羊膜由来間葉系細胞を有効成分とする心臓疾 患治療剤及びヒト羊膜由来間葉系細胞を心臓疾患患者に投与することを特徴と する心臓疾患治療法に関する。  The present invention relates to a therapeutic agent for diabetes comprising human amniotic membrane-derived epithelial cells as an active ingredient and a method for treating diabetes comprising administering human amniotic membrane-derived epithelial cells to a diabetic patient. The present invention relates to a method for treating bone metabolism disorders, which comprises administering a therapeutic agent for bone metabolism disorder as an active ingredient and human amniotic membrane-derived mesenchymal cells to a patient with bone metabolism disorders. The present invention relates to a method for treating a heart disease, which comprises administering a therapeutic agent for a heart disease and a human amniotic membrane-derived mesenchymal cell to a patient with a heart disease.
[背景技術] [Background technology]
近年、 再生医療なる技術が脚光を浴びるようになった。 すなわち、 ヒト又は 他の生物の体は、 一生の間に外傷や疾病によって組織の一部を失ったり、 大き な障害を受けたりするが、 この場合の再生能力は、 動物種によっても、 また、 組織によっても異なっている。 自然には再生できない臓器や組織を再生させ、 機能を回復させるベく、 未分化の細胞より生物機能を有する細胞を分化せしめ る技術が発達した。 このような技術は、 一般に再生医療と呼ばれている。 しかしながら、 従来、 このような技術に供される細胞として、 一般にヒト胎 児由来の幹細胞が使用されており、 倫理的な側面より問題視されていた。  In recent years, the technology of regenerative medicine has come into the spotlight. In other words, the body of humans or other organisms loses part of their tissues or suffers severe damage due to trauma or disease during their lives. It varies from organization to organization. In order to regenerate organs and tissues that cannot be naturally regenerated and restore their functions, techniques have been developed to differentiate cells with biological functions from undifferentiated cells. Such a technique is generally called regenerative medicine. However, hitherto, stem cells derived from a human fetus have generally been used as cells subjected to such a technique, which has been regarded as a problem from an ethical aspect.
[発明の開示] [Disclosure of the Invention]
本発明者は、 かかる問題を克服すべく、 ヒト胎児由来の幹細胞に代替し得る 細胞を模索していたところ、 ヒト羊膜由来の上皮細胞又は間葉系細胞を一定条 件下で生物体内に投与すれば、 生物体の病態を改善せしめることを見出し、 本 発明を完成した。 本発明は第一に、 ヒト羊膜由来上皮細胞を有効成分とする医薬組成物に関す る。 第二に、 ヒト羊膜由来上皮細胞を有効成分とする糖尿病治療剤に関する。 第三に、 ヒト羊膜由来上皮細胞を糖尿病患者に投与することを特徴とする糖 尿病治療法に関する。 第四に、 ヒ卜羊膜由来間葉系細胞を有効成分とする医薬組成物に関する。 第五に、 ヒト羊膜由来間葉系細胞を有効成分とする骨代謝異常症治療剤に関 する。 第六に、 ヒト羊膜由来間葉系細胞を骨代謝異常症患者に投与することを特徴 とする骨代謝異常症治療法に関する。 第七に、 ヒト羊膜由来間葉系細胞を有効成分とする心疾患治療剤に関する。 第八に、 ヒト羊膜由来間葉系細胞を心疾患患者に投与することを特徴とする 心疾患治療法に関する。 本発明において、 ヒト羊膜細胞は、 インフォ一ムドコンセントを得たヒト妊 婦を帝王切開し、 通常の外科的手法で得ることができる。 羊膜より上皮細胞は以下の方法で得ることができる。 得られた羊膜を、 滅菌したハサミ等を用い断片化し、 ダルベッコ修正ィーグ ル培地等の通常の動物細胞の培養に用いられる培養液に入れ、 振盪する。 この 場合、 羊膜断片を個々の細胞に分散化するため、 トリプシン等の蛋白質分解酵 素を共存させることが望ましい。 好適な蛋白質分解酵素の濃度は、 0.1 %から 0.5 %である。 また、 振盪する場合の温度は、 25でから 40°Cの間が望ましい。 振盪の間隔は、 50rpm〜200rpmが望ましく、 振盪時間は、 20分から 60分が 望ましい。 遠心操作で細胞を集めた後、上清を除去後、該細胞を前記と同様の条件下で、 蛋白質分解酵素が共存する培養液中で再度振盪する。 但し、 振盪の間隔は、 400rpm〜600rpmが望ましく、 振盪時間は、 20分から 60分が望ましい。 この後、培養溶液を滅菌ガーゼなどのメッシュで越し、上皮細胞を分離する。 上皮細胞は遠心によって集め、 10%の血清を含む培養液中で、 C02インキュべ —夕中で培養する。 培養温度は 370C、 C02 濃度は 5%が好ましい。 血清は子牛 血清又は子牛胎児血清が好ましい。 また、 培養液には、 濃度 1 %前後の抗生物 質 (例えば、 ペニシリン G、 100単位 Zml:ストレプトマイシン、 lOO g Z ml:アンフォテリシン B、 0.25 g /ml) を共存させることが好ましい。 ガー ゼに残された羊膜は、 上記と同様の方法で、 再度断片化、 さらに、 細胞分散化 することができる。 得られた上皮細胞は、 遠心で集め、 上記と同様に、 10%の血清を含む培養液 中で、 C02インキュベータ中で培養する。 培養の細胞濃度は、 1 X 105 〜1 X 106 Zmlが好ましい。 通常羊膜一枚から 5 X K)7個の上皮細胞が分離できる。 羊膜間葉系の細胞は、 上皮細胞が完全に取り除かれた羊膜より、 分離するこ とができる。 ガーゼなどのメッシュ上に残された羊膜を 0.1〜1.0mgZmlのコ ラゲナーゼ (和光純薬 (株) 製) 等の蛋白質分解酵素及 0.01〜0.1mgZml の DNAァーゼ (シグマ社) 等の核酸分解酵素により振盪し、 消化することができ る。 振盪の温度は、 20〜40Τ:が好ましく、 振盪時間は 30分間〜 120分間が好ま しく、 振盪の間隔は、 100〜: LOOOrpmが好ましい。 消化された羊膜をガーゼなどのメッシュで越し、 間葉系細胞を分離すること ができる。 得られた間葉系細胞を遠心で集め、 10%の血清を含む培養液中で、 C02インキュベータ中で培養する。 培養温度は 37QC、 C02 濃度は 5%が好まし い。 血清は子牛血清又は子牛胎児血清が好ましい。 また、 培養液には、 濃度 1 % 前後の抗生物質 (前記と同様) を共存させることが好ましい。 細胞密度は、 10 5 〜: 106 細胞 Zmlが好ましい。 間葉系細胞は、 通常羊膜一枚から 5 X 106個細 胞が分離できる。 本発明の医薬組成物は、 上述の方法で得られた、 ヒト羊膜由来の上皮細胞又 は間葉系細胞を含むことを特徴とする。 上皮細胞又は間葉系細胞を患者に投与する場合、 共存する抗生物質や異種血 清を完全に除去するために、 生理食塩水で細胞を丹念に洗^する。 細胞を投与 する場合には、 細胞を生理食塩水又は培養液にサスペンジョンしたものを用い る。 細胞濃度は、 106〜: L08 細胞/ mlが好ましい。 細胞を補助するために、 増 殖因子や細胞安定剤を混在させてもよい。 投与部位は、 特に限定はないが、 疾患に応じて、 塍臓、 脾臓、 リウマチ疾患 部位等の局所や種々の血管内が選択される。 投与する総細胞数は、 患者の病態、 体重等に依存する。 本発明の医薬組成物又は治療法により治療可能な疾患は、以下の通りである。 高脂血症、 高血糖症、 耐糖能不全 (i即 a i red gl ucose t o l erance: IGT) 状 態、糖尿病合併症(例えば網膜症、 腎症、 白内障、冠動脈疾患等)、動脈硬化症、 心血管性疾患 (例えば虚血性心疾患等)。 一次骨粗鬆症、 内分泌骨粗鬆症 (甲状腺機能亢進症、 副甲状腺機能亢進症、 クッシング症候群、 及び末端肥大症)、 遺伝性及び先天性形態の骨粗鬆症 (骨形 成不完全、 ホモシスチン尿症、 メンケス症候群、 及びライリーディ症候群) 及 び四肢の固定による骨粗鬆症のような骨粗鬆症。 青年及び少年少女における骨のパジエツト病 (変形性骨炎)。 固形腫瘍 (乳房、 肺及び腎臓) に由来する高カルシウム血症及び血液学的悪 性疾患 (多発性骨髄腫、 リンパ種及び白血病)、 特発性高カルシウム血症、 及び 甲状腺機能亢進症及び腎機能不全に関連する高カルシウム血症。 外科手術後の、 ステロイド投与に誘発された、 及び小腸及び大腸の障害に関 連した、 及び慢性肝炎及び腎臓病に関連した骨減少症。 外傷性負傷に関連した、 またはゴジェ病、 鎌形赤血球性貧血、 全身性紅斑性 狼瘡及び他の疾患に関連した非外傷性壊死に関連した、 骨壊死、 または細胞死 滅。 慢性間接リウマチによる骨減少。 歯周骨喪失。 骨溶解性転移。 . 変形性関節症、 慢性間接リウマチ又は変形性脊椎症及びこれらに由来する間 接の疼痛や類肩腕痛、 腰痛及び麻痺。 虚血性心疾患 (例えば、 狭心症、 心筋梗塞、 川崎病等)、 弁膜症 (例えば、 僧 帽弁狭窄症、 僧帽弁閉鎖不全症、 僧帽弁逸脱症候群、 大動脈弁狭窄症、 大動脈 弁閉鎖不全症等)、 不整脈 (例えば、 心房細動、 心房粗動、 発作性上室性頻拍、 W P W症候群、 房室結節回帰性頻拍、 心室性期外収縮、 心室頻拍、 心室細動、 徐脈性不整脈、洞不全症候群等)、大動脈 ·血管疾患(例えば、上行部大動脈瘤、 弓部大動脈瘤、 胸部大動脈瘤、 胸部下行大動脈瘤、 胸腹部大動脈瘤、 腹部大動 脈瘤、 閉塞性動脈硬化症、静脈血栓、肺血栓閉塞症等)、先天性心疾患(例えば、 心房中隔欠損症、 心室中隔欠損症、 E i senmenger 症候群、 肺動脈還流異常、 動 脈管開存症、 心内膜床欠損症、 完全大血管転移症、 修正大血管転移症、 肺動脈 狭窄症、 Fa l l o t四徴症、 大動脈縮窄症、 Val salva洞動脈瘤、 Ebs te i n奇形等)、 心筋症 (拡張型心筋症、 肥大型心筋症、 拘束型心筋症等)。 In order to overcome such a problem, the present inventors have been searching for cells that can be substituted for stem cells derived from human fetuses.However, epithelial cells or mesenchymal cells derived from human amniotic membrane were administered to an organism under certain conditions. Then, they found that the pathology of living organisms could be improved, and completed the present invention. First, the present invention relates to a pharmaceutical composition containing human amniotic membrane-derived epithelial cells as an active ingredient. Second, the present invention relates to a therapeutic agent for diabetes containing human amniotic membrane-derived epithelial cells as an active ingredient. Third, the present invention relates to a method for treating diabetes mellitus, which comprises administering human amniotic membrane-derived epithelial cells to a diabetic patient. Fourth, the present invention relates to a pharmaceutical composition containing human amniotic membrane-derived mesenchymal cells as an active ingredient. Fifthly, the present invention relates to a therapeutic agent for bone metabolism disorder containing human amniotic membrane-derived mesenchymal cells as an active ingredient. Sixth, the present invention relates to a method for treating bone metabolism disorder, which comprises administering human amniotic membrane-derived mesenchymal cells to a patient with bone metabolism disorder. Seventh, the present invention relates to a therapeutic agent for heart disease containing human amniotic membrane-derived mesenchymal cells as an active ingredient. Eighth, the present invention relates to a method for treating heart disease, which comprises administering human amniotic membrane-derived mesenchymal cells to a heart disease patient. In the present invention, human amniotic cells can be obtained by cesarean section of a human pregnant woman who has obtained informal consent and by a usual surgical technique. Epithelial cells can be obtained from amniotic membrane by the following method. The obtained amniotic membrane is fragmented using sterilized scissors or the like, and is placed in a culture medium such as Dulbecco's modified Figur medium used for culturing normal animal cells, and shaken. In this case, to disperse the amniotic membrane fragments into individual cells, a protease such as trypsin is used. It is desirable that elements coexist. Suitable concentrations of proteolytic enzymes are between 0.1% and 0.5%. The temperature for shaking is preferably between 25 and 40 ° C. The shaking interval is preferably from 50 rpm to 200 rpm, and the shaking time is preferably from 20 minutes to 60 minutes. After collecting the cells by centrifugation and removing the supernatant, the cells are shaken again in the culture medium containing the protease under the same conditions as described above. However, the shaking interval is preferably from 400 rpm to 600 rpm, and the shaking time is preferably from 20 minutes to 60 minutes. Thereafter, the culture solution is passed through a mesh such as sterile gauze, and the epithelial cells are separated. Epithelial cells were collected by centrifugation, in a culture medium containing 10% serum, C0 2 incubator base - cultured in the evening. Culture temperature is 370C, C0 2 concentration is preferably 5%. The serum is preferably calf serum or fetal calf serum. Further, it is preferable that an antibiotic having a concentration of about 1% (for example, penicillin G, 100 units Zml: streptomycin, 100 g Zml: amphotericin B, 0.25 g / ml) is co-present in the culture solution. The amniotic membrane remaining on the gauze can be re-fragmented and further dispersed in cells in the same manner as described above. Epithelial cells obtained are collected by centrifugation, as above, in a culture medium containing 10% serum, cultured in C0 2 incubator. The cell concentration of the culture is preferably 1 × 10 5 to 1 × 10 6 Zml. Normally, 5 XK) 7 epithelial cells can be isolated from one amniotic membrane. Amniotic mesenchymal cells can be isolated from amniotic membrane from which epithelial cells have been completely removed. The amniotic membrane remaining on the mesh such as gauze is treated with a protease such as 0.1-1.0 mgZml of collagenase (manufactured by Wako Pure Chemical Industries, Ltd.) and a nuclease such as 0.01-0.1 mgZml of DNAase (Sigma). Shake and digest. The shaking temperature is preferably 20 to 40 ° C, and the shaking time is preferably 30 to 120 minutes. Preferably, the shaking interval is 100 to: LOOO rpm. The digested amniotic membrane can be passed through a mesh such as gauze to separate mesenchymal cells. Collected resulting mesenchymal cells by centrifugation, in a culture medium containing 10% serum, cultured in C0 2 incubator. Culture temperature 37QC, C0 2 concentration has preferably 5%. The serum is preferably calf serum or fetal calf serum. In addition, it is preferable that an antibiotic (as described above) having a concentration of about 1% coexist in the culture solution. The cell density is preferably from 10 5 to 10 6 cells Zml. Mesenchymal cells can usually be separated into 5 x 10 6 cells from one amniotic membrane. The pharmaceutical composition of the present invention is characterized by containing human amniotic epithelial cells or mesenchymal cells obtained by the above-mentioned method. When epithelial cells or mesenchymal cells are administered to a patient, wash the cells carefully with saline to completely remove coexisting antibiotics and xenogenic serum. When administering cells, use cells suspended in physiological saline or culture solution. The cell concentration, 10 6 ~: L0 8 cells / ml are preferred. Growth factors and cell stabilizers may be mixed in to assist the cells. The administration site is not particularly limited. Depending on the disease, a site such as a spleen, a spleen, or a site of a rheumatic disease or a variety of blood vessels is selected. The total number of cells to be administered depends on the patient's condition, weight, and the like. The diseases that can be treated by the pharmaceutical composition or the therapeutic method of the present invention are as follows. Hyperlipidemia, hyperglycemia, impaired glucose tolerance (i-immediate ai red glucose tolerance: IGT), diabetic complications (eg retinopathy, nephropathy, cataract, coronary artery disease, etc.), arteriosclerosis, heart Vascular disease (eg, ischemic heart disease, etc.). Primary osteoporosis, endocrine osteoporosis (hyperthyroidism, hyperparathyroidism, Cushing's syndrome, and acromegaly), hereditary and congenital forms of osteoporosis (incomplete osteogenesis, homocystinuria, Menkes syndrome, and Riley) Osteoporosis such as osteoporosis due to di syndrome and limb fixation. Bone Paget's disease in adolescents and boys and girls (osteoarthritis). Hypercalcemia and hematological malignancies (solid myeloma, lymphoma and leukemia) from solid tumors (breast, lung and kidney), idiopathic hypercalcemia, and hyperthyroidism and renal function Hypercalcemia associated with insufficiency. Post-surgical steroid-induced osteopenia associated with small and large bowel disorders, and chronic hepatitis and kidney disease. Osteonecrosis or cell death associated with traumatic injury or with non-traumatic necrosis associated with Goge's disease, sickle cell anemia, systemic lupus erythematosus and other diseases. Bone loss due to chronic indirect rheumatism. Periodontal bone loss. Osteolytic metastases. Osteoarthritis, rheumatoid arthritis or spondyloarthropathy and indirect pain, shoulder and arm pain, low back pain and paralysis resulting therefrom. Ischemic heart disease (eg, angina, myocardial infarction, Kawasaki disease, etc.), valvular disease (eg, monk) Mitral stenosis, mitral regurgitation, mitral valve prolapse syndrome, aortic stenosis, aortic regurgitation, etc.), arrhythmia (eg, atrial fibrillation, atrial flutter, paroxysmal supraventricular tachycardia) , WPW syndrome, atrioventricular nodal recurrent tachycardia, ventricular extrasystole, ventricular tachycardia, ventricular fibrillation, bradyarrhythmia, sinus dysfunction syndrome, etc., aortic and vascular diseases (eg, ascending aortic aneurysm, arch Bilateral aortic aneurysm, thoracic aortic aneurysm, descending thoracic aortic aneurysm, thoracoabdominal aortic aneurysm, abdominal aortic aneurysm, obstructive atherosclerosis, venous thrombosis, pulmonary thromboembolism, etc.), congenital heart disease (eg, atrial septum) Deficiency, ventricular septal defect, Ei senmenger syndrome, pulmonary artery perfusion defect, patent arteriovascular patency, endocardial bed defect, complete macrovascular metastasis, modified macrovascular metastasis, pulmonary artery stenosis, Falot Tetralogy, aortic coarctation, Val salva sinus aneurysm, Ebstein malformation, etc.), cardiomyopathy (extended) Dilated cardiomyopathy, hypertrophic cardiomyopathy, restricted cardiomyopathy, etc.).
[図面の簡単な説明] [Brief description of drawings]
図 1は、 羊膜由来上皮細胞及び間葉系細胞図を表す。  FIG. 1 shows an amniotic membrane-derived epithelial cell and mesenchymal cell diagram.
図 2は、 羊膜由来上皮細胞及び間葉系細胞投与マウスにおける血糖値変動図 を表す。 図中 Mは間葉系細胞投与マウス、 HAEは上皮系細胞投与マウスの血 糖値の変動を表す。  FIG. 2 shows a blood glucose level fluctuation diagram in mice administered with amniotic epithelial cells and mesenchymal cells. In the figure, M represents the change in blood glucose level in mice administered with mesenchymal cells, and HAE represents changes in blood glucose levels in mice administered with epithelial cells.
図 3は、 上皮細胞移植脾臓におけるヒト β2-ミクログロプリン及びヒトインシ ュリンの発現を表す免疫組織染色図を表す。  FIG. 3 shows immunohistochemical staining showing the expression of human β2-microglobulin and human insulin in epithelial cell transplanted spleen.
図 4は、 培養羊膜由来間葉系細胞におけるヒトコラーゲンタイプ II遺伝子の 発現図。 左図は位相差顕微鏡写真であり、 形態変化を起こした細胞が観察され た。 右図は抗ヒトコラーゲンタイプ II抗体による免疫染色図であり、 形態変化 を起こした細胞にヒトコラーゲンタイプ IIの発現が観察された。  Figure 4 shows the expression of human collagen type II gene in cultured amnion-derived mesenchymal cells. The left figure is a phase contrast micrograph, in which cells that have undergone morphological changes were observed. The right figure is an immunostaining diagram using an anti-human collagen type II antibody. Expression of human collagen type II was observed in cells that had undergone morphological changes.
図 5は、分離した間葉系細胞を 100ng/mlの FGFまたは 5 x Mの 5·ァザシチ ヂン存在下で約 1ヶ月間培養し、 心筋特異的な NKX2.5に対する抗体で蛍光免 疫染色法により染色した図を表す。  Figure 5 shows that the isolated mesenchymal cells were cultured for about one month in the presence of 100 ng / ml FGF or 5 x M 5-azacitidine, and immunofluorescent staining was performed using a myocardial-specific antibody against NKX2.5. FIG.
図 6は、分離した間葉系細胞を 100ng/mlの FGFまたは の 5·ァザシチ ヂン存在下で約 1ヶ月間培養した後に RNAを抽出し、 ヒト NKX2.5遺伝子が 発現しているか否かを RT-PCR法で確認した図。 [発明を実施するための最良の形態] Figure 6 shows that the isolated mesenchymal cells were cultured in the presence of 100 ng / ml FGF or 5-azacitidine for about one month, and then RNA was extracted to determine whether the human NKX2.5 gene was expressed. FIG. 3 is a diagram in which RT was confirmed by the RT-PCR method. [Best Mode for Carrying Out the Invention]
以下実施例を挙げて本発明をさらに詳細に説明するが、 本発明はこれらに限 定されない。 実施例 1 .ヒト上皮細胞及び間葉細胞の分離  Hereinafter, the present invention will be described in more detail with reference to Examples, but the present invention is not limited thereto. Example 1 Isolation of human epithelial cells and mesenchymal cells
インフォームドコンセントを得たヒト妊婦を帝王切開し、 羊膜を入手した。 得られた羊膜を滅菌した手術用ハサミで断片化し、 0.2%トリブシンを含む細胞 培養液 (ダルベッコ修正イーグル培地:以下、 「DMEM」 という。) と共に 50ml のファルコンチューブに入れ、 37Q Cのシェーカーで lOOrpmで 30分間振盪し た。遠心後上清を除去し、 再度滅菌した手術用ハサミで断片化し、 37Q Cのシェ 一力一で 400-600rpmで 30分間振盪し、 0.2%トリプシンで消化した。 溶液を 滅菌ガーゼで越し、 上皮細胞を分離した。 上皮細胞は遠心によって集め、 培養 液 10%子牛胎児血清及び抗生物質 (ベニシリン G、 100単位 Zml:ストレブト マイシン、 100 g /ml:アンフォテリシン B、 0.25 g /ml) を含む DMEM で 370C、 5% C02下で培養した。 ガーゼに残された羊膜は、 再度滅菌した手術 用ハサミで断片化し、 同様に 0.2%トリプシンで 30分間消化した。 トリプシン 消化と細胞回収の操作を 3回繰り返し、上皮細胞を遠心で集め、 3 X 105細胞/ ml の細胞密度で、 10%子牛胎児血清及び上記と同様の抗生物質を含む DMEM で 370C、 5% C02下で培養した。 上皮細胞は、 通常羊膜一枚から 5 X 107個細胞が 分離できる。 羊膜間葉系の細胞は、 上皮細胞が完全に取り除かれた羊膜より、 分離した。 ガーゼ上に残された羊膜をコラゲナーゼ (0.75mgZml、 和光純薬 (株) 製)、 DNAァ一ゼ (0.075mgZml, シグマ社製) で、 37° C 60分間 600rpmで振盪 し、 消化した。 消化された羊膜をガーゼで越し、 間葉系細胞を分離した。 得ら れた間葉系細胞を遠心で集め、 4 X 105細胞/ mlの密度で 10%子牛胎児血清を含 む DMEMで 370C、 5% C02下で抗生物質 (ペニシリン G、 100単位 Zml:ス トレプトマイシン、 100 g /ml:アンフォテリシン B、 0.25 M g /ml) 存在 下で培養した。 間葉系細胞は、 通常羊膜一枚から 5 X 106個細胞が分離できた。 分離した羊膜由来上皮細胞と間葉系細胞の写真を図 1に示す。 試験例 1 .ヒト羊膜由来上皮細胞による糖尿病治療効果 A pregnant woman with informed consent was subjected to cesarean section to obtain an amniotic membrane. The resulting amniotic fragmented with surgical scissors sterile, cell culture medium containing 0.2% Toribushin (Dulbecco's modified Eagle's medium:. Hereinafter referred to as "DMEM") with placed in 50ml Falcon tubes, shaker of 37 Q C Shake at 100 rpm for 30 minutes. Removing the supernatant after centrifugation, fragmented with surgical scissors were sterilized again, shaking at 400-600rpm at 37 Q C Chez one force one 30 minutes, and digested with 0.2% trypsin. The solution was passed through sterile gauze to separate epithelial cells. Epithelial cells are collected by centrifugation, and the culture medium is 370C in DMEM containing 10% fetal calf serum and antibiotics (benicillin G, 100 units Zml: streptomycin, 100 g / ml: amphotericin B, 0.25 g / ml), 5% C0 2 and cultured under. The amniotic membrane remaining on the gauze was fragmented with re-sterilized surgical scissors and similarly digested with 0.2% trypsin for 30 minutes. The procedure of trypsin digestion and cell harvest was repeated three times, and the epithelial cells were collected by centrifugation at a cell density of 3 × 10 5 cells / ml, 370C in DMEM containing 10% fetal calf serum and the same antibiotics as above, 5% C0 2 were cultured under. As for epithelial cells, usually 5 × 10 7 cells can be separated from one amniotic membrane. Amniotic mesenchymal cells were isolated from amniotic membrane from which epithelial cells had been completely removed. The amniotic membrane left on the gauze was digested with collagenase (0.75 mgZml, manufactured by Wako Pure Chemical Industries, Ltd.) and DNAse (0.075 mgZml, manufactured by Sigma) at 37 ° C. for 60 minutes at 600 rpm for digestion. The digested amniotic membrane was passed through gauze to separate mesenchymal cells. Collected mesenchymal cells obtained al a centrifugal, 4 X 10 5 cells / ml with 10% fetal calf serum at a density in including DMEM and 370C, 5% C0 2 under antibiotic (penicillin G, 100 units (Zml: streptomycin, 100 g / ml: amphotericin B, 0.25 Mg / ml). Normally, 5 × 10 6 mesenchymal cells could be separated from one amniotic membrane. FIG. 1 shows photographs of the separated amniotic membrane-derived epithelial cells and mesenchymal cells. Test Example 1 Therapeutic effect of diabetes by human amniotic membrane-derived epithelial cells
( 1 ) 糖尿病マウスの作成  (1) Creation of diabetic mice
8-12週の免疫不全の SCID(sever combined immunodeficient )マウスをヒト 羊膜由来上皮細胞 (以下、 単に 「上皮細胞」 という。) 又はヒト羊膜由来間葉系 細胞 (以下、 単に 「間葉系細胞」 という。) の移植動物として使用した。 SCID マウスに 250mg/kgのストレプトゾトシン (streptozotocin:以下 「STZ」 とい う。) を濃度で腹腔内投与する事によって糖尿病を誘発し、 該マウスの体重の低 下や血糖値の上昇 (>300mg/dl) を確認した。 STZ 投与から 1週間後、 麻酔下 で、マウス脾臓に 1 X 1 0 6細胞 /0.1mlの上皮細胞または間葉系細胞を移植した。 移植マウスの血糖値は、 尻尾から採血した血清について、 一日おきにアベンテ イスファーマ株式会社のダイァセンサーを使用し同社の実験プロトコールに従 つて午前 9-11時の間に測定した。 移植後 2〜3週間経過後、 血糖値が正常範囲 «100mg/dl) に回復した後、 マウスを開腹し、 脾臓および滕臓を切除し、 以 後の解析に供した。 Eight to 12 weeks of immunodeficient SCID (sever combined immunodeficient) mice can be treated with human amnion-derived epithelial cells (hereinafter simply referred to as “epithelial cells”) or human amniotic-derived mesenchymal cells (hereinafter simply referred to as “mesenchymal cells”). ). Intraperitoneal administration of 250mg / kg streptozotocin (STZ) to SCID mice at a concentration induces diabetes, resulting in a decrease in body weight and an increase in blood glucose (> 300mg / dl). ) It was confirmed. One week after STZ administration, under anesthesia, 1 × 10 6 cells / 0.1 ml of epithelial cells or mesenchymal cells were transplanted into the mouse spleen. The blood glucose level of the transplanted mice was measured every 9 days between 9 and 11 am using the Aventis Pharma Co., Ltd. diasensor according to the experimental protocol of serum collected from the tail. Two to three weeks after the transplantation, after the blood glucose level recovered to the normal range <100 mg / dl), the mouse was laparotomized, and the spleen and the spleen were excised and subjected to the subsequent analysis.
( 2 ) 免疫染色 (2) Immunostaining
( 1 ) で得られたマウス脾臓及び滕臓にっき、 10%ホルマリン固定パラフィ ン切片を、 4乃至 6 ミクロンに薄切し、 スライドグラスに貼付して作成し、 使 用した。 免疫染色又は蛍光免疫のための一次抗体として、 以下のマウス由来モノク口 ーナル抗体を使用した。抗ヒトインスリン抗体(オンコジーン社製: Ab-lZl:400 希釈)、 抗ヒト β2·ミクログロプリン抗体(フアルミノーゲン社製: Col^Zl OO 希釈)、 及び、 抗ヒトコラーゲンタイプ II 抗体 (シグマ社製: Tu-99 1:1000 希釈)。 二次抗体としてはゥサギ抗マウス抗体を使用し、 抗原検出システムとし てストレプトアビジン-ピオチン法 (SAB-POキット:ニチレイ (株) 製) を使用 した。 但し、 ヒトコラ一ゲンタイプ IIの検出に限り二次抗体として FITC標識 ゥサギ抗マウス抗体を使用した。 0.01M-pH6.0クェン酸緩衝中で電子レンジで 1 5分間加熱処理を行い、 脱パラフィン後、 内因性パーォキシダーゼ阻止操作 (0.3 %過酸化水素添加メタノール中に 30分静止) を行った。 一次抗体結合反 応は室温で 1時間行い、 ピオチン標識二次抗体結合反応は室温で 30分行い、パ 一ォキシダーゼ標識ストレプトアビジン (酵素試薬)は室温で 30分結合させ、 そ の後 DAB (ジァミノベンチジン) 発色させた。 The mouse spleen and tentum obtained in (1) were cut into 10% formalin-fixed paraffin slices, sliced to 4 to 6 microns, attached to a slide glass, and used. The following mouse-derived monoclonal antibodies were used as primary antibodies for immunostaining or fluorescence immunization. Anti-human insulin antibody (manufactured by Oncogene: Ab-lZl: 400 dilution), anti-human β2-microglobulin antibody (manufactured by Pharmaluminogen: Col ^ ZlOO dilution), and anti-human collagen type II antibody (manufactured by Sigma) : 1:99 dilution of Tu-99). A heron anti-mouse antibody was used as a secondary antibody, and a streptavidin-biotin method (SAB-PO kit: manufactured by Nichirei Co., Ltd.) was used as an antigen detection system. However, only for the detection of human collagen type II, FITC labeling as a secondary antibody ゥ A heron anti-mouse antibody was used. Heat treatment in a microwave oven for 15 minutes in 0.01M-pH6.0 citrate buffer, deparaffinization, and endogenous peroxidase inhibition (rest in methanol with 0.3% hydrogen peroxide for 30 minutes) . The primary antibody binding reaction is performed at room temperature for 1 hour, the biotin-labeled secondary antibody binding reaction is performed at room temperature for 30 minutes, and the peroxidase-labeled streptavidin (enzyme reagent) is allowed to bind at room temperature for 30 minutes, followed by DAB Aminobenzidine) The color was developed.
( 3 ) PCRによる検討 (3) Examination by PCR
( 2 ) で得られたマウス脾臓及び塍臓にヒトの細胞が存在することを検討す るために、 これら組織より DNAを抽出し、 ヒト特異的な β2·ミクログロブリン 遺伝子が存在するか否かを PCR法で確認した。 以下に詳細を記載する。  In order to examine the presence of human cells in the mouse spleen and spleen obtained in (2), DNA was extracted from these tissues and the presence or absence of the human-specific β2-microglobulin gene was examined. Was confirmed by the PCR method. The details are described below.
モレキュラーリサーチセン夕一社の DNAzolを使って、 同社の実験方法に従 つて (2 ) で得られたマウス脾臓及び塍臓より DNA を抽出した。 得られた DNAl gを基質として、ヒト β2-ミクログロプリン遺伝子中のヒト特異ヌクレオ チド配列に基づき設計したプライマ一:  DNA was extracted from the mouse spleen and spleen obtained in (2) using DNAzol from Molecular Research Senyuichi according to the company's experimental method. Using the obtained DNAlg as a substrate, a primer designed based on the human-specific nucleotide sequence in the human β2-microglobulin gene:
5,- gtgtctgggt ttcatcaatc -3' (酉己列番号 1 ) ; 5, -gtgtctgggt ttcatcaatc -3 '(Rooster column number 1);
および and
5'- ggcaggcata ctcatctttt -3' (酉己歹 iJ番号 2 )  5'- ggcaggcata ctcatctttt -3 '(Toshimi system iJ number 2)
を用いて、 アマ一シャム社の Ready-To-Go PCR Beadsを同社の実験プロトコ —ルに従い使用して、 940C 1分、 560C 1分、 720C2分、 30サイクルで PCRを 行った。 PCR後得られた DNA鎖につき、 2 %ァガロースゲルを用い 1 0 0 V で一時間電気泳動を行い、 紫外線下で、 ヒト β2-ミクログロブリン遺伝子に相当 する 163bpのバンドを同定した。 Using the Ready-To-Go PCR Beads flax one Siamese Inc. its experimental protocol - using accordance Le, 940C 1 minute, 56 0 C 1 min, 720C2 minutes, PCR was performed in 30 cycles. The DNA strand obtained after PCR was subjected to electrophoresis at 100 V for 1 hour using a 2% agarose gel, and a 163 bp band corresponding to the human β2-microglobulin gene was identified under ultraviolet light.
( 4 ) 結果 (4) Result
8- 12週の免疫不全の SCIDマウスに STZ投与後 2日目、 血糖値は 300mg/dl を超え糖尿病が誘導できた。 STZ投与後一週間目に、 上皮細胞または間葉細胞 を脾臓に移植した。 血糖値は、 間葉細胞投与マウスでは 3例中 3例で高値のま まで低下せず、 一方、 上皮細胞投与マウスでは 4例中 1例で高値のままで低下 しなかったものの、 3 例で正常範囲 KlOOmgZdl) の値に回復した (図 2 )。 更に血糖値が正常範囲に回復したマウスで、上皮細胞を移植した脾臓において、 ヒトインスリンの発現が免疫組織学的に同定された (図 3 )。 さらに同脾臓で ヒト β2-ミクログロプリンが発現することが免疫組織学的に確認された(図 3 )。 さらに、 PCR により同脾臓中にヒト β2·ミクログロブリン遺伝子の存在が確認 できた。 これらの結果より、 ヒト羊膜上皮細胞より、 β 細胞が誘導されたこと が確認された。 試験例 2 . ヒト羊膜由来間葉系細胞の軟骨細胞への分化 On day 2 after STZ administration to 8- to 12-week immunodeficient SCID mice, blood glucose levels exceeded 300 mg / dl and diabetes was induced. One week after STZ administration, epithelial cells or mesenchymal cells were transplanted into the spleen. Blood glucose levels did not decrease to high levels in 3 of 3 mice treated with mesenchymal cells, while blood glucose levels remained low in 1 of 4 mice treated with epithelial cells Although they did not, they recovered to the normal range (KlOOmgZdl) in 3 cases (Fig. 2). Furthermore, in mice whose blood glucose levels had returned to the normal range, the expression of human insulin was identified immunohistologically in the spleen transplanted with epithelial cells (FIG. 3). Furthermore, it was confirmed immunohistologically that human β2-microglobulin was expressed in the spleen (FIG. 3). In addition, PCR confirmed the presence of the human β2-microglobulin gene in the spleen. From these results, it was confirmed that β cells were induced from human amniotic epithelial cells. Test Example 2. Differentiation of human amniotic membrane-derived mesenchymal cells into chondrocytes
分離した間葉系細胞を in vitroで 100ng/mlの Bone Mo rop ho genie Protein (BMP) 存在下で、 約 1ヶ月間高分子乳酸一ポリエチレングリコール (分子量約 9500) と 10%子牛胎児血清を含む DMEMで 370C、 5% C02で抗生物質 (ぺニ シリン G、 100単位 Zml:ストレプトマイシン、 100 ^ g Zml:アンフォテリ シン B、 0.25 g /ml) 存在下で共培養後、 軟骨細胞特異的なコラーゲンタイ プ IIの抗体で下記のように蛍光免疫染色法により解析した。 上記の培養間葉系 細胞をリン酸緩衝生理食塩水 (PBS) で洗净し、 100 %メタノールで - 20でで 10分間、 100%アセトンで - 20°C 1分間処理で細胞を固定した。 次に抗ヒトコ ラーゲンタイプ Π抗体 (シグマ社製: Tu-99/ l: 1000 希釈) を用い室温で 1 時間反応させ PBSで洗浄後、 二次抗体として FITC標識ゥサギ抗マウス抗体で 室温で 3 0分間反応させた後 PBSで洗诤した後、 蛍光顕微鏡で観察した。 その 結果、 形態が変化した細胞にのみ特異的にヒトコラーゲンタイプ II遺伝子の発 現が見られた (図 4 )。 この結果より、 ヒト羊膜間葉系細胞より、 軟骨細胞が 誘導できることが示された。 試験例 3 . ヒト羊膜由来間葉系細胞の心筋細胞への分化 The isolated mesenchymal cells were cultured in vitro in the presence of 100 ng / ml Bone Moropogenie Protein (BMP) for about 1 month with high molecular lactate-polyethylene glycol (molecular weight: about 9500) and 10% fetal calf serum. in containing DMEM 370C, 5% C0 2 with antibiotics (Bae two cylindrical G, 100 units ZML: streptomycin, 100 ^ g Zml: Anfoteri Singh B, 0.25 g / ml) after co-cultured in the presence of, cartilage cell-specific Analysis was performed by immunofluorescent staining with a collagen type II antibody as described below. The above cultured mesenchymal cells were washed with phosphate buffered saline (PBS), and fixed by treatment with 100% methanol at −20 for 10 minutes and 100% acetone at −20 ° C. for 1 minute. Then react with an anti-human collagen type II antibody (Sigma: Tu-99 / l: diluted 1000) for 1 hour at room temperature, wash with PBS, and use FITC-labeled heron anti-mouse antibody as a secondary antibody at room temperature for 30 hours. After reacting for 1 minute, the cells were washed with PBS and observed with a fluorescence microscope. As a result, the expression of the human collagen type II gene was specifically observed only in the cells whose morphology had changed (Fig. 4). The results showed that chondrocytes could be induced from human amniotic mesenchymal cells. Test Example 3. Differentiation of human amniotic membrane-derived mesenchymal cells into cardiomyocytes
( 1 )分離した間葉系細胞を in vitroで 100ng/mlの FGFまたは の 5-ァザ シチヂン(5-AZA)存在下で、約 1ヶ月間、 10%子牛胎児血清を含む 370 5% C0G 下で抗生物質 (ペニシリン G、 100単位ノ ml:ストレプトマイシン、 lOO g ml:アンフォテリシン B、 0.25 g/ml) 存在下で培養後、 心筋細胞特異的な転 写因子である NKX2.5(Komuro, I.; Izumo, S.: Csx: a murine (1) Isolated mesenchymal cells in vitro in the presence of 100 ng / ml FGF or 5-azacitidine (5-AZA) for about 1 month containing 10% fetal calf serum 370 5 % C0 G under antibiotics (penicillin G, 100 units Roh ml: streptomycin, Loo g ml: amphotericin B, 0.25 g / ml) after culture in the presence, cardiomyocyte-specific translocation NKX2.5 (Komuro, I .; Izumo, S .: Csx: a murine
homeobox-containing gene specifically expressed in the developing heart. Proc. Nat. Acad. Sci. 90: 8145-8149, 1993.)に対する抗体を用いた蛍光免疫染色法によ り解析した。 上記の培養間葉系細胞をリン酸緩衝生理食塩水 (PBS) で洗浄し、 100%メタノールで- 200C、 10分間、 100%アセトンで- 20。C、 1分間処理して細胞 を固定した。 次に抗ヒト NKX2.5抗体 (SANTA CRUZ社製: sc-14033を 1:200 希釈したもの) を用い室寧で 1時間反応させ、 PBSで洗浄した後、 二次抗体とし て FITC標識ャギ抗ゥサギ抗体で室温で 30分間反応させた後、 PBSで洗浄し、 蛍光顕微鏡で観察した。 その結果、 FGFで処理した羊膜間葉系細胞のみで、 Nat. Acad. Sci. 90: 8145-8149, 1993.) homeobox-containing gene specifically expressed in the developing heart. The above cultured mesenchymal cells are washed with phosphate buffered saline (PBS) and then washed with 100% methanol at -200C for 10 minutes and with 100% acetone at -20. C, treated for 1 minute to fix cells. Next, using an anti-human NKX2.5 antibody (manufactured by SANTA CRUZ: sc-14033 diluted 1: 200), react for 1 hour in a room, wash with PBS, and use FITC-labeled antibody as a secondary antibody. After reacting with an anti-Peacock antibody at room temperature for 30 minutes, the plate was washed with PBS and observed with a fluorescence microscope. As a result, only amniotic mesenchymal cells treated with FGF
NKX2.5蛋白質の発現が認められた (図 5 )。 この結果により、 ヒト羊膜間葉系細 胞から、 心筋細胞が誘導できることが示された。 Expression of the NKX2.5 protein was observed (FIG. 5). The results showed that cardiomyocytes could be induced from human amniotic mesenchymal cells.
( 2 ) RT-PCRによる検討  (2) Examination by RT-PCR
( 1 ) で得られた細胞で心筋細胞特異的な転写因子 NKX2.5遺伝子の転写が亢 進しているか否かを検討するために、 上記細胞から total RNA を抽出し、 NKX2.5遺伝子の発現を RT-PCR法で確認した。 RNAzol (モレキユラ一リサー チセンター社製) を添付のプロトコールに従って用いて、 (1 ) で得られた羊膜 細胞より total RNAを抽出した。 Total RNA、 l ^ gを基質とし、 ヒト NKX2.5 遺伝子中のヒト特異的ヌクレオチド配列に基づき設計したプライマー:  In order to examine whether the transcription of the NKX2.5 gene, a cardiomyocyte-specific transcription factor, is enhanced in the cells obtained in (1), total RNA was extracted from the above cells, and the NKX2.5 gene Expression was confirmed by RT-PCR. Total RNA was extracted from the amniotic cells obtained in (1) using RNAzol (Molecular Research Center) according to the attached protocol. Primers designed based on human-specific nucleotide sequence in human NKX2.5 gene using total RNA, l ^ g as substrate:
5'- cttcaagcca gaggcctacg -3' (酉己列表の酉己列番号 3 ) ; 5'-cttcaagcca gaggcctacg -3 '(Rooster column number 3 in Rooster column);
および and
5'· ccgcctctgt cttcttcagc -3' (配列表の配列番号 4 ) ;  5 ′ · ccgcctctgt cttcttcagc -3 ′ (SEQ ID NO: 4 in the sequence listing);
を用いて、 RT-PCRを行った。 反応は RT-PCRキット (TAKARA社製) を添付 のプロトコールに従って用い、 逆転写酵素で 420C、 30分間反応させて cDNA を作製し、 940Cで 30秒、 550Cで 30秒、 720Cで 30秒の温度サイクルを 40 回繰り返して PCR反応を行った。 得られた産物を 2%ァガロースゲルを用いて 100ポルトで 1時間、電気泳動を行い、紫外線下でヒト NKX2.5の mRNAから の増幅産物に相当する 233bpのバンドを同定した。その結果、 FGF存在下で培 養した細胞のみでヒ卜 NKX2.5特異的な増幅産物が確認された (図 6 )。 [産業上の利用の可能性] Was used to perform RT-PCR. The reaction is used according to the protocol attached RT-PCR kit (TAKARA Co.), a reverse transcriptase is reacted 420C, 30 minutes to prepare a cDNA, 30 seconds at 940C, 30 seconds at 55 0 C, 30 seconds at 720C The temperature cycle was repeated 40 times to perform the PCR reaction. The obtained product was subjected to electrophoresis using a 2% agarose gel at 100 ports for 1 hour, and a 233 bp band corresponding to an amplification product from human NKX2.5 mRNA was identified under ultraviolet light. As a result, a human NKX2.5-specific amplification product was confirmed only in cells cultured in the presence of FGF (FIG. 6). [Possibility of industrial use]
本発明を利用することにより、 倫理的問題の生じないように、 再生医療技術 を用い、 糖尿病治療薬等を提供することが可能になり、 または、 糖尿病患者等 を治療することが可能になる。  By utilizing the present invention, it is possible to provide a remedy for diabetes or the like using regenerative medicine technology or to treat a diabetic patient or the like without causing ethical problems.

Claims

請求の範囲 The scope of the claims
1 . ヒト羊膜由来上皮細胞を有効成分とする医薬組成物。  1. A pharmaceutical composition containing human amniotic membrane-derived epithelial cells as an active ingredient.
2 . ヒト羊膜由来上皮細胞を有効成分とする糖尿病治療剤。  2. An antidiabetic agent comprising human amniotic membrane-derived epithelial cells as an active ingredient.
3 . ヒト羊膜由来上皮細胞を糖尿病患者に投与することを特徴とする糖尿 病治療法。  3. A method for treating diabetes, comprising administering human amniotic membrane-derived epithelial cells to a diabetic patient.
4 . ヒト羊膜由来間葉系細胞を有効成分とする医薬組成物。  4. A pharmaceutical composition comprising human amniotic membrane-derived mesenchymal cells as an active ingredient.
5 . ヒト羊膜由来間葉系細胞を有効成分とする骨代謝異常症治療剤。  5. An agent for treating bone metabolism disorders comprising human amniotic membrane-derived mesenchymal cells as an active ingredient.
6 . ヒト羊膜由来間葉系細胞を骨代謝異常症患者に投与することを特徴と する骨代謝異常症治療法。  6. A method for treating bone metabolism disorders, which comprises administering human amniotic membrane-derived mesenchymal cells to patients with bone metabolism disorders.
7 . ヒト羊膜由来間葉系細胞を有効成分とする心疾患治療剤。  7. A therapeutic agent for heart disease comprising human amniotic membrane-derived mesenchymal cells as an active ingredient.
8 . ヒト羊膜由来間葉系細胞を心疾患患者に投与することを特徴とする心 疾患治療法。  8. A method for treating heart disease, comprising administering human amniotic membrane-derived mesenchymal cells to a patient with heart disease.
PCT/JP2002/012761 2001-12-06 2002-12-05 Medicinal compositions containing human amnion-origin cells WO2003047607A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU2002354101A AU2002354101A1 (en) 2001-12-06 2002-12-05 Medicinal compositions containing human amnion-origin cells

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
JP2001372878 2001-12-06
JP2001-372878 2001-12-06
JP2002262265A JP2005139067A (en) 2002-09-09 2002-09-09 Human amnion-originated cell-containing medicine composition for treating cardiac disease
JP2002-262265 2002-09-09

Publications (1)

Publication Number Publication Date
WO2003047607A1 true WO2003047607A1 (en) 2003-06-12

Family

ID=26624909

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP2002/012761 WO2003047607A1 (en) 2001-12-06 2002-12-05 Medicinal compositions containing human amnion-origin cells

Country Status (3)

Country Link
AU (1) AU2002354101A1 (en)
TW (1) TW200301132A (en)
WO (1) WO2003047607A1 (en)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1535994A1 (en) * 2002-08-23 2005-06-01 Srl, Inc. Human bone stem cells
JP2007528705A (en) * 2003-06-27 2007-10-18 エチコン、インコーポレイテッド Postpartum-derived cells for use in the treatment of heart and cardiovascular diseases
EP1833494A4 (en) * 2005-01-07 2010-05-19 Univ Wake Forest Health Sciences Regeneration of pancreatic islets by amniotic fluid stem cell therapy
US9085755B2 (en) 2004-08-16 2015-07-21 Cellresearch Corporation Pte Ltd. Isolation, cultivation and uses of stem/progenitor cells

Citations (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE2715887A1 (en) * 1977-04-09 1978-10-12 Ayguen Suerayya Tahsin Prof Dr Culturing human foetal cells - and injectable compsns. contg. human foetal cells for treating heart, kidney, liver, nervous, genital and circulatory diseases
WO1981002674A1 (en) * 1980-03-26 1981-10-01 F Taguchi Virus-inhibiting substance and process for preparing the same
JPS57171924A (en) * 1981-04-15 1982-10-22 Fumiaki Taguchi Virus controlling substance and its preparation
WO1984004669A1 (en) * 1983-05-24 1984-12-06 American Hospital Supply Corp Barrier layer for implantable tendons and ligaments
EP0230672A2 (en) * 1985-12-30 1987-08-05 American Hospital Supply Corporation Method for implantation of mammary prosthesis
WO1989007425A2 (en) * 1988-02-17 1989-08-24 Genethics Limited Clinical developments using amniotic cells
WO1997026902A1 (en) * 1996-01-23 1997-07-31 Srl, Inc. Cells for treating dementia
WO1999046366A1 (en) * 1998-03-13 1999-09-16 Osiris Therapeutics, Inc. Uses for humane non-autologous mesenchymal stem cells
WO2000029002A2 (en) * 1998-11-13 2000-05-25 Osiris Therapeutics, Inc. In utero transplantation of human mesenchymal stem cells
WO2000073421A2 (en) * 1999-06-02 2000-12-07 Lifebank Services, L.L.C. Methods of isolation, cryopreservation, and therapeutic use of human amniotic epithelial cells
JP2001161353A (en) * 1999-12-09 2001-06-19 Japan Ophthalmic Consultants:Kk Cell piece for transplantation and method for preparing the same
JP2002326943A (en) * 2001-02-28 2002-11-15 Japan Tissue Engineering:Kk Antiinflammatory agent

Patent Citations (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE2715887A1 (en) * 1977-04-09 1978-10-12 Ayguen Suerayya Tahsin Prof Dr Culturing human foetal cells - and injectable compsns. contg. human foetal cells for treating heart, kidney, liver, nervous, genital and circulatory diseases
WO1981002674A1 (en) * 1980-03-26 1981-10-01 F Taguchi Virus-inhibiting substance and process for preparing the same
JPS57171924A (en) * 1981-04-15 1982-10-22 Fumiaki Taguchi Virus controlling substance and its preparation
WO1984004669A1 (en) * 1983-05-24 1984-12-06 American Hospital Supply Corp Barrier layer for implantable tendons and ligaments
EP0230672A2 (en) * 1985-12-30 1987-08-05 American Hospital Supply Corporation Method for implantation of mammary prosthesis
WO1989007425A2 (en) * 1988-02-17 1989-08-24 Genethics Limited Clinical developments using amniotic cells
WO1997026902A1 (en) * 1996-01-23 1997-07-31 Srl, Inc. Cells for treating dementia
WO1999046366A1 (en) * 1998-03-13 1999-09-16 Osiris Therapeutics, Inc. Uses for humane non-autologous mesenchymal stem cells
WO2000029002A2 (en) * 1998-11-13 2000-05-25 Osiris Therapeutics, Inc. In utero transplantation of human mesenchymal stem cells
WO2000073421A2 (en) * 1999-06-02 2000-12-07 Lifebank Services, L.L.C. Methods of isolation, cryopreservation, and therapeutic use of human amniotic epithelial cells
JP2001161353A (en) * 1999-12-09 2001-06-19 Japan Ophthalmic Consultants:Kk Cell piece for transplantation and method for preparing the same
JP2002326943A (en) * 2001-02-28 2002-11-15 Japan Tissue Engineering:Kk Antiinflammatory agent

Non-Patent Citations (20)

* Cited by examiner, † Cited by third party
Title
ADINOLFI M. ET AL.: "Expression of HLA antigens, beta2-microglobulin and enzymes by human amniotic epithelial cells", NATURE, vol. 295, no. 5847, 1992, pages 325 - 327, XP002962928 *
ANDERSON H.C.: "Osteogenetic epithelial-mesenchymal cell interactions", CLINICAL ORTHOPAEDICS AND RELATED RESEARCH, no. 119, 1976, pages 211 - 223 *
ANDERSON H.C.: "The role of cells versus matrix in bone induction", CONNECTIVE TISSUE RESEARCH, vol. 24, no. 1, 1990, pages 3 - 12, XP002962930 *
ANDREA-ROMANA P. ET AL.: "Amniotic fluid cells and human cell research: a new connection", MEDICAL SCIENCE MONITOR, vol. 8, no. 11, November 2002 (2002-11-01), pages RA253 - RA257 *
BOYAN B.D. ET AL.: "Epithelial cell lines that induce bone formation in vivo produce alkaline phosphatase-enriched matrix vesicles in culture", CLINICAL ORTHOPAEDICS AND RELATED RESEARCH, no. 277, 1992, pages 266 - 276, XP002962931 *
CASEY M.L. ET AL.: "Interstitial collagen synthesis and processing in human amnion: a property of the mesenchymal cells", BIOL. REPROD., vol. 55, no. 6, 1996, pages 1253 - 1260, XP002962932 *
DAVIS G.E. ET AL.: "Human amnion membrane serves as a substratum for growing axons in vitro and in vivo", SCIENCE, vol. 236, no. 4805, 1987, pages 1106 - 1109, XP002962938 *
HUNT J.S. ET AL.: "Interferon-gamma induces class I HLA and beta2-microglobulin expression by human amnion cells", J. IMMUNOL., vol. 136, no. 2, 1986, pages 364 - 367, XP002962940 *
KAZUOMI HANADA: "Shashin seminar 205 yomaku deno kaiyo heisa", THE JOURNAL OF THE EYE, vol. 18, no. 6, June 2001 (2001-06-01), pages 731 - 732, XP002962937 *
KAZUTOMO INOUE: "Kansaibo to saisei iryo - rinsho oyo no tenbo -", SAIBO, vol. 34, no. 11, November 2002 (2002-11-01), pages 432 - 433, XP002962926 *
LEE S.W. ET AL.: "Immunohistological study of the localization of collagen rypes I-XI in bovine term placenta", ANIMAL SCIENCE JOURNAL, vol. 71, no. 5, 2000, pages 501 - 508, XP002962941 *
LIECHTY K.W. ET AL.: "Distribution and engraftment of human mesenchymal stem cells (MSC) after in utero transplantation in fatal sheep", BLOOD, vol. 92, no. 10, SUPPL. 1, 1998, pages 117A, XP000914604 *
MIGNATTI P. ET AL.: "In vitro angiogenesis on the human amniotic membrane: requirement for basic fibroblast growth factor-induced proteinases", J. CELL. BIOL., vol. 108, no. 2, 1989, pages 671 - 682, XP002962939 *
NATSUKI NAGATA ET AL.: "Saisei iryo toshite no saibo ryoho no genjo to kadai - suito saisei iryo eno approach", BIOTHERAPY, vol. 15, no. 2, March 2001 (2001-03-01), pages 105 - 110, XP002962929 *
NOBUO SAKURAGAWA: "Tokushu kansaibo kenkyu no shintenkai chusu shinkeikei kansaibo", SEITAI NO KAGAKU, vol. 49, no. 3, 1998, pages 196 - 202, XP002962934 *
NORIO KUMAGAYA: "Shorei zenshin 95% o jusho shita nessho kanji no jika baiyo hyohi ishoku ni yoru kyumeirei", BIOMEDICINE & THERAPEUTICS, vol. 35, no. 10, October 2001 (2001-10-01), pages 1131 - 1134, XP002962935 *
SAWANOBU ENO ET AL.: "Yomaku johi saibo no kansaibo-yo bunka to saisei iryo eno oyo kanosei ni tsuite", ORGAN BIOLOGY, vol. 9, no. 3, August 2002 (2002-08-01), pages 265 - 274, XP002962927 *
SHIGETO HARIMURA ET AL.: "Ganka ryoiki no tissue engineering", GENE & MEDICINE, vol. 5, no. 4, 10 November 2001 (2001-11-10), pages 609 - 613, XP002962936 *
WHITTLE W.L. ET AL.: "The characterization of human amnion epithelial and mesenchymal cells: the cellular expression, activity and glucocorticoid regulation of prostaglandin output", PLACENTA, vol. 21, no. 4, 2000, pages 394 - 401, XP002962933 *
WLODARSKI K.: "Failure of heterotopic osteogenesis by epithelial mesenchymal cell interactions in xenogeneic transplants in the kidney", CALCIF. TISSUE RES., vol. 25, no. 1, 1978, pages 7 - 12 *

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1535994A1 (en) * 2002-08-23 2005-06-01 Srl, Inc. Human bone stem cells
EP1535994A4 (en) * 2002-08-23 2005-12-07 Srl Inc Human bone stem cells
JP2007528705A (en) * 2003-06-27 2007-10-18 エチコン、インコーポレイテッド Postpartum-derived cells for use in the treatment of heart and cardiovascular diseases
US9085755B2 (en) 2004-08-16 2015-07-21 Cellresearch Corporation Pte Ltd. Isolation, cultivation and uses of stem/progenitor cells
US10363275B2 (en) 2004-08-16 2019-07-30 Cellresearch Corporation Pte Ltd Isolation, cultivation and uses of stem/progenitor cells
EP1833494A4 (en) * 2005-01-07 2010-05-19 Univ Wake Forest Health Sciences Regeneration of pancreatic islets by amniotic fluid stem cell therapy
US9056093B2 (en) 2005-01-07 2015-06-16 Wake Forest University Health Sciences Regeneration of pancreatic islets by amniotic fluid stem cell therapy

Also Published As

Publication number Publication date
AU2002354101A1 (en) 2003-06-17
TW200301132A (en) 2003-07-01

Similar Documents

Publication Publication Date Title
Comhair et al. Human primary lung endothelial cells in culture
CN110577931B (en) Intermittent hypoxia treatment stem cell source exosome and application thereof in myocardial tissues
CN102282252A (en) Amnion derived adherent cells
JPH07502651A (en) Senescent cell-derived DNA synthesis inhibitor
CN107028983A (en) The treatment of PUD D and illness
CN1728946A (en) Method of obtaining viable human liver cells, including hepatic stem/progenitor cells
Wruck et al. Human induced pluripotent stem cell-derived mesenchymal stem cells acquire rejuvenation and reduced heterogeneity
Ennis et al. Isolation, characterization, and differentiation of human umbilical cord perivascular cells (HUCPVCs)
CN110616190B (en) Method for regulating and controlling periodontal ligament stem cell osteogenic differentiation based on extracellular matrix
JP2007511219A5 (en)
JP2024041867A (en) Therapeutic use of gene-edited fibroblasts
FR2810045A1 (en) Preparation of specific cell populations from muscle biopsy, useful e.g. for reconstruction and repair of muscular, articular and bone tissue, by selective culture
CN111944748A (en) high-IL-10-expression human adipose-derived mesenchymal stem cell exosome for treating myocardial infarction and application thereof
WO2003047607A1 (en) Medicinal compositions containing human amnion-origin cells
WO2020158924A1 (en) Method for inducing feto-maternal immune tolerance
Chen et al. Mesenchymal stem cells promote tumor progression via inducing stroma remodeling on rabbit VX2 bladder tumor model
US20230233604A1 (en) Regulatory macrophages for treating angiopathies
US20140242142A1 (en) Cell secreted proteins for the treatment of myocardial infarction
JP2003231639A (en) Pharmaceutical composition containing cell derived from human amnion
Wang et al. Biomimetic polydopamine-laced hydroxyapatite collagen material orients osteoclast behavior to an anti-resorptive pattern without compromising osteoclasts’ coupling to osteoblasts
CN111925983A (en) Preparation method of high-IL-10-expression human adipose-derived mesenchymal stem cell exosome for treating myocardial infarction
CN111944747A (en) Human adipose-derived mesenchymal stem cell exosome for treating myocardial infarction and application thereof
EP3795677A1 (en) Composition for promoting stem cell differentiation, comprising progenitor cell culture solution and multilayer graphene film, and use thereof
CN114058591B (en) Recombinant mesenchymal stem cell and application thereof
CN114381425B (en) Modified mesenchymal stem cells, preparation method thereof and pharmaceutical composition

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NO NZ OM PH PL PT RO RU SC SD SE SG SK SL TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR IE IT LU MC NL PT SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
121 Ep: the epo has been informed by wipo that ep was designated in this application
122 Ep: pct application non-entry in european phase
NENP Non-entry into the national phase

Ref country code: JP