JPWO2020012991A1 - Mesenchymal stem cells and neuropathy therapeutic agents - Google Patents

Mesenchymal stem cells and neuropathy therapeutic agents Download PDF

Info

Publication number
JPWO2020012991A1
JPWO2020012991A1 JP2020530111A JP2020530111A JPWO2020012991A1 JP WO2020012991 A1 JPWO2020012991 A1 JP WO2020012991A1 JP 2020530111 A JP2020530111 A JP 2020530111A JP 2020530111 A JP2020530111 A JP 2020530111A JP WO2020012991 A1 JPWO2020012991 A1 JP WO2020012991A1
Authority
JP
Japan
Prior art keywords
cells
mesenchymal stem
cell
stem cells
medium
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
JP2020530111A
Other languages
Japanese (ja)
Inventor
格靖 石川
格靖 石川
陽子 堀内
陽子 堀内
崇史 瀧尻
崇史 瀧尻
輝 黒木
輝 黒木
真代 湯本
真代 湯本
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.)
Rohto Pharmaceutical Co Ltd
Original Assignee
Rohto Pharmaceutical Co Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Rohto Pharmaceutical Co Ltd filed Critical Rohto Pharmaceutical Co Ltd
Publication of JPWO2020012991A1 publication Critical patent/JPWO2020012991A1/en
Priority to JP2023215423A priority Critical patent/JP2024023760A/en
Pending legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/28Bone marrow; Haematopoietic stem cells; Mesenchymal stem cells of any origin, e.g. adipose-derived stem cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/48Reproductive organs
    • A61K35/51Umbilical cord; Umbilical cord blood; Umbilical stem cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system

Abstract

本発明は、神経障害の新規治療剤を提供することを目的とする。上記課題を解決するための本発明は、HGF, SHH, OLIG2, VEGFA, NEUROG1, GRPR, IL1R1, CRHR2, CCKAR, APOE, PAX3, PAX5, EGF, CXCL1, GDNF, NRCAM, DLL1, HEYL, BMP2, NTN1, ASCL1, NRP2のいずれか一つ、または二つ以上が高発現であることを特徴とする、間葉系幹細胞である。An object of the present invention is to provide a novel therapeutic agent for neuropathy. The present invention for solving the above problems is HGF, SHH, OLIG2, VEGFA, NEUROG1, GRPR, IL1R1, CRHR2, CCKAR, APOE, PAX3, PAX5, EGF, CXCL1, GDNF, NRCAM, DLL1, HEYL, BMP2, NTN1. , ASCL1, NRP2, one or more of which are highly expressed, are mesenchymal stem cells.

Description

本発明は、間葉系幹細胞及び神経障害治療剤に関する。 The present invention relates to mesenchymal stem cells and therapeutic agents for neuropathy.

高齢化社会の進行に伴い、脳の神経細胞の障害である神経変性疾患が年々増加を続けている。神経変性疾患としては、アルツハイマー病、パーキンソン病、筋萎縮性側索硬化症(ALS)や脊髄小脳変性症などが挙げられる。アルツハイマー病では、大脳皮質や海馬といった場所の神経細胞が脱落し、記憶能力が減退していく。また、パーキンソン病では、黒質と呼ばれる場所にある神経細胞が脱落し、運動機能に障害が出る。さらに、脳の神経細胞の障害によっておこる疾患としては、神経変性疾患の他にも脳卒中による脳梗塞や脳出血が知られている。 With the progress of an aging society, neurodegenerative diseases, which are disorders of nerve cells in the brain, continue to increase year by year. Neurodegenerative diseases include Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis (ALS) and spinocerebellar degeneration. In Alzheimer's disease, nerve cells in places such as the cerebral cortex and hippocampus are shed, and memory capacity declines. In Parkinson's disease, nerve cells in a place called the substantia nigra are shed, resulting in impaired motor function. Furthermore, cerebral infarction and cerebral hemorrhage due to stroke are known as diseases caused by damage to nerve cells in the brain, in addition to neurodegenerative diseases.

また、周産期の新生児脳障害は出生1,000人に1〜2人の頻度で発症し、その後の生涯にわたる脳性麻痺の原因となる。このような脳性麻痺の原因となる周産期脳障害には、低酸素性虚血性脳症、脳出血、脳室周囲白質軟化症などがあり、これらの主病態はミトコンドリア機能不全から生じる活性酸素の上昇、マクロファージの活性化とそれに伴う高サイトカイン血症という炎症病態である。上述の神経変性疾患や脳卒中、脳性麻痺は、疾患の原因及び症状は異なっているが、神経細胞の数が減るという一点においては共通していると言える。 In addition, perinatal neonatal cerebral disorders occur at a frequency of 1 to 2 in 1,000 live births and cause cerebral palsy for the rest of their lives. Perinatal cerebral disorders that cause such cerebral palsy include hypoxic-ischemic encephalopathy, cerebral hemorrhage, and periventricular leukomalacia, and these main pathological conditions are elevated active oxygen caused by mitochondrial dysfunction. , Macrophage activation and associated hypercytokineemia, an inflammatory condition. The above-mentioned neurodegenerative diseases, strokes, and cerebral palsy can be said to be common in that the causes and symptoms of the diseases are different, but the number of nerve cells is reduced.

アルツハイマー病にはドネペジル、パーキンソン病にはレボドパなどの治療薬が用いられているが、これらの治療薬は神経細胞が減少したことによる神経ネットワークの情報処理機能を、化学伝達物質シグナルの補充によって回復させることを目的としたものであり、神経細胞の減少自身を抑えることはできない。また、神経変性疾患や脳卒中に対する既存の治療薬の中で、神経細胞を保護する作用を示すものはない。そのため神経細胞を保護する作用を有する新規治療薬の開発が望まれている。 Therapeutic agents such as donepezil for Alzheimer's disease and levodopa for Parkinson's disease are used, and these therapeutic agents restore the information processing function of the neural network due to the decrease in nerve cells by supplementing the chemical transmitter signal. The purpose is to make it, and the decrease of nerve cells itself cannot be suppressed. In addition, none of the existing therapeutic agents for neurodegenerative diseases and strokes have a protective effect on nerve cells. Therefore, the development of a new therapeutic agent having an action of protecting nerve cells is desired.

間葉系幹細胞は、1982年にFriedensteinによって初めて骨髄から単離された多分化能を有する前駆細胞である(非特許文献1参照)。間葉系幹細胞は、骨髄、臍帯、脂肪等の様々な組織に存在することが明らかにされており、間葉系幹細胞移植は、様々な難治性疾患に対する新しい治療方法として、期待されている(特許文献1〜2参照)。最近では、脂肪組織、胎盤、臍帯、卵膜等の間質細胞に同等の機能を有する細胞が存在することが知られている。従って、間葉系幹細胞を間質細胞(Mesenchymal Stromal Cell)と称することもある。 Mesenchymal stem cells are pluripotent progenitor cells first isolated from bone marrow by Friedenstein in 1982 (see Non-Patent Document 1). It has been clarified that mesenchymal stem cells are present in various tissues such as bone marrow, umbilical cord, and adipose tissue, and mesenchymal stem cell transplantation is expected as a new therapeutic method for various intractable diseases ( See Patent Documents 1 and 2). Recently, it is known that there are cells having the same function as stromal cells such as adipose tissue, placenta, umbilical cord, and fetal membrane. Therefore, mesenchymal stem cells are sometimes referred to as stromal cells (Mesenchymal Stromal Cells).

特開2012−157263号公報Japanese Unexamined Patent Publication No. 2012-157263 特表2012−508733号公報Japanese Patent Application Laid-Open No. 2012-508733

Pittenger F.M.et al.Science ,(1999),284,pp.143-147Pittenger F.M.et al.Science, (1999), 284, pp.143-147

本発明は、上述のような状況の中、神経障害の新規治療剤を提供することを目的とする。 An object of the present invention is to provide a novel therapeutic agent for neuropathy under the above-mentioned circumstances.

上記課題を解決するために鋭意研究した結果、本発明者らは、HGF, SHH, OLIG2, VEGFA, NEUROG1, GRPR, IL1R1, CRHR2, CCKAR, APOE, PAX3, PAX5, EGF, CXCL1, GDNF, NRCAM, DLL1, HEYL, BMP2, NTN1, ASCL1, NRP2のいずれか一つ、または二つ以上が高発現する間葉系幹細胞(mesenchymal stem(stromal) cell; MSC)が、神経障害の治療に有効であることを見出し、本発明を完成させた。本発明によれば、神経障害の治療のために有効な治療剤を提供できる。すなわち本発明の要旨は、以下の通りである。 As a result of diligent research to solve the above problems, the present inventors have HGF, SHH, OLIG2, VEGFA, NEUROG1, GRPR, IL1R1, CRHR2, CCKAR, APOE, PAX3, PAX5, EGF, CXCL1, GDNF, NRCAM, Mesenchymal stem (stromal) cells (MSCs) in which one or more of DLL1, HEYL, BMP2, NTN1, ASCL1, and NRP2 are highly expressed are effective in treating neuropathy. And completed the present invention. According to the present invention, it is possible to provide an effective therapeutic agent for the treatment of neuropathy. That is, the gist of the present invention is as follows.

すなわち、本発明は、下記に関するものである:
[1]HGF, SHH, OLIG2, VEGFA, NEUROG1, GRPR, IL1R1, CRHR2, CCKAR, APOE, PAX3, PAX5, EGF, CXCL1, GDNF, NRCAM, DLL1, HEYL, BMP2, NTN1, ASCL1, NRP2のいずれか一つ、または二つ以上が高発現であることを特徴とする、間葉系幹細胞。
[2]他家由来である、[1]に記載の間葉系幹細胞。
[3]臍帯組織もしくは脂肪組織由来である、[1]又は[2]に記載の間葉系幹細胞。
[4]浮遊培養法により調製される[1]から[3]のいずれかに記載の間葉系幹細胞。
[5][1]から[4]のいずれかに記載の間葉系幹細胞を含有する神経障害治療剤。
That is, the present invention relates to:
[1] One of HGF, SHH, OLIG2, VEGFA, NEUROG1, GRPR, IL1R1, CRHR2, CCKAR, APOE, PAX3, PAX5, EGF, CXCL1, GDNF, NRCAM, DLL1, HEYL, BMP2, NTN1, ASCL1, NRP2 Mesenchymal stem cells characterized by high expression of one or more.
[2] The mesenchymal stem cell according to [1], which is derived from an allogeneic family.
[3] The mesenchymal stem cell according to [1] or [2], which is derived from umbilical cord tissue or adipose tissue.
[4] The mesenchymal stem cell according to any one of [1] to [3] prepared by the suspension culture method.
[5] The therapeutic agent for neuropathy containing mesenchymal stem cells according to any one of [1] to [4].

本発明によると、神経障害の新規治療剤を提供することができる。 According to the present invention, it is possible to provide a novel therapeutic agent for neuropathy.

図1は、各種培地による培養で得られた間葉系幹細胞における、各種因子のmRNA発現量を比較した結果を示す図である。FIG. 1 is a diagram showing the results of comparing the mRNA expression levels of various factors in mesenchymal stem cells obtained by culturing in various media. 図2は、間葉系幹細胞について、平面培養細胞(ADH)と浮遊培養細胞(SUS)における各種因子のmRNA発現量を比較した結果を示す図である。FIG. 2 is a diagram showing the results of comparing the mRNA expression levels of various factors in planar cultured cells (ADH) and suspension cultured cells (SUS) for mesenchymal stem cells. 各種培養法による培養で得られた間葉系幹細胞の、ラット一過性脳虚血モデルへの投与効果を示す図である(体重)。It is a figure which shows the administration effect to the rat transient ischemic attack model of the mesenchymal stem cell obtained by culture by various culture methods (body weight). 各種培養法による培養で得られた間葉系幹細胞の、ラット一過性脳虚血モデルへの投与効果を示す図である(神経症状スコア)。It is a figure which shows the administration effect to the rat transient ischemic attack model of the mesenchymal stem cell obtained by culture by various culture methods (neurosymptom score). 各種培養法による培養で得られた間葉系幹細胞の、ラット一過性脳虚血モデルへの投与効果を示す図である(ステップ回数)。It is a figure which shows the administration effect to the rat transient ischemic attack model of the mesenchymal stem cell obtained by culture by various culture methods (the number of steps). 各種培養法による培養で得られた間葉系幹細胞の、ラット一過性脳虚血モデルへの投与効果を示す図である(テープ剥がしテスト)。It is a figure which shows the administration effect to the rat transient ischemic attack model of the mesenchymal stem cell obtained by culture by various culture methods (tape peeling test). 神経細胞と間葉系幹細胞の細胞間相互作用の様子を示すタイムラプス画像である。It is a time-lapse image showing the state of cell-cell interaction between nerve cells and mesenchymal stem cells. 無血清培地(Rohto社)による培養で得られた間葉系幹細胞の、神経細胞死抑制効果を示す図である。It is a figure which shows the nerve cell death inhibitory effect of the mesenchymal stem cell obtained by culturing in the serum-free medium (Rohto). 無血清培地(Rohto社)による培養で得られた間葉系幹細胞の、用量依存的な神経細胞賦活効果を示す図である。It is a figure which shows the dose-dependent nerve cell activation effect of the mesenchymal stem cell obtained by culturing in the serum-free medium (Rohto). 無血清培地(Rohto社)を用いて平面(ADH)もしくは浮遊(SUS)培養で得られた間葉系幹細胞の、神経細胞賦活効果を示す図である(線維芽細胞との比較)。It is a figure which shows the nerve cell activation effect of the mesenchymal stem cell obtained by the plane (ADH) or suspension (SUS) culture using serum-free medium (Rohto) (comparison with fibroblast). 無血清培地(Rohto社)による培養で得られた間葉系幹細胞の、神経細胞賦活効果を示す図である(他の培地で培養された細胞との比較)。It is a figure which shows the nerve cell activation effect of the mesenchymal stem cell obtained by culturing in a serum-free medium (Rohto) (comparison with the cell cultured in another medium). 無血清培地(Rohto社)による培養で得られた間葉系幹細胞の、神経細胞死抑制効果を示す細胞写真である(他の培地で培養された細胞との比較)。It is a cell photograph which shows the nerve cell death inhibitory effect of the mesenchymal stem cell obtained by culturing in a serum-free medium (Rohto) (comparison with the cell cultured in another medium). 無血清培地(Rohto社)による培養で得られた間葉系幹細胞の、神経細胞死抑制効果を示す図である(他の培地で培養された細胞との比較)。It is a figure which shows the nerve cell death inhibitory effect of the mesenchymal stem cell obtained by culturing in a serum-free medium (Rohto) (comparison with the cell cultivated in another medium).

以下、本発明の間葉系幹細胞、神経障害治療剤について詳細に説明する。 Hereinafter, the mesenchymal stem cells and the therapeutic agent for neuropathy of the present invention will be described in detail.

[間葉系幹細胞]
本発明の間葉系幹細胞は、HGF(hepatocyte growth factor), SHH(sonic hedgehog), OLIG2(oligodendrocyte transcription factor 2), VEGFA(vascular endothelial growth factor A), NEUROG1(neurogenin 1), GRPR(gastrin releasing peptide receptor), IL1R1(interleukin-1 receptor 1), CRHR2(corticotropin releasing hormone receptor 2), CCKAR(cholecystokinin A receptor), APOE(apolipoprotein E), PAX3(paired box 3), PAX5(paired box 5), EGF(epidermal growth factor), CXCL1(C-X-C motif chemokine ligand 1), GDNF(glial cell derived neurotrophic factor), NRCAM(neuronal cell adhesion molecule), DLL1(delta like canonical Notch ligand 1), HEYL(hes related family bHLH transcription factor with YRPW motif-like), BMP2(bone morphogenetic protein 2), NTN1(netrin 1), ASCL1(achaete-scute family bHLH transcription factor 1), NRP2(neuropilin 2)のいずれか一つ、または二つ以上が高発現であることを特徴とする。
[Mesenchymal stem cells]
The mesenchymal stem cells of the present invention are HGF (hepatocyte growth factor), SHH (sonic hedgehog), OLIG2 (oligodendrocyte transcription factor 2), VEGFA (vascular endothelial growth factor A), NEUROG1 (neurogenin 1), GRPR (gastrin releasing peptide). receptor), IL1R1 (interleukin-1 receptor 1), CRHR2 (corticotropin releasing hormone receptor 2), CCKAR (cholecystokinin A receptor), APOE (apolipoprotein E), PAX3 (paired box 3), PAX5 (paired box 5), EGF ( epidermal growth factor), CXCL1 (CXC motif chemokine ligand 1), GDNF (glial cell derived neurotrophic factor), NRCAM (neuronal cell adhesion molecule), DLL1 (delta like canonical Notch ligand 1), HEYL (hes related family bHLH transcription factor with High expression of any one or more of YRPW motif-like), BMP2 (bone morphogenetic protein 2), NTN1 (netrin 1), ASCL1 (achaete-scute family bHLH transcription factor 1), NRP2 (neuropilin 2) It is characterized by being.

HGFは成長因子の一種であり、神経保護作用を有することが知られている。SHHはヘッジホッグファミリーに属する遺伝子であり、酸化ストレスに対する神経細胞の保護に関与することが知られている。VEGFAは成長因子の一種であり、血管新生を誘導することで神経細胞の保護に関与することが知られている。IL1R1は炎症性サイトカインの一種であるIL1の受容体であり、炎症応答に関与することが知られている。DLL1はNotchリガンドであるDSLファミリーに属し、Notchシグナルの調節に関与することが知られている。 HGF is a type of growth factor and is known to have a neuroprotective effect. SHH is a gene belonging to the hedgehog family and is known to be involved in the protection of nerve cells against oxidative stress. VEGFA is a type of growth factor and is known to be involved in the protection of nerve cells by inducing angiogenesis. IL1R1 is a receptor for IL1, which is a type of inflammatory cytokine, and is known to be involved in the inflammatory response. DLL1 belongs to the DSL family, which is a Notch ligand, and is known to be involved in the regulation of Notch signaling.

なお、上記HGF, SHH, OLIG2, VEGFA, NEUROG1, GRPR, IL1R1, CRHR2, CCKAR, APOE, PAX3, PAX5, EGF, CXCL1, GDNF, NRCAM, DLL1, HEYL, BMP2, NTN1, ASCL1, NRP2が高発現であるとは、それぞれのmRNA発現が高発現であること、若しくはそれぞれのタンパク質が高発現であること、又はその両方が高発現であることを含む。 The above HGF, SHH, OLIG2, VEGFA, NEUROG1, GRPR, IL1R1, CRHR2, CCKAR, APOE, PAX3, PAX5, EGF, CXCL1, GDNF, NRCAM, DLL1, HEYL, BMP2, NTN1, ASCL1, NRP2 are highly expressed. Being includes that each mRNA expression is highly expressed, each protein is highly expressed, or both are highly expressed.

また、本発明の間葉系幹細胞は、他の細胞に比べ、上記因子を高発現していればよいが、具体的には、本発明の間葉系幹細胞は、従来の培養条件下(例えば、10%FBS含有MEM-α培地による培養)で得られる間葉系幹細胞に比べて、上記因子を高発現していればよい。好ましくは従来の培養条件下で得られる間葉系幹細胞に比べ2倍以上、より好ましくは5倍以上、さらに好ましくは10倍以上、特に好ましくは100倍以上発現している。 Further, the mesenchymal stem cell of the present invention may express the above-mentioned factors higher than other cells, but specifically, the mesenchymal stem cell of the present invention may be subjected to conventional culture conditions (for example,). It suffices if the above factors are highly expressed as compared with the mesenchymal stem cells obtained by culturing in MEM-α medium containing 10% FBS). It is preferably expressed twice or more, more preferably five times or more, still more preferably ten times or more, and particularly preferably 100 times or more, as compared with the mesenchymal stem cells obtained under conventional culture conditions.

本発明の間葉系幹細胞は、線維芽細胞に比べて、HGF, SHH, OLIG2, VEGFA, NEUROG1, GRPR, IL1R1, CRHR2, CCKAR, APOE, PAX3, PAX5, EGF, CXCL1, GDNF, NRCAM, DLL1, HEYL, BMP2, NTN1, ASCL1, NRP2のいずれか一つ、または二つ以上を高発現していればよい。好ましくは皮膚線維芽細胞に対して2倍以上、より好ましくは5倍以上、さらに好ましくは10倍以上、特に好ましくは100倍以上発現している。 Compared with fibroblasts, the mesenchymal stem cells of the present invention have HGF, SHH, OLIG2, VEGFA, NEUROG1, GRPR, IL1R1, CRHR2, CCKAR, APOE, PAX3, PAX5, EGF, CXCL1, GDNF, NRCAM, DLL1, Any one or more of HEYL, BMP2, NTN1, ASCL1 and NRP2 may be highly expressed. It is preferably expressed twice or more, more preferably five times or more, still more preferably ten times or more, and particularly preferably 100 times or more with respect to skin fibroblasts.

本発明において間葉系幹細胞とは、間葉系に属する一種以上の細胞(骨細胞、心筋細胞、軟骨細胞、腱細胞、脂肪細胞など)への分化能を有し、当該能力を維持したまま増殖できる細胞を意味する。本発明において用いる間葉系幹細胞なる用語は、間質細胞と同じ細胞を意味し、両者を特に区別するものではない。また、単に間葉系細胞と表記される場合もある。間葉系幹細胞を含む組織としては、例えば、脂肪組織、臍帯、骨髄、臍帯血、子宮内膜、胎盤、羊膜、絨毛膜、脱落膜、真皮、骨格筋、骨膜、歯小嚢、歯根膜、歯髄、歯胚等が挙げられる。例えば脂肪組織由来間葉系幹細胞とは、脂肪組織に含有される間葉系幹細胞を意味し、脂肪組織由来間質細胞と称してもよい。これらのうち、神経障害疾患の治療に対する有効性の観点、入手容易性の観点等から、脂肪組織由来間葉系幹細胞、臍帯由来間葉系幹細胞、骨髄由来間葉系幹細胞、胎盤由来間葉系幹細胞、歯髄由来間葉系幹細胞が好ましく、脂肪組織由来間葉系幹細胞、臍帯由来間葉系幹細胞がより好ましく、臍帯由来間葉系幹細胞が最も好ましい。 In the present invention, the mesenchymal stem cell has an ability to differentiate into one or more cells belonging to the mesenchymal system (osteocytes, myocardial cells, chondrocytes, tendon cells, adipocytes, etc.), and maintains the ability. It means a cell that can proliferate. The term mesenchymal stem cell used in the present invention means the same cell as a stromal cell, and does not particularly distinguish between the two. It may also be simply referred to as mesenchymal cells. Tissues containing mesenchymal stem cells include, for example, adipose tissue, umbilical cord, bone marrow, umbilical cord blood, endometrial membrane, placenta, amniotic membrane, chorion, decidua, dermatitis, skeletal muscle, bone membrane, dental follicle, periodontal ligament, etc. Examples include dental pulp and tooth germ. For example, adipose tissue-derived mesenchymal stem cells mean mesenchymal stem cells contained in adipose tissue, and may be referred to as adipose tissue-derived stromal cells. Of these, adipose tissue-derived mesenchymal stem cells, umbilical band-derived mesenchymal stem cells, bone marrow-derived mesenchymal stem cells, and placenta-derived mesenchymal stem cells are used from the viewpoints of effectiveness in treating neuropathy diseases and availability. Stem cells and mesenchymal stem cells derived from dental pulp are preferable, mesenchymal stem cells derived from adipose tissue and mesenchymal stem cells derived from umbilical cord are more preferable, and mesenchymal stem cells derived from umbilical cord are most preferable.

本発明における間葉系幹細胞は、処置される対象(被検体)と同種由来であってもよいし、異種由来であってもよい。本発明における間葉系幹細胞の種として、ヒト、ウマ、ウシ、ヒツジ、ブタ、イヌ、ネコ、ラビット、マウス、ラットが挙げられ、好ましくは処置される対象(被検体)と同種由来細胞である。本発明における間葉系幹細胞は、処置される対象(被検体)に由来、すなわち自家細胞であってもよいし、同種の別の対象に由来、すなわち他家細胞であってもよい。好ましくは他家細胞である。 The mesenchymal stem cells in the present invention may be derived from the same species as the subject (subject) to be treated, or may be derived from a different species. Examples of the species of mesenchymal stem cells in the present invention include humans, horses, cows, sheep, pigs, dogs, cats, rabbits, mice, and rats, and preferably cells derived from the same species as the subject (subject) to be treated. .. The mesenchymal stem cells in the present invention may be derived from a subject (subject) to be treated, that is, an autologous cell, or may be derived from another subject of the same species, that is, an allogeneic cell. It is preferably an allogeneic cell.

間葉系幹細胞は同種異系の被験体に対しても拒絶反応を起こしにくいため、あらかじめ調製されたドナーの細胞を拡大培養して凍結保存したものを、本発明の疾患治療剤における間葉系幹細胞として使用することができる。そのため、自己の間葉系幹細胞を調製して用いる場合と比較して、商品化も容易であり、かつ安定して一定の効果を得られ易いという観点から、本発明における間葉系幹細胞は、同種異系であることがより好ましい。 Since mesenchymal stem cells are less likely to cause rejection of allogeneic subjects, pre-prepared donor cells are expanded and cryopreserved in the mesenchymal system in the disease therapeutic agent of the present invention. It can be used as a stem cell. Therefore, the mesenchymal stem cells in the present invention can be easily commercialized and can obtain a stable and constant effect as compared with the case where self-mesenchymal stem cells are prepared and used. It is more preferable that they are allogeneic.

本発明において間葉系幹細胞とは、間葉系幹細胞を含む任意の細胞集団を意味する。当該細胞集団は、少なくとも20%以上、好ましくは、30%、40%、50%、60%、70%、75%、80%、85%、90%、93%、96%、97%、98%又は99%以上が間葉系幹細胞である。 In the present invention, the mesenchymal stem cell means an arbitrary cell population including the mesenchymal stem cell. The cell population is at least 20% or more, preferably 30%, 40%, 50%, 60%, 70%, 75%, 80%, 85%, 90%, 93%, 96%, 97%, 98. % Or 99% or more are mesenchymal stem cells.

本発明において臍帯とは、胎児と胎盤を結ぶ白い管状の組織であり、臍帯静脈、臍帯動脈、膠様組織(ウォートンジェリー;Wharton's Jelly)、臍帯基質自体等から構成され、間葉系幹細胞を多く含む。臍帯は、本発明の疾患治療剤を使用する被験体(投与対象)と同種動物から入手されることが好ましく、本発明の疾患治療剤をヒトへ投与することを考慮すると、より好ましくは、ヒトの臍帯である。 In the present invention, the umbilical cord is a white tubular tissue that connects the foetation and the placenta, and is composed of umbilical veins, umbilical arteries, glue-like tissues (Wharton's Jelly), umbilical cord matrix itself, and many other mesenchymal stem cells. Including. The umbilical cord is preferably obtained from an animal of the same species as the subject (administration target) using the therapeutic agent for the disease of the present invention, and more preferably for humans in consideration of administering the therapeutic agent for the disease of the present invention to humans. Umbilical cord.

本発明において脂肪組織とは、脂肪細胞、及び微小血管細胞等を含む間質細胞を含有する組織を意味し、例えば、哺乳動物の皮下脂肪を外科的切除又は吸引して得られる組織である。脂肪組織は、皮下脂肪より入手され得る。後述する脂肪組織由来間葉系幹細胞の投与対象と同種動物から入手されることが好ましく、ヒトへ投与することを考慮すると、より好ましくは、ヒトの皮下脂肪である。皮下脂肪の供給個体は、生存していても死亡していてもよいが、本発明において用いる脂肪組織は、好ましくは、生存個体から採取された組織である。個体から採取する場合、脂肪吸引は、例えば、PAL(パワーアシスト)脂肪吸引、エルコーニアレーザー脂肪吸引、又は、ボディジェット脂肪吸引などが例示され、細胞の状態を維持するという観点から、超音波を用いないことが好ましい。 In the present invention, the adipose tissue means a tissue containing stromal cells including adipocytes and microvascular cells, and is, for example, a tissue obtained by surgically excising or aspirating subcutaneous fat of a mammal. Adipose tissue can be obtained from subcutaneous fat. It is preferably obtained from an animal of the same species as the adipose tissue-derived mesenchymal stem cell to be administered, which will be described later, and more preferably human subcutaneous fat in consideration of administration to humans. The subcutaneous fat supply individual may be alive or dead, but the adipose tissue used in the present invention is preferably a tissue collected from a surviving individual. When collected from an individual, liposuction includes, for example, PAL (power assist) liposuction, erconia laser liposuction, body jet liposuction, etc., and ultrasonic waves from the viewpoint of maintaining the state of cells. It is preferable not to use.

本発明において骨髄とは、骨の内腔を満たしている柔組織のことをいい、造血器官である。骨髄中には骨髄液が存在し、その中に存在する細胞を骨髄細胞と呼ぶ。骨髄細胞には、赤血球、顆粒球、巨核球、リンパ球、脂肪細胞等の他、間葉系幹細胞、造血幹細胞、血管内皮前駆細胞等が含まれている。骨髄細胞は、例えば、ヒト腸骨、長管骨、又はその他の骨から採取することができる。 In the present invention, the bone marrow refers to parenchyma that fills the lumen of bone and is a hematopoietic organ. Bone marrow fluid exists in the bone marrow, and the cells existing in it are called bone marrow cells. Bone marrow cells include erythrocytes, granulocytes, megakaryocytes, lymphocytes, adipocytes and the like, as well as mesenchymal stem cells, hematopoietic stem cells, vascular endothelial progenitor cells and the like. Bone marrow cells can be harvested from, for example, human ilium, long bone, or other bone.

本発明において、脂肪組織由来間葉系幹細胞、臍帯由来間葉系幹細胞、骨髄由来間葉系幹細胞といった各組織由来間葉系幹細胞とは、それぞれ脂肪組織由来間葉系幹細胞、臍帯由来間葉系幹細胞、骨髄由来間葉系幹細胞といった各組織由来間葉系幹細胞を含む任意の細胞集団を意味する。当該細胞集団は、少なくとも20%以上、好ましくは、30%、40%、50%、60%、70%、75%、80%、85%、90%、93%、96%、97%、98%又は99%以上が、脂肪組織由来間葉系幹細胞、臍帯由来間葉系幹細胞、骨髄由来間葉系幹細胞である各組織由来間葉系幹細胞である。 In the present invention, each tissue-derived mesenchymal stem cell such as adipose tissue-derived mesenchymal stem cell, umbilical band-derived mesenchymal stem cell, and bone marrow-derived mesenchymal stem cell refers to adipose tissue-derived mesenchymal stem cell and umbilical band-derived mesenchymal stem cell, respectively. It means an arbitrary cell population including mesenchymal stem cells derived from each tissue such as stem cells and mesenchymal stem cells derived from bone marrow. The cell population is at least 20% or more, preferably 30%, 40%, 50%, 60%, 70%, 75%, 80%, 85%, 90%, 93%, 96%, 97%, 98. % Or 99% or more are adipose tissue-derived mesenchymal stem cells, umbilical cord-derived mesenchymal stem cells, and bone marrow-derived mesenchymal stem cells.

本発明における間葉系幹細胞は、HGF, SHH, OLIG2, VEGFA, NEUROG1, GRPR, IL1R1, CRHR2, CCKAR, APOE, PAX3, PAX5, EGF, CXCL1, GDNF, NRCAM, DLL1, HEYL, BMP2, NTN1, ASCL1, NRP2のいずれか一つ、または二つ以上を高発現することに加えて、例えば、成長特徴(例えば、継代から老化までの集団倍加能力、倍加時間)、核型分析(例えば、正常な核型、母体系統又は新生児系統)、フローサイトメトリー(例えば、FACS分析)による表面マーカー発現、免疫組織化学及び/又は免疫細胞化学(例えば、エピトープ検出)、遺伝子発現プロファイリング(例えば、遺伝子チップアレイ;逆転写PCR、リアルタイムPCR、従来型PCR等のポリメラーゼ連鎖反応)、miRNA発現プロファイリング、タンパク質アレイ、サイトカイン等のタンパク質分泌(例えば、血漿凝固解析、ELISA、サイトカインアレイ)、代謝産物(メタボローム解析)、本分野で知られている他の方法等によって、特徴付けられてもよい。 The mesenchymal stem cells in the present invention are HGF, SHH, OLIG2, VEGFA, NEUROG1, GRPR, IL1R1, CRHR2, CCKAR, APOE, PAX3, PAX5, EGF, CXCL1, GDNF, NRCAM, DLL1, HEYL, BMP2, NTN1, ASCL1. , In addition to high expression of any one or more of NRP2, for example, growth characteristics (eg, population doubling ability from passage to aging, doubling time), nuclear type analysis (eg, normal) Nuclear type, maternal or neonatal lineage, surface marker expression by flow cytometry (eg, FACS analysis), immunohistochemistry and / or immunocytokine (eg, epitope detection), gene expression profiling (eg, gene chip array; Reverse transcription PCR, real-time PCR, polymerase chain reaction such as conventional PCR), miRNA expression profiling, protein array, protein secretion such as cytokine (eg, plasma coagulation analysis, ELISA, cytokine array), metabolite (metabolome analysis), book It may be characterized by other methods known in the art and the like.

(間葉系幹細胞の調製方法)
HGF, SHH, OLIG2, VEGFA, NEUROG1, GRPR, IL1R1, CRHR2, CCKAR, APOE, PAX3, PAX5, EGF, CXCL1, GDNF, NRCAM, DLL1, HEYL, BMP2, NTN1, ASCL1, NRP2のいずれか一つ、または二つ以上が高発現の間葉系幹細胞の調製方法は特に限定されないが、例えば以下のようにして調製することができる。すなわち、脂肪、臍帯、骨髄等の組織から、当業者に公知の方法に従って、間葉系幹細胞を分離、培養し、HGF, SHH, OLIG2, VEGFA, NEUROG1, GRPR, IL1R1, CRHR2, CCKAR, APOE, PAX3, PAX5, EGF, CXCL1, GDNF, NRCAM, DLL1, HEYL, BMP2, NTN1, ASCL1もしくはNRP2に特異的に結合する抗体を用いて、HGF, SHH, OLIG2, VEGFA, NEUROG1, GRPR, IL1R1, CRHR2, CCKAR, APOE, PAX3, PAX5, EGF, CXCL1, GDNF, NRCAM, DLL1, HEYL, BMP2, NTN1, ASCL1もしくはNRP2高発現細胞をセルソーター、磁気ビーズ等で分離することにより取得することができる。また、特定の培地を用いた培養により、間葉系幹細胞におけるHGF, SHH, OLIG2, VEGFA, NEUROG1, GRPR, IL1R1, CRHR2, CCKAR, APOE, PAX3, PAX5, EGF, CXCL1, GDNF, NRCAM, DLL1, HEYL, BMP2, NTN1, ASCL1もしくはNRP2の発現を誘導することで、HGF, SHH, OLIG2, VEGFA, NEUROG1, GRPR, IL1R1, CRHR2, CCKAR, APOE, PAX3, PAX5, EGF, CXCL1, GDNF, NRCAM, DLL1, HEYL, BMP2, NTN1, ASCL1もしくはNRP2高発現の間葉系幹細胞を取得することもできる。この誘導によって得られる細胞集団において、細胞集団の50%以上がHGF, SHH, OLIG2, VEGFA, NEUROG1, GRPR, IL1R1, CRHR2, CCKAR, APOE, PAX3, PAX5, EGF, CXCL1, GDNF, NRCAM, DLL1, HEYL, BMP2, NTN1, ASCL1もしくはNRP2が高発現であることが好ましく、70%以上がHGF, SHH, OLIG2, VEGFA, NEUROG1, GRPR, IL1R1, CRHR2, CCKAR, APOE, PAX3, PAX5, EGF, CXCL1, GDNF, NRCAM, DLL1, HEYL, BMP2, NTN1, ASCL1もしくはNRP2が高発現であることがより好ましく、80%以上がHGF, SHH, OLIG2, VEGFA, NEUROG1, GRPR, IL1R1, CRHR2, CCKAR, APOE, PAX3, PAX5, EGF, CXCL1, GDNF, NRCAM, DLL1, HEYL, BMP2, NTN1, ASCL1もしくはNRP2を高発現であることがさらに好ましく、90%以上がHGF, SHH, OLIG2, VEGFA, NEUROG1, GRPR, IL1R1, CRHR2, CCKAR, APOE, PAX3, PAX5, EGF, CXCL1, GDNF, NRCAM, DLL1, HEYL, BMP2, NTN1, ASCL1もしくはNRP2を高発現であることが特に好ましく、実質的にHGF, SHH, OLIG2, VEGFA, NEUROG1, GRPR, IL1R1, CRHR2, CCKAR, APOE, PAX3, PAX5, EGF, CXCL1, GDNF, NRCAM, DLL1, HEYL, BMP2, NTN1, ASCL1もしくはNRP2の高発現の均一な細胞集団であることが最も好ましい。以下に、HGF, SHH, OLIG2, VEGFA, NEUROG1, GRPR, IL1R1, CRHR2, CCKAR, APOE, PAX3, PAX5, EGF, CXCL1, GDNF, NRCAM, DLL1, HEYL, BMP2, NTN1, ASCL1もしくはNRP2高発現の間葉系幹細胞の調製方法を具体的に説明する。
(Method of preparing mesenchymal stem cells)
HGF, SHH, OLIG2, VEGFA, NEUROG1, GRPR, IL1R1, CRHR2, CCKAR, APOE, PAX3, PAX5, EGF, CXCL1, GDNF, NRCAM, DLL1, HEYL, BMP2, NTN1, ASCL1, NRP2, or The method for preparing mesenchymal stem cells in which two or more are highly expressed is not particularly limited, but can be prepared, for example, as follows. That is, mesenchymal stem cells are separated and cultured from tissues such as fat, umbilical cord, and bone marrow according to a method known to those skilled in the art, and HGF, SHH, OLIG2, VEGFA, NEUROG1, GRPR, IL1R1, CRHR2, CCKAR, APOE, HGF, SHH, OLIG2, VEGFA, NEUROG1, GRPR, IL1R1, CRHR2, using an antibody that specifically binds to PAX3, PAX5, EGF, CXCL1, GDNF, NRCAM, DLL1, HEYL, BMP2, NTN1, ASCL1 or NRP2. It can be obtained by separating CCKAR, APOE, PAX3, PAX5, EGF, CXCL1, GDNF, NRCAM, DLL1, HEYL, BMP2, NTN1, ASCL1 or NRP2 high-expressing cells with a cell sorter, magnetic beads, or the like. In addition, HGF, SHH, OLIG2, VEGFA, NEUROG1, GRPR, IL1R1, CRHR2, CCKAR, APOE, PAX3, PAX5, EGF, CXCL1, GDNF, NRCAM, DLL1, in mesenchymal stem cells by culturing using a specific medium. By inducing the expression of HEYL, BMP2, NTN1, ASCL1 or NRP2, HGF, SHH, OLIG2, VEGFA, NEUROG1, GRPR, IL1R1, CRHR2, CCKAR, APOE, PAX3, PAX5, EGF, CXCL1, GDNF, NRCAM, DLL1 , HEYL, BMP2, NTN1, ASCL1 or NRP2 high expression mesenchymal stem cells can also be obtained. In the cell population obtained by this induction, more than 50% of the cell population is HGF, SHH, OLIG2, VEGFA, NEUROG1, GRPR, IL1R1, CRHR2, CCKAR, APOE, PAX3, PAX5, EGF, CXCL1, GDNF, NRCAM, DLL1, High expression of HEYL, BMP2, NTN1, ASCL1 or NRP2 is preferred, with 70% or more HGF, SHH, OLIG2, VEGFA, NEUROG1, GRPR, IL1R1, CRHR2, CCKAR, APOE, PAX3, PAX5, EGF, CXCL1, High expression of GDNF, NRCAM, DLL1, HEYL, BMP2, NTN1, ASCL1 or NRP2 is more preferable, and 80% or more are HGF, SHH, OLIG2, VEGFA, NEUROG1, GRPR, IL1R1, CRHR2, CCKAR, APOE, PAX3. , PAX5, EGF, CXCL1, GDNF, NRCAM, DLL1, HEYL, BMP2, NTN1, ASCL1 or NRP2 are more preferably highly expressed, with 90% or more being HGF, SHH, OLIG2, VEGFA, NEUROG1, GRPR, IL1R1, High expression of CRHR2, CCKAR, APOE, PAX3, PAX5, EGF, CXCL1, GDNF, NRCAM, DLL1, HEYL, BMP2, NTN1, ASCL1 or NRP2 is particularly preferred, substantially HGF, SHH, OLIG2, VEGFA, Most preferably, it is a homogeneous cell population with high expression of NEUROG1, GRPR, IL1R1, CRHR2, CCKAR, APOE, PAX3, PAX5, EGF, CXCL1, GDNF, NRCAM, DLL1, HEYL, BMP2, NTN1, ASCL1 or NRP2. Below, between HGF, SHH, OLIG2, VEGFA, NEUROG1, GRPR, IL1R1, CRHR2, CCKAR, APOE, PAX3, PAX5, EGF, CXCL1, GDNF, NRCAM, DLL1, HEYL, BMP2, NTN1, ASCL1 or NRP2 high expression The method for preparing leaf stem cells will be specifically described.

間葉系幹細胞は、当業者に周知の方法により調製することができる。以下に、例として、臍帯組織由来間葉系幹細胞及び脂肪組織由来間葉系幹細胞の調製方法を説明する。 Mesenchymal stem cells can be prepared by methods well known to those skilled in the art. Below, as an example, a method for preparing mesenchymal stem cells derived from umbilical cord tissue and mesenchymal stem cells derived from adipose tissue will be described.

臍帯は、経膣分娩および帝王切開にて娩出された胎盤および臍帯を含む産褥組織から適宜胎盤を取り除き回収することができる。回収した臍帯から臍帯血を除去した後、無菌または制菌処理を行っても良い。臍帯血の除去は、ヘパリン含有溶液などの抗凝固溶液ですすぐことによって行われる。無菌または制菌処理は、特に限定されるものではないが、ポピドンヨードの塗布、またはペニシリン、ストレプトマイシン、アムホテリシンB、ゲンタマイシン、およびナイスタチンなどの1種類以上の抗生剤および/または抗真菌剤を添加した培地またはバッファー中に浸漬してもよい。また、必要に応じて、赤血球を選択的に溶解する工程を含んでも良い。赤血球を選択的に溶解する方法として、例えば、塩化アンモニウムによる溶解による高張培地または低張培地中でのインキュベーションなど、当技術分野で周知の方法を使用することができる。 The umbilical cord can be recovered by appropriately removing the placenta from the placenta delivered by vaginal delivery and cesarean section and the puerperal tissue including the umbilical cord. After removing the umbilical cord blood from the collected umbilical cord, sterile or bacteriostatic treatment may be performed. Cord blood removal is performed by rinsing with an anticoagulant solution such as a heparin-containing solution. Aseptic or bacteriostatic treatment is not particularly limited, but is a medium supplemented with povidone iodine or one or more antibiotics and / or antifungal agents such as penicillin, streptomycin, amphotericin B, gentamicin, and nystatin. Alternatively, it may be immersed in a buffer. Further, if necessary, a step of selectively lysing red blood cells may be included. As a method for selectively lysing erythrocytes, a method well known in the art can be used, for example, incubation in a hypertonic medium or a hypotonic medium by lysis with ammonium chloride.

本発明の臍帯由来細胞とは、臍帯を原材料として、調製された細胞集団を意味し、公知の製造方法によって得られれば良く、例えば、以下の工程(i)〜(iii)を含む方法で製造することができる:
(i)臍帯を切断する工程;
(ii)(i)の工程で得られた臍帯を培養する工程;ならびに
(iii)継代する工程。
The umbilical cord-derived cell of the present invention means a cell population prepared using the umbilical cord as a raw material, and may be obtained by a known production method. For example, the cell is produced by a method including the following steps (i) to (iii). can do:
(I) Step of cutting the umbilical cord;
(Ii) The step of culturing the umbilical cord obtained in step (i); and (iii) the step of subculturing.

また、他の当該細胞の調製方法として、(i)臍帯を切断する工程の代わりに、(i')臍帯を酵素処理することにより組織を解離させる工程を含んでもよい。さらに、(i)臍帯を切断する工程に加えて、(i')臍帯を酵素処理することにより組織を解離させる工程を含んでもよい。 Further, as another method for preparing the cells, instead of (i) the step of cutting the umbilical cord, (i') a step of dissociating the tissue by enzymatically treating the umbilical cord may be included. Further, in addition to the step of (i) cutting the umbilical cord, a step of (i') dissociating the tissue by enzymatically treating the umbilical cord may be included.

本発明の(i)臍帯を切断する工程では、上述の方法で入手した臍帯を、羊膜、血管、血管周囲組織およびワルトンジェリーを含む状態にて機械力(細断力または剪断力)によって切断することによって行い得る。特に限定されないが、切断により得られた臍帯切片は、1から10mm、1から5mm、1から4mm、1から3mmまたは1から2mmの大きさが例示される。本発明の(i')臍帯を酵素処理することにより組織を解離させる工程では、上述の方法で入手した臍帯を、羊膜、血管、血管周囲組織およびワルトンジェリーを含む状態にて酵素処理にて、組織を解離させる工程にて行い得る。特に限定されないが、酵素処理には、コラゲナーゼ、ディスパーゼ及びヒアルロニダーゼなどの1種又は2種以上の酵素を用いた酵素処理が例示される。In the step (i) of the present invention for cutting the umbilical cord, the umbilical cord obtained by the above method is cut by a mechanical force (shred force or shear force) including the amniotic membrane, blood vessels, perivascular tissue and Walton jelly. Can be done by Although not particularly limited, the umbilical cord section obtained by cutting is exemplified in the size of 1 to 10 mm 3 , 1 to 5 mm 3 , 1 to 4 mm 3 , 1 to 3 mm 3 or 1 to 2 mm 3. In the step of dissociating the tissue by enzymatically treating the (i') umbilical cord of the present invention, the umbilical cord obtained by the above method is enzymatically treated with the amniotic membrane, blood vessels, perivascular tissue and Walton jelly. This can be done in the step of dissociating the tissue. The enzyme treatment is not particularly limited, and examples thereof include enzyme treatment using one or more kinds of enzymes such as collagenase, dispase and hyaluronidase.

本発明の(ii)(i)の工程で得られた臍帯を培養する工程は、固体表面上で、適切な細胞培地を使用して、(i)の工程で得られた臍帯を適切な細胞密度及び培養条件で培養する。 In the step of culturing the umbilical cord obtained in the steps (ii) and (i) of the present invention, the umbilical cord obtained in the step (i) is used as an appropriate cell on a solid surface using an appropriate cell medium. Incubate under density and culture conditions.

本工程で用いる培地は、間葉系幹細胞を培養できる培地であれば、特に限定されないが、このような培地は、基礎培地に、血清を添加する、及び/又は、アルブミン、トランスフェリン、脂肪酸、インスリン、亜セレン酸ナトリウム、コレステロール、コラーゲン前駆体、微量元素、2−メルカプトエタノール、3’−チオールグリセロール等の1つ以上の血清代替物を添加して作製してもよい。これらの培地には、必要に応じて、さらに脂質、アミノ酸、タンパク質、多糖、ビタミン、増殖因子、低分子化合物、抗生物質、抗酸化剤、ピルビン酸、緩衝剤、無機塩類等の物質を添加してもよい。 The medium used in this step is not particularly limited as long as it can culture mesenchymal stem cells, but such a medium is obtained by adding serum to the basal medium and / or albumin, transferrin, fatty acid, insulin. , Sodium selenate, cholesterol, collagen precursors, trace elements, 2-mercaptoethanol, 3'-thiolglycerol and the like may be added with one or more serum substitutes. If necessary, substances such as lipids, amino acids, proteins, polysaccharides, vitamins, growth factors, low molecular weight compounds, antibiotics, antioxidants, pyruvate, buffers, and inorganic salts are added to these media. You may.

上記基礎培地としては、例えば、IMDM培地、Medium 199培地、Eagle's Minimum Essential Medium(EMEM)培地、MEM-α培地、Dulbecco's modified Eagle's Medium(DMEM)培地、Ham's F12培地、RPMI 1640培地、Fischer's培地、MCDB201培地及びこれらの混合培地等が挙げられる。 Examples of the basal medium include IMDM medium, Medium 199 medium, Eagle's Minimum Essential Medium (EMEM) medium, MEM-α medium, Dulbecco's modified Eagle's Medium (DMEM) medium, Ham's F12 medium, RPMI 1640 medium, Fischer's medium, and MCDB201. Examples thereof include a medium and a mixed medium thereof.

上記血清としては、例えば、ヒト血清、ウシ胎児血清(FBS)、ウシ血清、仔ウシ血清、ヤギ血清、ウマ血清、ブタ血清、ヒツジ血清、ウサギ血清、ラット血清等が挙げられるがこれらに限定されない。血清を用いる場合、基礎培地に対して、5v/v%から15v/v%、好ましくは、10v/v%を添加してもよい。 Examples of the serum include, but are not limited to, human serum, fetal bovine serum (FBS), bovine serum, calf serum, goat serum, horse serum, pig serum, sheep serum, rabbit serum, rat serum and the like. .. When serum is used, 5 v / v% to 15 v / v%, preferably 10 v / v% may be added to the basal medium.

上記脂肪酸としては、リノール酸、オレイン酸、リノレイン酸、アラキドン酸、ミリスチン酸、パルミトイル酸、パルミチン酸、及びステアリン酸等が例示されるが、これらに限定されない。脂質は、フォスファチジルセリン、フォスファチジルエタノールアミン、フォスファチジルコリン等が例示されるが、これらに限定されない。アミノ酸は、例えば、L−アラニン、L−アルギニン、L−アスパラギン酸、L−アスパラギン、L−システイン、L−シスチン、L−グルタミン酸、L−グルタミン、L−グリシンなどを含むが、これらに限定されない。タンパク質は、例えば、エコチン、還元型グルタチオン、フィブロネクチン及びβ2−ミクログロブリン等が例示されるが、これらに限定されない。多糖は、グリコサミノグリカンが例示され、グリコサミノグリカンのうち特に、ヒアルロン酸、ヘパラン硫酸等が例示されるが、これらに限定されない。増殖因子は、例えば、血小板由来増殖因子(PDGF)、塩基性線維芽細胞成長因子(bFGF)、トランスフォーミング増殖因子ベータ(TGF-β)、肝細胞増殖因子(HGF)、上皮成長因子(EGF)、結合組織増殖因子(CTGF)、血管内皮細胞増殖因子(VEGF)等が例示されるが、これらに限定されない。本発明において得られる臍帯由来間葉系幹細胞を細胞移植に用いるという観点から、血清等の異種由来成分を含まない(ゼノフリー)培地を用いることが好ましい。このような培地は、例えば、PromoCell社、Lonza社、Biological Industries社、Veritas社、R&D Systems社、Corning社及びRohto社などから間葉系幹細胞(間質細胞)用として予め調製された培地として提供されている。 Examples of the fatty acid include, but are not limited to, linoleic acid, oleic acid, linoleic acid, arachidonic acid, myristic acid, palmitoyl acid, palmitic acid, stearic acid and the like. Examples of the lipid include, but are not limited to, phosphatidylserine, phosphatidylethanolamine, and phosphatidylcholine. Amino acids include, but are not limited to, for example, L-alanine, L-arginine, L-aspartic acid, L-asparagine, L-cysteine, L-cystine, L-glutamic acid, L-glutamine, L-glycine and the like. .. Examples of the protein include, but are not limited to, ecotin, reduced glutathione, fibronectin, β2-microglobulin and the like. Examples of the polysaccharide include glycosaminoglycans, and among glycosaminoglycans, hyaluronic acid, heparan sulfate and the like are exemplified, but the polysaccharides are not limited thereto. Growth factors include, for example, platelet-derived growth factor (PDGF), basic fibroblast growth factor (bFGF), transforming growth factor beta (TGF-β), hepatocellular growth factor (HGF), epithelial growth factor (EGF). , Binding tissue growth factor (CTGF), vascular endothelial cell growth factor (VEGF) and the like, but are not limited thereto. From the viewpoint of using the umbilical cord-derived mesenchymal stem cells obtained in the present invention for cell transplantation, it is preferable to use a medium containing no heterologous components such as serum (Zenofree). Such a medium is provided as a medium prepared in advance for mesenchymal stem cells (stromal cells) from, for example, PromoCell, Lonza, Biological Industries, Veritas, R & D Systems, Corning, Rohto and the like. Has been done.

本発明において、「固体表面」とは、本発明における脂肪組織由来間葉系幹細胞の結合・接着を可能とする任意の材料を意味する。特定の態様では、このような材料は、その表面への哺乳類細胞の結合・接着を促すように処理されたプラスチック材料である。固体表面を有する培養容器の形状は特に限定されないが、シャーレやフラスコなどが好適に用いられる。非結合状態の細胞及び細胞の破片を除去するために、インキュベーション後に細胞を洗浄する。 In the present invention, the "solid surface" means any material that enables the binding and adhesion of adipose tissue-derived mesenchymal stem cells in the present invention. In certain embodiments, such a material is a plastic material that has been treated to facilitate the binding and adhesion of mammalian cells to its surface. The shape of the culture vessel having a solid surface is not particularly limited, but a petri dish, a flask, or the like is preferably used. The cells are washed after incubation to remove unbound cells and cell debris.

本発明では、最終的に固体表面に結合・接着した状態で留まる細胞を、臍帯組織由来間葉系幹細胞の細胞集団として選択することができる。 In the present invention, cells that finally remain bound and adhered to the solid surface can be selected as a cell population of umbilical cord tissue-derived mesenchymal stem cells.

本発明の臍帯組織由来間葉系幹細胞は、浮遊培養製造法を用いても、製造することができる。浮遊培養製造法として、細胞を凝集させてスフェア上の細胞塊として撹拌培養する方法、マイクロキャリア上に細胞を接着させてマイクロキャリアを撹拌することにより培養する方法などがある。なお、撹拌は容器内の撹拌翼をスターラーで回転させる方法、培養液と細胞の入ったバッグを振盪機に乗せてバッグごと揺らすことで培養液を懸濁する方法などがある。また、浮遊培養製造法で用いる培地は、間葉系幹細胞を培養できる培地であれば、特に限定されず、上記のような培地が例示される。マイクロキャリアとしては、浮遊培養で用いることができるものであれば、特に限定されないが、ポリエステル、ポリスチレン、ガラス、デキストラン等が例示される。 The mesenchymal stem cells derived from the umbilical cord tissue of the present invention can also be produced by using the suspension culture production method. As a suspension culture production method, there are a method of aggregating cells and stirring and culturing them as a cell mass on a sphere, a method of adhering cells on microcarriers and culturing by stirring the microcarriers, and the like. For stirring, there are a method of rotating a stirring blade in a container with a stirrer, a method of placing a bag containing a culture solution and cells on a shaker, and shaking the bag together to suspend the culture solution. The medium used in the suspension culture production method is not particularly limited as long as it is a medium capable of culturing mesenchymal stem cells, and the above-mentioned medium is exemplified. The microcarrier is not particularly limited as long as it can be used in suspension culture, and examples thereof include polyester, polystyrene, glass, and dextran.

脂肪組織由来間葉系幹細胞は、例えば米国特許第6,777,231号に記載の製造方法によって得られれば良く、例えば、以下の工程(i)〜(iii)を含む方法で製造することができる:
(i) 脂肪組織を酵素による消化により細胞懸濁物を得る工程;
(ii) 細胞を沈降させ、細胞を適切な培地に再懸濁する工程;ならびに
(iii) 細胞を固体表面で培養し、固体表面への結合を示さない細胞を除去する工程。
The adipose tissue-derived mesenchymal stem cells may be obtained, for example, by the production method described in US Pat. No. 6,777,231, and can be produced, for example, by a method including the following steps (i) to (iii). it can:
(I) Step of obtaining cell suspension by enzymatic digestion of adipose tissue;
(Ii) The steps of precipitating cells and resuspending the cells in a suitable medium; and (iii) culturing the cells on a solid surface and removing cells that do not show binding to the solid surface.

工程(i)において用いる脂肪組織は、洗浄されたものを用いることが好ましい。洗浄は、生理学的に適合する生理食塩水溶液(例えばリン酸緩衝食塩水(PBS))を用いて、激しく攪拌して沈降させることによって行い得る。これは、脂肪組織に含まれる夾雑物(デブリとも言い、例えば損傷組織、血液、赤血球など)を組織から除去するためである。したがって、洗浄及び沈降は一般に、上清からデブリが総体的に除去されるまで繰り返される。残存する細胞は、さまざまなサイズの塊として存在するので、細胞そのものの損傷を最小限に抑えながら解離させるため、洗浄後の細胞塊を、細胞間結合を弱めるか、又は破壊する酵素(例えば、コラゲナーゼ、ディスパーゼ又はトリプシンなど)で処理することが好ましい。このような酵素の量及び処理期間は、使用される条件に依存して変わるが、当技術分野で既知である。このような酵素処理に代えて、又は併用して、細胞塊を、機械的な攪拌、超音波エネルギー、熱エネルギーなどの他の処理法で分解することができるが、細胞の損傷を最小限に抑えるため、酵素処理のみで行うことが好ましい。酵素を用いた場合、細胞に対する有害な作用を最小限に抑えるために、適切な期間をおいた後に培地等を用いて酵素を失活させることが望ましい。 As the adipose tissue used in the step (i), it is preferable to use a washed adipose tissue. Washing can be performed by vigorous stirring and precipitation with a physiologically compatible aqueous saline solution (eg, phosphate buffered saline (PBS)). This is to remove impurities (also called debris, for example, damaged tissue, blood, red blood cells, etc.) contained in adipose tissue from the tissue. Therefore, washing and sedimentation are generally repeated until debris is totally removed from the supernatant. Since the remaining cells exist as clumps of various sizes, enzymes that weaken or destroy intercellular connections in the washed cell clumps (eg, for example) in order to dissociate the cells while minimizing damage to the cells themselves. Treatment with collagenase, dispase, trypsin, etc.) is preferred. The amount and duration of such enzymes will vary depending on the conditions used and are known in the art. The cell mass can be degraded by other treatment methods such as mechanical agitation, ultrasonic energy, thermal energy, etc., in place of or in combination with such enzymatic treatment, but with minimal cell damage. In order to suppress it, it is preferable to carry out only the enzyme treatment. When an enzyme is used, it is desirable to inactivate the enzyme using a medium or the like after an appropriate period in order to minimize harmful effects on cells.

工程(i)により得られる細胞懸濁物は、凝集状の細胞のスラリー又は懸濁物、ならびに各種夾雑細胞、例えば赤血球、平滑筋細胞、内皮細胞、及び線維芽細胞を含む。従って、続いて凝集状態の細胞とこれらの夾雑細胞を分離、除去してもよいが、後述する工程(iii)での接着及び洗浄により、除去可能であることから、当該分離、除去は割愛してもよい。夾雑細胞を分離、除去する場合、細胞を上清と沈殿に強制的に分ける遠心分離によって達成しえる。得られた夾雑細胞を含む沈殿は、生理学的に適合する溶媒に懸濁させる。懸濁状の細胞には、赤血球を含む恐れがあるが、後述する個体表面への接着による選択により、赤血球は除外されるため、溶解する工程は必ずしも必要ではない。赤血球を選択的に溶解する方法として、例えば、塩化アンモニウムによる溶解による高張培地又は低張培地中でのインキュベーションなど、当技術分野で周知の方法を使用することができる。溶解後、例えば濾過、遠心沈降、又は密度分画によって溶解物を所望の細胞から分離してもよい。 The cell suspension obtained in step (i) includes a slurry or suspension of aggregated cells and various contaminant cells such as erythrocytes, smooth muscle cells, endothelial cells, and fibroblasts. Therefore, the aggregated cells and these contaminating cells may be subsequently separated and removed, but the separation and removal are omitted because they can be removed by adhesion and washing in the step (iii) described later. You may. Separation and removal of contaminating cells can be achieved by centrifugation, which forces the cells into a supernatant and a precipitate. The resulting precipitate containing contaminating cells is suspended in a physiologically compatible solvent. Suspended cells may contain erythrocytes, but the lysis step is not always necessary because erythrocytes are excluded by selection by adhesion to the solid surface, which will be described later. As a method for selectively lysing erythrocytes, a method well known in the art can be used, for example, incubation in a hypertonic medium or a hypotonic medium by lysis with ammonium chloride. After lysis, the lysate may be separated from the desired cells, for example by filtration, centrifugation, or density fractionation.

工程(ii)において、懸濁状の細胞において、間葉系幹細胞の純度を高めるために、1回もしくは連続して複数回洗浄し、遠心分離し、培地に再懸濁してもよい。この他にも、細胞を、細胞表面マーカープロファイルを基に、又は細胞のサイズ及び顆粒性を基に分離してもよい。 In step (ii), the suspended cells may be washed once or multiple times in succession, centrifuged, and resuspended in the medium in order to increase the purity of the mesenchymal stem cells. Alternatively, cells may be separated based on cell surface marker profiles or based on cell size and granularity.

再懸濁において用いる培地は、間葉系幹細胞を培養できる培地であれば、特に限定されないが、このような培地は、基礎培地に、血清を添加する、及び/又は、アルブミン、トランスフェリン、脂肪酸、インスリン、亜セレン酸ナトリウム、コレステロール、コラーゲン前駆体、微量元素、2−メルカプトエタノール、3’−チオールグリセロール等の1つ以上の血清代替物を添加して作製してもよい。これらの培地には、必要に応じて、さらに脂質、アミノ酸、タンパク質、多糖、ビタミン、増殖因子、低分子化合物、抗生物質、抗酸化剤、ピルビン酸、緩衝剤、無機塩類等の物質を添加してもよい。 The medium used for resuspension is not particularly limited as long as it can culture mesenchymal stem cells, but such a medium is a basal medium to which serum is added and / or albumin, transferrin, fatty acid, etc. It may be prepared by adding one or more serum substitutes such as insulin, sodium selenate, cholesterol, collagen precursor, trace element, 2-mercaptoethanol, 3'-thiolglycerol and the like. If necessary, substances such as lipids, amino acids, proteins, polysaccharides, vitamins, growth factors, low molecular weight compounds, antibiotics, antioxidants, pyruvate, buffers, and inorganic salts are added to these media. You may.

上記基礎培地としては、例えば、IMDM培地、Medium 199培地、Eagle's Minimum Essential Medium(EMEM)培地、MEM-α培地、Dulbecco's modified Eagle’s Medium(DMEM)培地、Ham's F12培地、RPMI 1640培地、Fischer's培地、MCDB201培地及びこれらの混合培地等が挙げられる。 Examples of the basal medium include IMDM medium, Medium 199 medium, Eagle's Minimum Essential Medium (EMEM) medium, MEM-α medium, Dulbecco's modified Eagle's Medium (DMEM) medium, Ham's F12 medium, RPMI 1640 medium, Fischer's medium, and MCDB201. Examples thereof include a medium and a mixed medium thereof.

上記血清としては、例えば、ヒト血清、ウシ胎児血清(FBS)、ウシ血清、仔ウシ血清、ヤギ血清、ウマ血清、ブタ血清、ヒツジ血清、ウサギ血清、ラット血清等が挙げられるがこれらに限定されない。血清を用いる場合、基礎培地に対して、5v/v%から15v/v%、好ましくは、10v/v%を添加してもよい。 Examples of the serum include, but are not limited to, human serum, fetal bovine serum (FBS), bovine serum, calf serum, goat serum, horse serum, pig serum, sheep serum, rabbit serum, rat serum and the like. .. When serum is used, 5 v / v% to 15 v / v%, preferably 10 v / v% may be added to the basal medium.

上記脂肪酸としては、リノール酸、オレイン酸、リノレイン酸、アラキドン酸、ミリスチン酸、パルミトイル酸、パルミチン酸、及びステアリン酸等が例示されるが、これらに限定されない。脂質は、フォスファチジルセリン、フォスファチジルエタノールアミン、フォスファチジルコリン等が例示されるが、これらに限定されない。アミノ酸は、例えば、L−アラニン、L−アルギニン、L−アスパラギン酸、L−アスパラギン、L−システイン、L−シスチン、L−グルタミン酸、L−グルタミン、L−グリシンなどを含むがこれらに限定されない。タンパク質は、例えば、エコチン、還元型グルタチオン、フィブロネクチン及びβ2−ミクログロブリン等が例示されるが、これらに限定されない。多糖は、グリコサミノグリカンが例示され、グリコサミノグリカンのうち特に、ヒアルロン酸、ヘパラン硫酸等が例示されるが、これらに限定されない。増殖因子は、例えば、血小板由来増殖因子(PDGF)、塩基性線維芽細胞成長因子(bFGF)、トランスフォーミング増殖因子ベータ(TGF-β)、肝細胞増殖因子(HGF)、上皮成長因子(EGF)、結合組織増殖因子(CTGF)、血管内皮細胞増殖因子(VEGF)等が例示されるが、これらに限定されない。本発明において得られる脂肪由来間葉系幹細胞を細胞移植に用いるという観点から、血清等の異種由来成分を含まない(ゼノフリー)培地を用いることが好ましい。このような培地は、例えば、PromoCell社、Lonza社、Biological Industries社、Veritas社、R&D Systems社、Corning社及びRohto社などから間葉系幹細胞(間質細胞)用として予め調製された培地として提供されている。 Examples of the fatty acid include, but are not limited to, linoleic acid, oleic acid, linoleic acid, arachidonic acid, myristic acid, palmitoyl acid, palmitic acid, stearic acid and the like. Examples of the lipid include, but are not limited to, phosphatidylserine, phosphatidylethanolamine, and phosphatidylcholine. Amino acids include, but are not limited to, for example, L-alanine, L-arginine, L-aspartic acid, L-asparagine, L-cysteine, L-cystine, L-glutamic acid, L-glutamine, L-glycine and the like. Examples of the protein include, but are not limited to, ecotin, reduced glutathione, fibronectin, β2-microglobulin and the like. Examples of the polysaccharide include glycosaminoglycans, and among glycosaminoglycans, hyaluronic acid, heparan sulfate and the like are exemplified, but the polysaccharides are not limited thereto. Growth factors include, for example, platelet-derived growth factor (PDGF), basic fibroblast growth factor (bFGF), transforming growth factor beta (TGF-β), hepatocellular growth factor (HGF), epithelial growth factor (EGF). , Binding tissue growth factor (CTGF), vascular endothelial cell growth factor (VEGF) and the like, but are not limited thereto. From the viewpoint of using the adipose-derived mesenchymal stem cells obtained in the present invention for cell transplantation, it is preferable to use a medium containing no heterologous components such as serum (Zenofree). Such a medium is provided as a medium prepared in advance for mesenchymal stem cells (stromal cells) from, for example, PromoCell, Lonza, Biological Industries, Veritas, R & D Systems, Corning, Rohto and the like. Has been done.

続いて、工程(iii)では、工程(ii)で得られた細胞懸濁液中の細胞を分化させずに固体表面上で、上述の適切な細胞培地を使用して、適切な細胞密度及び培養条件で培養する。本発明において、「固体表面」とは、本発明における脂肪組織由来間葉系幹細胞の結合・接着を可能とする任意の材料を意味する。特定の態様では、このような材料は、その表面への哺乳類細胞の結合・接着を促すように処理されたプラスチック材料である。固体表面を有する培養容器の形状は特に限定されないが、シャーレやフラスコなどが好適に用いられる。非結合状態の細胞及び細胞の破片を除去するために、インキュベーション後に細胞を洗浄する。 Subsequently, in step (iii), on a solid surface without differentiating the cells in the cell suspension obtained in step (ii), using the appropriate cell medium described above, the appropriate cell density and Incubate under culture conditions. In the present invention, the "solid surface" means any material that enables the binding and adhesion of adipose tissue-derived mesenchymal stem cells in the present invention. In certain embodiments, such a material is a plastic material that has been treated to facilitate the binding and adhesion of mammalian cells to its surface. The shape of the culture vessel having a solid surface is not particularly limited, but a petri dish, a flask, or the like is preferably used. The cells are washed after incubation to remove unbound cells and cell debris.

本発明では、最終的に固体表面に結合・接着した状態で留まる細胞を、脂肪組織由来間葉系幹細胞の細胞集団として選択することができる。 In the present invention, cells that finally remain bound and adhered to the solid surface can be selected as a cell population of adipose tissue-derived mesenchymal stem cells.

選択された細胞について、本発明における間葉系幹細胞であることを確認するために、表面抗原についてフローサイトメトリー等を用いて従来の方法で解析してもよい。さらに、各細胞系列に分化する能力について検査してもよく、このような分化は、従来の方法で行うことができる。 In order to confirm that the selected cells are mesenchymal stem cells in the present invention, the surface antigen may be analyzed by a conventional method using flow cytometry or the like. In addition, the ability to differentiate into each cell lineage may be tested, and such differentiation can be performed by conventional methods.

本発明における間葉系幹細胞は、上述の通り調製することができるが、次の特性を持つ細胞として定義してもよい;
(1)標準培地での培養条件で、プラスチックに接着性を示す、
(2)表面抗原CD73、CD90が陽性であり、CD45が陰性であり、及び
(3)培養条件にて骨細胞、脂肪細胞、軟骨細胞に分化可能。
The mesenchymal stem cells in the present invention can be prepared as described above, but may be defined as cells having the following characteristics;
(1) Shows adhesiveness to plastic under culture conditions in standard medium.
(2) Surface antigens CD73 and CD90 are positive, CD45 is negative, and (3) can be differentiated into osteoocytes, adipocytes, and chondrocytes under culture conditions.

上記工程によって得られた間葉系幹細胞から、HGF, SHH, OLIG2, VEGFA, NEUROG1, GRPR, IL1R1, CRHR2, CCKAR, APOE, PAX3, PAX5, EGF, CXCL1, GDNF, NRCAM, DLL1, HEYL, BMP2, NTN1, ASCL1もしくはNRP2タンパクを高発現している細胞を、セルソータ―、磁気ビーズ等を用いた免疫学的手法により選択的に分離することで、HGF, SHH, OLIG2, VEGFA, NEUROG1, GRPR, IL1R1, CRHR2, CCKAR, APOE, PAX3, PAX5, EGF, CXCL1, GDNF, NRCAM, DLL1, HEYL, BMP2, NTN1, ASCL1もしくはNRP2タンパクを高発現している間葉系幹細胞を取得することができる。またHGF, SHH, OLIG2, VEGFA, NEUROG1, GRPR, IL1R1, CRHR2, CCKAR, APOE, PAX3, PAX5, EGF, CXCL1, GDNF, NRCAM, DLL1, HEYL, BMP2, NTN1, ASCL1もしくはNRP2の発現を誘導できる特定の培地による培養を行うことにより、間葉系幹細胞におけるHGF, SHH, OLIG2, VEGFA, NEUROG1, GRPR, IL1R1, CRHR2, CCKAR, APOE, PAX3, PAX5, EGF, CXCL1, GDNF, NRCAM, DLL1, HEYL, BMP2, NTN1, ASCL1もしくはNRP2発現を誘導し、効率的にHGF, SHH, OLIG2, VEGFA, NEUROG1, GRPR, IL1R1, CRHR2, CCKAR, APOE, PAX3, PAX5, EGF, CXCL1, GDNF, NRCAM, DLL1, HEYL, BMP2, NTN1, ASCL1もしくはNRP2高発現の間葉系幹細胞を取得することもできる。一例として、セルソータ―を用いた免疫学的手法による選択的分離の具体的方法を以下に説明する。 From the mesenchymal stem cells obtained by the above steps, HGF, SHH, OLIG2, VEGFA, NEUROG1, GRPR, IL1R1, CRHR2, CCKAR, APOE, PAX3, PAX5, EGF, CXCL1, GDNF, NRCAM, DLL1, HEYL, BMP2, By selectively separating cells highly expressing NTN1, ASCL1 or NRP2 protein by immunological methods using cell sorters, magnetic beads, etc., HGF, SHH, OLIG2, VEGFA, NEUROG1, GRPR, IL1R1 , CRHR2, CCKAR, APOE, PAX3, PAX5, EGF, CXCL1, GDNF, NRCAM, DLL1, HEYL, BMP2, NTN1, ASCL1 or mesenchymal stem cells highly expressing NRP2 protein can be obtained. Identification that can induce the expression of HGF, SHH, OLIG2, VEGFA, NEUROG1, GRPR, IL1R1, CRHR2, CCKAR, APOE, PAX3, PAX5, EGF, CXCL1, GDNF, NRCAM, DLL1, HEYL, BMP2, NTN1, ASCL1 or NRP2 HGF, SHH, OLIG2, VEGFA, NEUROG1, GRPR, IL1R1, CRHR2, CCKAR, APOE, PAX3, PAX5, EGF, CXCL1, GDNF, NRCAM, DLL1, HEYL, Induces BMP2, NTN1, ASCL1 or NRP2 expression and efficiently HGF, SHH, OLIG2, VEGFA, NEUROG1, GRPR, IL1R1, CRHR2, CCKAR, APOE, PAX3, PAX5, EGF, CXCL1, GDNF, NRCAM, DLL1, HEYL , BMP2, NTN1, ASCL1 or NRP2 high expression mesenchymal stem cells can also be obtained. As an example, a specific method of selective separation by an immunological method using a cell sorter will be described below.

上記調製した間葉系幹細胞をトリプシン・EDTA溶液等により処理して得られた細胞懸濁液を遠心(室温、400G、5分)して上清を除去する。細胞にStaining Buffer(1%BSA−PBS)を加え、1×10cells/500μLとなるように調製し、ピペッティングにより細胞懸濁液濃度を均一にした後、新しい1.5mLマイクロチューブに50μLずつ分注する。分注した細胞懸濁液に1次抗体(Mouse anti human TFPI、Sekisui diagnostics社製、ADG4903)を5〜20μg/mLの濃度で添加し懸濁した後に、遮光・冷蔵下で30分間〜1時間反応させる。Staining Buffer 1mLで3回洗浄を行った後に、Staining Bufferを加え50μLとし、2次抗体(Anti Mouse IgG alexar488、Thermofisher scientific社製、A21202)を1〜10μg/mLの濃度で添加し懸濁した後に、遮光・冷蔵下で30分間〜1時間反応させる。Staining Buffer 1mLで3回洗浄を行った後に、PI Buffer(Staining buffer 14.4mLにPropidium iodide solution(SIGMA社製、P4864)28.8μLを添加して調製)300μLを加えてよく懸濁し、セルストレーナ付チューブに通し、fluorescence activated cell sorting(FACS)で分離を行うことができる。The mesenchymal stem cells prepared above are treated with a trypsin / EDTA solution or the like, and the cell suspension obtained is centrifuged (room temperature, 400 G, 5 minutes) to remove the supernatant. Staining Buffer (1% BSA-PBS) was added to the cells to prepare 1 × 10 6 cells / 500 μL, and the cell suspension concentration was made uniform by pipetting, and then 50 μL was placed in a new 1.5 mL microtube. Dispense one by one. A primary antibody (Mouse anti-human TFPI, manufactured by Sekisui diagnostics, ADG4903) was added to the dispensed cell suspension at a concentration of 5 to 20 μg / mL, suspended, and then kept in the dark and refrigerated for 30 minutes to 1 hour. React. After washing 3 times with 1 mL of Staining Buffer, add Staining Buffer to make 50 μL, add a secondary antibody (Anti Mouse IgG alexar488, Thermofisher scientific, A21202) at a concentration of 1 to 10 μg / mL and suspend. React for 30 minutes to 1 hour under shading and refrigeration. After washing 3 times with 1 mL of Staining Buffer, add 300 μL of PI Buffer (prepared by adding 28.8 μL of Propidium iodide solution (SIGMA, P4864) to 14.4 mL of Staining buffer), suspend well, and suspend well. Separation can be performed by fluorescence activated cell sorting (FACS) through a tube.

(間葉系幹細胞の凍結保存)
本発明における間葉系幹細胞は、疾患治療効果を備えていれば、適宜、凍結保存及び融解を繰り返した細胞であってもよい。本発明において、凍結保存は、当業者に周知の凍結保存液へ間葉系幹細胞を懸濁し、冷却することによって行い得る。懸濁は、必要に応じて細胞をトリプシンなどの剥離剤によって剥離し、凍結保存容器に移し、適宜、処理した後、凍結保存液を加えることによって行い得る。
(Cyropreservation of mesenchymal stem cells)
The mesenchymal stem cells in the present invention may be cells that have been cryopreserved and thawed as appropriate as long as they have a disease therapeutic effect. In the present invention, cryopreservation can be performed by suspending mesenchymal stem cells in a cryopreservation solution well known to those skilled in the art and cooling the cells. Suspension can be carried out by, if necessary, exfoliating the cells with a release agent such as trypsin, transferring to a cryopreservation container, appropriately treating the cells, and then adding a cryopreservation solution.

凍結保存液は、凍害防御剤として、DMSO(Dimethyl sulfoxide)を含有していてもよいが、DMSOは、細胞毒性に加えて、間葉系幹細胞を分化誘導する特性を有することから、DMSO含有量を減らすことが好ましい。DMSOの代替物として、グリセロール、プロピレングリコール又は多糖類が例示される。DMSOを用いる場合、5%〜20%の濃度、好ましくは5%〜10%の濃度、より好ましくは10%の濃度を含有する。この他にも、WO2007/058308に記載の添加剤を含んでもよい。このような凍結保存液として、例えば、バイオベルデ社、日本ジェネティクス株式会社、リプロセル社、ゼノアック社、コスモ・バイオ社、コージンバイオ株式会社、サーモフィッシャーサイエンティフィック社などから提供されている凍結保存液を用いてもよい。 The cryopreservation solution may contain DMSO (Dimethyl sulfoxide) as a frost damage protective agent, but DMSO has a property of inducing differentiation of mesenchymal stem cells in addition to cytotoxicity, and therefore has a DMSO content. It is preferable to reduce. Alternatives to DMSO are exemplified by glycerol, propylene glycol or polysaccharides. When DMSO is used, it contains a concentration of 5% to 20%, preferably a concentration of 5% to 10%, more preferably a concentration of 10%. In addition to this, the additives described in WO2007 / 058308 may be included. As such cryopreservation liquids, for example, cryopreservation provided by Bioverde, Japan Genetics Co., Ltd., Reprocell, Xenoac, Cosmo Bio, Kojin Bio Co., Ltd., Thermo Fisher Scientific Co., Ltd., etc. A liquid may be used.

上述の懸濁した細胞を凍結保存する場合、−80℃〜−100℃の間の温度(例えば、−80℃)で凍結することで良く、当該温度に達成しえる任意のフリーザーを用いて行い得る。特に限定されないが、急激な温度変化を回避するため、プログラムフリーザーを用いて、冷却速度を適宜制御してもよい。冷却速度は、凍結保存液の成分によって適宜選択しても良く、凍結保存液の製造者指示に従って行われ得る。 When the above-mentioned suspended cells are cryopreserved, they may be frozen at a temperature between −80 ° C. and −100 ° C. (for example, −80 ° C.), using an arbitrary freezer capable of achieving the temperature. obtain. Although not particularly limited, the cooling rate may be appropriately controlled by using a program freezer in order to avoid a sudden temperature change. The cooling rate may be appropriately selected depending on the components of the cryopreservation solution, and may be performed according to the manufacturer's instructions of the cryopreservation solution.

保存期間は、上記条件で凍結保存した細胞が融解した後、凍結前と同等の性質を保持している限り、特に上限は限定されないが、例えば、1週間以上、2週間以上、3週間以上、4週間以上、2か月以上、3か月以上、4か月以上、5か月以上、6か月以上、1年以上、又はそれ以上が挙げられる。より低い温度で保存することで細胞障害を抑制することができるため、液体窒素上の気相(約−150℃以下から−180℃以上)へ移して保存してもよい。液体窒素上の気相で保存する場合、当業者に周知の保存容器を用いて行うことができる。特に限定されないが、例えば、2週間以上保存する場合、液体窒素上の気相で保存することが好ましい。 The storage period is not particularly limited as long as the cells cryopreserved under the above conditions retain the same properties as before freezing after thawing, but for example, 1 week or more, 2 weeks or more, 3 weeks or more, 4 weeks or more, 2 months or more, 3 months or more, 4 months or more, 5 months or more, 6 months or more, 1 year or more, or more. Since cell damage can be suppressed by storing at a lower temperature, the cells may be transferred to a gas phase (from about −150 ° C. or lower to −180 ° C. or higher) on liquid nitrogen for storage. When storing in the gas phase on liquid nitrogen, it can be carried out using a storage container well known to those skilled in the art. Although not particularly limited, for example, when storing for 2 weeks or more, it is preferable to store in a gas phase on liquid nitrogen.

融解した間葉系幹細胞は、次の凍結保存までに適宜、培養してもよい。間葉系幹細胞の培養は、上述した間葉系幹細胞を培養できる培地を用いて行われ、特に限定されないが、約30〜40℃、好ましくは約37℃の培養温度で、CO含有空気の雰囲気下で行われてもよい。CO濃度は、約2〜10%、好ましくは約5〜10%である。培養において、培養容器に対して適切なコンフルエンシー(例えば、培養容器に対して、50%から80%を細胞が占有することが挙げられる)に達した後に、細胞をトリプシンなどの剥離剤によって剥離し、別途用意した培養容器に適切な細胞密度で播種して培養を継続してもよい。細胞を播種する際において、典型的な細胞密度として、100細胞/cm〜100,000細胞/cm、500細胞/cm〜50,000細胞/cm、1,000〜10,000細胞/cm、2,000〜10,000細胞/cmなどが例示される。特定の態様では、細胞密度は2,000〜10,000細胞/cmである。適切なコンフルエンシーに達するまでの期間が、3日間から7日間となるように調整することが好ましい。培養中、必要に応じて、適宜、培地を交換してもよい。The thawed mesenchymal stem cells may be appropriately cultured before the next cryopreservation. The culture of the mesenchymal stem cells is carried out using the medium capable of culturing the above-mentioned mesenchymal stem cells, and is not particularly limited, but at a culture temperature of about 30 to 40 ° C., preferably about 37 ° C., in CO 2- containing air. It may be done in an atmosphere. The CO 2 concentration is about 2-10%, preferably about 5-10%. In culturing, cells are detached with a release agent such as trypsin after reaching the appropriate confluency for the culture vessel (eg, 50% to 80% of the culture vessel is occupied by cells). Then, the cells may be seeded in a separately prepared culture vessel at an appropriate cell density to continue the culture. When seeding cells, typical cell densities are 100 cells / cm 2 to 100,000 cells / cm 2 , 500 cells / cm 2 to 50,000 cells / cm 2 , 1,000 to 10,000 cells. Examples include / cm 2 , 2,000 to 10,000 cells / cm 2 . In certain embodiments, the cell density is 2,000 to 10,000 cells / cm 2 . It is preferable to adjust the time to reach the appropriate confluency from 3 days to 7 days. During culturing, the medium may be changed as needed.

凍結保存した細胞の融解は、当業者に周知の方法によって行い得る。例えば、37℃の恒温槽内又は湯浴中にて静置又は振とうすることによって行う方法が例示される。 Thawing of cryopreserved cells can be performed by methods well known to those skilled in the art. For example, a method performed by standing or shaking in a constant temperature bath at 37 ° C. or in a hot water bath is exemplified.

本発明の間葉系幹細胞は、いずれの状態の細胞であってもよいが、例えば培養中の細胞を剥離して回収された細胞でもよいし、凍結保存液中に凍結された状態の細胞でもよい。拡大培養して得られる同ロットの細胞を小分けして凍結保存したものを使用すると、安定して同様の作用効果が得られる点、取扱い性に優れる点等において好ましい。凍結保存状態の間葉系幹細胞は、使用直前に融解し、凍結保存液に懸濁したまま輸液もしくは培地等の溶液に直接混合してもよい。また、遠心分離等の方法により凍結保存液を除去してから輸液もしくは培地等の溶液に懸濁してもよい。ここで、本発明における「輸液」とは、ヒトの治療の際に用いられる溶液のことをいい、特に限定されないが、例えば、生理食塩水、日局生理食塩液、5%ブドウ糖液、日局ブドウ糖注射液、リンゲル液、日局リンゲル液、乳酸リンゲル液、酢酸リンゲル液、1号液(開始液)、2号液(脱水補給液)、3号液(維持液)、4号液(術後回復液)等が挙げられる。 The mesenchymal stem cell of the present invention may be a cell in any state, for example, a cell recovered by exfoliating a cell in culture, or a cell frozen in a cryopreservation solution. Good. It is preferable to use cells obtained by expanding and culturing the same lot of cells in small portions and cryopreserving them, in that the same action and effect can be stably obtained and the handling property is excellent. The mesenchymal stem cells in the cryopreserved state may be thawed immediately before use and may be directly mixed with a solution such as an infusion solution or a medium while being suspended in the cryopreserved solution. Alternatively, the cryopreservation solution may be removed by a method such as centrifugation and then suspended in a solution such as an infusion solution or a medium. Here, the "infusion solution" in the present invention refers to a solution used in the treatment of humans, and is not particularly limited, but for example, a physiological saline solution, a Japanese Pharmacopoeia physiological saline solution, a 5% glucose solution, and a Japanese Pharmacopoeia. Glucose injection, Ringer's solution, Japanese Pharmacopoeia Ringer's solution, Lactobacillus Ringer's solution, Ringer's acetate solution, No. 1 (starting solution), No. 2 (dehydration replacement fluid), No. 3 (maintenance solution), No. 4 (postoperative recovery solution) And so on.

[神経障害治療剤]
本発明の神経障害治療剤は、上述した本発明の、HGF, SHH, OLIG2, VEGFA, NEUROG1, GRPR, IL1R1, CRHR2, CCKAR, APOE, PAX3, PAX5, EGF, CXCL1, GDNF, NRCAM, DLL1, HEYL, BMP2, NTN1, ASCL1, NRP2のいずれか一つ、または二つ以上を高発現する間葉系幹細胞を含有する。本発明の神経障害治療剤によると、神経障害を効果的に保護することができる。本発明の神経障害治療剤を含む間葉系幹細胞については、上記間葉系幹細胞の項の説明を適用できる。
[Treatment for neuropathy]
The therapeutic agent for neurological disorders of the present invention is the above-mentioned HGF, SHH, OLIG2, VEGFA, NEUROG1, GRPR, IL1R1, CRHR2, CCKAR, APOE, PAX3, PAX5, EGF, CXCL1, GDNF, NRCAM, DLL1, HEYL. , BMP2, NTN1, ASCL1, NRP2, contains mesenchymal stem cells that highly express any one or more. According to the neuropathy therapeutic agent of the present invention, neuropathy can be effectively protected. The above description of the section on mesenchymal stem cells can be applied to the mesenchymal stem cells containing the therapeutic agent for neuropathy of the present invention.

本発明の神経障害治療剤は、本発明の効果を損なわない範囲であれば、上記間葉系幹細胞以外に、その用途や形態に応じて、常法に従い、薬学的に許容される担体や添加物を含有させてもよい。このような担体や添加物としては、例えば、等張化剤、増粘剤、糖類、糖アルコール類、防腐剤(保存剤)、殺菌剤又は抗菌剤、pH調節剤、安定化剤、キレート剤、油性基剤、ゲル基剤、界面活性剤、懸濁化剤、結合剤、賦形剤、滑沢剤、崩壊剤、発泡剤、流動化剤、分散剤、乳化剤、緩衝剤、溶解補助剤、抗酸化剤、甘味剤、酸味剤、着色剤、呈味剤、香料又は清涼化剤等が挙げられるが、これらに限定されない。代表的な成分として例えば次の担体、添加物等が挙げられる。 In addition to the above-mentioned mesenchymal stem cells, the therapeutic agent for neuropathy of the present invention can be added with a pharmaceutically acceptable carrier or addition according to a conventional method according to its use and morphology, as long as the effects of the present invention are not impaired. It may contain an object. Examples of such carriers and additives include tonicity agents, thickeners, sugars, sugar alcohols, preservatives (preservatives), bactericides or antibacterial agents, pH regulators, stabilizers, and chelating agents. , Oil-based bases, gel bases, surfactants, suspending agents, binders, excipients, lubricants, disintegrants, foaming agents, fluidizers, dispersants, emulsifiers, buffers, solubilizers , Antioxidants, sweeteners, acidity agents, colorants, flavoring agents, fragrances, refreshing agents and the like, but are not limited thereto. Typical components include, for example, the following carriers and additives.

担体としては、例えば、水、含水エタノール等の水性担体が;等張化剤(無機塩)としては、例えば、塩化ナトリウム、塩化カリウム、塩化カルシウム、塩化マグネシウム等が;多価アルコールとしては、例えば、グリセリン、プロピレングリコール、ポリエチレングリコール等が;増粘剤としては、例えば、カルボキシビニルポリマー、ヒドロキシエチルセルロース、ヒドロキシプロピルメチルセルロース、メチルセルロース、アルギン酸、ポリビニルアルコール(完全、又は部分ケン化物)、ポリビニルピロリドン、マクロゴール等が;糖類としては、例えば、シクロデキストリン、ブドウ糖等が;糖アルコール類としては、例えば、キシリトール、ソルビトール、マンニトール等(これらはd体、l体又はdl体のいずれでもよい)が;防腐剤、殺菌剤又は抗菌剤としては、例えば、ジブチルヒドロキシトルエン、ブチルヒドロキシアニソール、塩酸アルキルジアミノエチルグリシン、安息香酸ナトリウム、エタノール、塩化ベンザルコニウム、塩化ベンゼトニウム、グルコン酸クロルヘキシジン、クロロブタノール、ソルビン酸、ソルビン酸カリウム、トロメタモール、デヒドロ酢酸ナトリウム、パラオキシ安息香酸メチル、パラオキシ安息香酸エチル、パラオキシ安息香酸プロピル、パラオキシ安息香酸ブチル、硫酸オキシキノリン、フェネチルアルコール、ベンジルアルコール、ビグアニド化合物(具体的には、塩酸ポリヘキサニド(ポリヘキサメチレンビグアニド)等)、グローキル(ローディア社製商品名)等が;pH調節剤としては、例えば、塩酸、ホウ酸、アミノエチルスルホン酸、イプシロン−アミノカプロン酸、クエン酸、酢酸、水酸化ナトリウム、水酸化カリウム、水酸化カルシウム、水酸化マグネシウム、炭酸水素ナトリウム、炭酸ナトリウム、ホウ砂、トリエタノールアミン、モノエタノールアミン、ジイソプロパノールアミン、硫酸、硫酸マグネシウム、リン酸、ポリリン酸、プロピオン酸、シュウ酸、グルコン酸、フマル酸、乳酸、酒石酸、リンゴ酸、コハク酸、グルコノラクトン、酢酸アンモニウム等が;安定化剤としては、例えば、ジブチルヒドロキシトルエン、トロメタモール、ナトリウムホルムアルデヒドスルホキシレート(ロンガリット)、トコフェロール、ピロ亜硫酸ナトリウム、モノエタノールアミン、モノステアリン酸アルミニウム、モノステアリン酸グリセリン、亜硫酸水素ナトリウム、亜硫酸ナトリウム等が;油性基剤としては、例えば、オリーブ油、トウモロコシ油、大豆油、ゴマ油、綿実油等の植物油、中鎖脂肪酸トリグリセリド等が;水性基剤としては、例えば、マクロゴール400等が;ゲル基剤としては、例えば、カルボキシビニルポリマー、ガム質等が;界面活性剤としては、例えば、ポリソルベート80、硬化ヒマシ油、グリセリン脂肪酸エステル、セスキオレイン酸ソルビタン等が;懸濁化剤としては、例えば、サラシミツロウや各種界面活性剤、アラビアゴム、アラビアゴム末、キサンタンガム、大豆レシチン等が;結合剤としては、例えば、ヒドロキシエチルセルロース、ヒドロキシプロピルセルロース、ヒドロキシプロピルメチルセルロース、カルボキシメチルセルロースナトリウム、ポリビニルピロリドン、ポリビニルアルコール等が;賦形剤としては、例えば、ショ糖、乳糖、デンプン、コーンスターチ、結晶セルロース、軽質無水ケイ酸等が;滑沢剤としては、例えば、ショ糖脂肪酸エステル、ステアリン酸マグネシウム、タルク等が;崩壊剤としては、例えば、低置換度ヒドロキシプロピルセルロース、クロスポビドン、クロスカルメロースナトリウム等が;発泡剤としては、例えば、炭酸水素ナトリウム等が;流動化剤としては、例えば、メタケイ酸アルミン酸ナトリウム、軽質無水ケイ酸等が、それぞれ挙げられる。 As the carrier, for example, an aqueous carrier such as water or hydrous ethanol; as the tonicity agent (inorganic salt), for example, sodium chloride, potassium chloride, calcium chloride, magnesium chloride, etc .; as the polyhydric alcohol, for example. , Glycerin, propylene glycol, polyethylene glycol, etc .; Examples of thickeners include carboxyvinyl polymer, hydroxyethyl cellulose, hydroxypropyl methyl cellulose, methyl cellulose, argic acid, polyvinyl alcohol (completely or partially saponified), polyvinylpyrrolidone, macrogol. Etc.; as sugars, for example, cyclodextrin, glucose, etc .; as sugar alcohols, for example, xylitol, sorbitol, mannitol, etc. (these may be d-form, l-form, or dl-form); preservatives. Examples of the bactericidal agent or antibacterial agent include dibutylhydroxytoluene, butylhydroxyanisole, alkyldiaminoethylglycine hydrochloride, sodium benzoate, ethanol, benzalconium chloride, benzethonium chloride, chlorhexidine gluconate, chlorobutanol, sorbic acid, and sorbin. Potassium acid, tromethamole, sodium dehydroacetate, methyl paraoxybenzoate, ethyl paraoxybenzoate, propyl paraoxybenzoate, butyl paraoxybenzoate, oxyquinoline sulfate, phenethyl alcohol, benzyl alcohol, biguanide compounds (specifically, polyhexanide hydrochloride (specifically, polyhexanide hydrochloride (specifically) Polyhexamethylene biguanide), etc.), Gloquil (trade name manufactured by Rhodia), etc .; Examples of pH adjusters include hydrochloric acid, boric acid, aminoethylsulfonic acid, epsilon-aminocaproic acid, citric acid, acetic acid, sodium hydroxide. , Potassium hydroxide, Calcium hydroxide, Magnesium hydroxide, Sodium hydrogen carbonate, Sodium carbonate, Boso, Triethanolamine, Monoethanolamine, Diisopropanolamine, Sulfate, Magnesium sulfate, Phosphoric acid, Polyphosphate, Propionic acid, Shu Acids, gluconic acid, fumaric acid, lactic acid, tartaric acid, malic acid, succinic acid, gluconolactone, ammonium acetate, etc .; Stabilizers include, for example, dibutylhydroxytoluene, tromethamole, sodium formaldehyde sulfoxylate (longalit), Tocopherol, sodium pyrosulfate, monoethanolamine, aluminum monostearate, glycerin monostearate, sub Sodium hydrogensulfate, sodium sulfite, etc .; oil-based bases include, for example, olive oil, corn oil, soybean oil, sesame oil, cottonseed oil and other vegetable oils, medium-chain fatty acid triglycerides, etc .; as aqueous bases, for example, Macrogol 400. Etc .; as gel base, for example, carboxyvinyl polymer, gum quality, etc .; as surfactant, for example, polysorbate 80, hardened castor oil, glycerin fatty acid ester, sorbitan sesquioleate, etc .; Examples include sala shimitsuro, various surfactants, arabic rubber, arabic rubber powder, xanthan gum, soybean lecithin, and the like; as binders, for example, hydroxyethyl cellulose, hydroxypropyl cellulose, hydroxypropyl methyl cellulose, sodium carboxymethyl cellulose, polyvinyl. Pyrrolidone, polyvinyl alcohol, etc .; as excipients, for example, sucrose, lactose, starch, corn starch, crystalline cellulose, light anhydrous silicic acid, etc .; as lubricants, for example, sucrose fatty acid ester, magnesium stearate, etc. , Tarc, etc .; as disintegrants, for example, low-substituted hydroxypropyl cellulose, crospovidone, croscarmellose sodium, etc .; as effervescent agents, for example, sodium hydrogen carbonate, etc .; as fluidizers, for example, Examples thereof include sodium aluminometasilicate and light anhydrous silicic acid.

本発明の神経障害治療剤は、目的に応じて種々の形態、例えば、固形剤、半固形剤、液剤等の様々な剤形で提供することができる。例えば、固形剤(錠剤、粉末、散剤、顆粒剤、カプセル剤等)、半固形剤[軟膏剤(硬軟膏剤、軟軟膏剤等)、クリーム剤等]、液剤[ローション剤、エキス剤、懸濁剤、乳剤、シロップ剤、注射剤(輸液剤、埋め込み注射剤、持続性注射、用時調製型の注射剤を含む)、透析用剤、エアゾール剤、軟カプセル剤、ドリンク剤等]、貼付剤、パップ剤等の形態で利用できる。また、本発明の神経障害治療剤は、油性又は水性のビヒクル中の溶液又は乳液等の形態でも利用できる。さらに、本発明の神経障害治療剤は噴霧により、患部に適用することもでき、本発明の神経障害治療剤は噴霧した後に患部でゲル化もしくはシート化される形態でも利用できる。本発明の神経障害患治療剤は上記間葉系幹細胞をシート状または立体構造体とした後に、患部に適用することもできる。 The therapeutic agent for neuropathy of the present invention can be provided in various forms, for example, various dosage forms such as solid preparations, semi-solid preparations, and liquid preparations, depending on the intended purpose. For example, solids (tablets, powders, powders, granules, capsules, etc.), semi-solids [ointments (hard ointments, ointments, etc.), creams, etc.], liquids [lotions, extracts, suspensions, etc.] Turbids, emulsions, syrups, injections (including infusions, implantable injections, continuous injections, time-prepared injections), dialysis agents, aerosols, soft capsules, drinks, etc.], patch It can be used in the form of an agent, a poultice, or the like. The neuropathy therapeutic agent of the present invention can also be used in the form of a solution or emulsion in an oily or aqueous vehicle. Furthermore, the therapeutic agent for neuropathy of the present invention can be applied to the affected area by spraying, and the therapeutic agent for neuropathy of the present invention can also be used in the form of gelling or sheeting in the affected area after spraying. The therapeutic agent for neuropathy of the present invention can also be applied to the affected area after forming the mesenchymal stem cells into a sheet or three-dimensional structure.

本発明の神経障害治療剤は、生理食塩水、日局生理食塩液、5%ブドウ糖液、日局ブドウ糖注射液、リンゲル液、日局リンゲル液、乳酸リンゲル液、酢酸リンゲル液、重炭酸リンゲル液、1号液(開始液)、2号液(脱水補給液)、3号液(維持液)、4号液(術後回復液)等の輸液、又は、DMEM等の細胞培養培地を用いて、懸濁もしくは希釈して用いることができ、好ましくは生理食塩液、5%ブドウ糖液、1号液(開始液)で、より好ましくは5%ブドウ糖液、1号液(開始液)で懸濁もしくは希釈して用いることができる。 The neuropathy therapeutic agent of the present invention includes physiological saline, Japanese Pharmacopoeia physiological saline, 5% glucose solution, Japanese Pharmacopoeia glucose injection, Ringer's solution, Japanese Pharmacopoeia Ringer's solution, Lactobacillus ringer's solution, Ringer's acetate solution, Ringer's carbonate solution No. 1 ( Suspension or dilution using infusion fluids such as starting fluid), fluid 2 (dehydration replacement fluid), fluid 3 (maintenance fluid), fluid 4 (postoperative recovery fluid), or cell culture medium such as DMEM. It can be used by suspending or diluting with physiological saline solution, 5% glucose solution, No. 1 solution (starting solution), and more preferably 5% glucose solution, No. 1 solution (starting solution). be able to.

本発明の神経障害治療剤が液剤である場合、神経障害治療剤のpHは、医薬上、薬理学的に(製薬上)又は生理学的に許容される範囲内であれば特に限定されるものではないが、一例として、2.5〜9.0、好ましくは3.0〜8.5、より好ましくは3.5〜8.0となる範囲が挙げられる。 When the therapeutic agent for neuropathy of the present invention is a liquid preparation, the pH of the therapeutic agent for neuropathy is not particularly limited as long as it is within a pharmaceutically, pharmacologically (pharmaceutically) or physiologically acceptable range. However, as an example, a range of 2.5 to 9.0, preferably 3.0 to 8.5, and more preferably 3.5 to 8.0 can be mentioned.

本発明の神経障害治療剤が液剤である場合、神経障害治療剤の浸透圧については、生体に許容される範囲内であれば、特に制限されない。本発明の組成物の浸透圧比の一例として、好ましくは0.7〜5.0、より好ましくは0.8〜3.0、さらに好ましくは0.9〜1.4となる範囲が挙げられる。浸透圧の調整は無機塩、多価アルコール、糖アルコール、糖類等を用いて、当該技術分野で既知の方法で行うことができる。浸透圧比は、第十五改正日本薬局方に基づき286mOsm(0.9w/v%塩化ナトリウム水溶液)の浸透圧に対する試料の浸透圧の比とし、浸透圧は日本薬局方記載の浸透圧測定法(氷点降下法)を参考にして測定する。なお、浸透圧比測定用標準液(0.9w/v%塩化ナトリウム水溶液)は、塩化ナトリウム(日本薬局方標準試薬)を500〜650℃で40〜50分間乾燥した後、デシケーター(シリカゲル)中で放冷し、その0.900gを正確に量り、精製水に溶かし正確に100mLとして調製するか、市販の浸透圧比測定用標準液(0.9w/v%塩化ナトリウム水溶液)を用いる。 When the therapeutic agent for neuropathy of the present invention is a liquid preparation, the osmotic pressure of the therapeutic agent for neuropathy is not particularly limited as long as it is within the range acceptable to the living body. As an example of the osmotic pressure ratio of the composition of the present invention, a range of preferably 0.7 to 5.0, more preferably 0.8 to 3.0, and further preferably 0.9 to 1.4 can be mentioned. The osmotic pressure can be adjusted by a method known in the art using inorganic salts, polyhydric alcohols, sugar alcohols, sugars and the like. The osmotic pressure ratio is the ratio of the osmotic pressure of the sample to the osmotic pressure of 286 mOsm (0.9 w / v% sodium chloride aqueous solution) based on the 15th revised Japanese Pharmacy, and the osmotic pressure is the osmotic pressure measurement method described in the Japanese Pharmacy. Measure with reference to the freezing point descent method). The standard solution for measuring the osmotic pressure ratio (0.9 w / v% sodium chloride aqueous solution) is prepared by drying sodium chloride (standard reagent of the Japanese Pharmacopoeia) at 500 to 650 ° C. for 40 to 50 minutes and then in a desiccator (silica). Allow to cool, weigh accurately 0.900 g, dissolve in purified water to prepare exactly 100 mL, or use a commercially available standard solution for measuring osmotic pressure ratio (0.9 w / v% sodium chloride aqueous solution).

本発明の神経障害治療剤の対象への投与経路は、経口投与、皮下投与、筋肉内投与、静脈内投与、動脈内投与、脳室内投与、髄腔内投与、腹腔内投与、舌下投与、経直腸投与、経腟投与、眼内投与、経鼻投与、吸入、経皮投与、インプラント、臓器表面への噴霧及びシート等の貼付による直接投与等が挙げられるが、本発明の神経障害治療剤の有効性の観点から、好ましくは動脈内投与、静脈内投与、脳室内投与及び髄腔内投与であり、対象者の負担の軽減の観点から、より好ましくは静脈内投与、筋肉内投与、鼻腔内投与であり、効果の観点からは脳室内投与及び髄腔内投与が好ましい。 The routes of administration of the therapeutic agent for neuropathy of the present invention to a subject include oral administration, subcutaneous administration, intramuscular administration, intravenous administration, intraarterial administration, intraventricular administration, intrathecal administration, intraperitoneal administration, and sublingual administration. Examples thereof include transrectal administration, transvaginal administration, intraocular administration, nasal administration, inhalation, transdermal administration, implants, spraying on the surface of organs, and direct administration by attaching a sheet or the like. From the viewpoint of efficacy, it is preferably intraarterial administration, intravenous administration, intraventricular administration and intrathecal administration, and from the viewpoint of reducing the burden on the subject, it is more preferably intravenous administration, intramuscular administration, and nasal administration. It is an internal administration, and from the viewpoint of effect, intraventricular administration and intramuscular administration are preferable.

本発明の神経障害治療剤において、その用量(投与量)は、患者の状態(体重、年齢、症状、体調等)、及び本発明の神経障害治療剤の剤形等によって異なりうるが、十分な経障害治療剤の治療効果を奏する観点からは、その量は多い方が好ましい傾向にあり、一方、副作用の発現を抑制する観点からはその量は少ない方が好ましい傾向にある。通常、成人に投与する場合には、細胞数として、1×10〜1×1012個/回、好ましくは1×10〜1×1011個/回、より好ましくは1×10〜1×1010個/回、さらに好ましくは5×10〜1×10個/回である。また、患者の体重あたりの投与量としては、1×10〜5×1010個/kg、好ましくは1×10〜5×10個/kg、より好ましくは1×10〜5×10個/kg、さらに好ましくは1×10〜5×10個/kgである。新生児に投与する場合には、細胞数として、1×10〜1×1011個/回、好ましくは1×10〜1×1010個/回、より好ましくは1×10〜1×10個/回、さらに好ましくは5×10〜5×10個/回である。また、患者の体重あたりの投与量としては、1×10〜5×1010個/kg、好ましくは1×10〜5×10個/kg、より好ましくは1×10〜5×10個/kg、さらに好ましくは1×10〜5×10個/kgである。なお、本用量を1回量として、複数回投与してもよく、本用量を複数回に分けて投与してもよい。The dose (dose) of the therapeutic agent for neuropathy of the present invention may vary depending on the patient's condition (weight, age, symptom, physical condition, etc.), the dosage form of the therapeutic agent for neuropathy of the present invention, etc., but is sufficient. From the viewpoint of exerting the therapeutic effect of the therapeutic agent for transdisorder, a large amount tends to be preferable, while a small amount tends to be preferable from the viewpoint of suppressing the occurrence of side effects. Usually, when administered to an adult, the number of cells is 1 × 10 3 to 1 × 10 12 cells / time, preferably 1 × 10 4 to 1 × 10 11 cells / time, more preferably 1 × 10 5 to 1 × 10 10 pieces / time, more preferably 5 × 10 6 to 1 × 10 9 pieces / time. The dose per patient's body weight is 1 x 10 to 5 x 10 10 pieces / kg, preferably 1 x 10 2 to 5 x 10 9 pieces / kg, and more preferably 1 x 10 3 to 5 x 10. 8 pieces / kg, more preferably 1 × 10 4 to 5 × 10 7 pieces / kg. When administered to a newborn baby, the number of cells is 1 × 10 3 to 1 × 10 11 cells / time, preferably 1 × 10 4 to 1 × 10 10 cells / time, more preferably 1 × 10 5 to 1 ×. 10 9 pieces / time, more preferably 5 × 10 5 to 5 × 10 8 pieces / time. The dose per patient's body weight is 1 x 10 to 5 x 10 10 pieces / kg, preferably 1 x 10 2 to 5 x 10 9 pieces / kg, and more preferably 1 x 10 3 to 5 x 10. 8 pieces / kg, more preferably 1 × 10 4 to 5 × 10 7 pieces / kg. In addition, this dose may be administered as a single dose in a plurality of times, or this dose may be administered in a plurality of times.

本発明の神経障害治療剤は、一又は二以上の他の薬剤と共に投与してもよい。他の薬剤としては、神経障害の治療薬として用いることができる任意の剤を薬剤が挙げられ、たとえば、レボドパ、アマンタジン、カルビドパ等の抗パーキンソン病薬、ブロモクリプチン、プルゴリド、ロピニロール、プラミペキソール等のドパミン作動薬、セレギリン、ラサリジン等の選択的B型モノアミン酸化酵素阻害剤(MAO-B)、エンタカポン、トルカポン等のカテコール−O−メチルトランスフェラーゼ(COMT)阻害薬ベンズトロポン、トリヘキシフェニジル等の抗コリン薬、ジフェンヒドラミン、オルフェナドリン等の抗ヒスタミン薬、ドネペジル、リバスチグミン、ガランタミン、タクリン等のコリンエステラーゼ阻害薬、メマンチン等のN−メチル−D−アスパラギン酸受容体拮抗薬、ハロペリドール、チオリダジン、チオチキセン、オランザピン、リスペリドン、クエチアピン、クロザピン等の抗精神薬、プロプラノロール等のβ遮断薬、ベンゾジアゼピン等の鎮静薬、ヘパリン、低分子ヘパリン、ワルファリン等の抗凝固剤、カルバマゼピン、ガバペンチン、フェニトイン、プレガバリン、バルプロサン、ラモトリジン等の抗痙攣薬、三環系、ベンラファキシン、ブプロピオン、アミトリプチリン、デシプラミン、パロキセチン等の抗うつ薬、クロニジン、チザニジン等の中枢性α−2アドレナリン作用薬、デキサメタゾン、プレドニゾロン等のコルチコステロイド、アマンタジン、デキストリメトロファン等のNMDA受容体拮抗薬、リドカイン、メキシレチン、カプサイシン等の局所麻酔薬、エトドラク、インドメタシン、スリン諾、トルメタチン、ナブメトン、ピロキシカム、アセトアミノフェン、フェノビプロン、フルルビプロン、イブプロフェン、ケトプロフェン、ナプロキセン、ナプロキセンナトリウム、オキサプロシン、アスピリン、コリンマグネシウム三サリチル酸、ジフルニサル、メクロフェナム酸塩、メフェナム酸、フェニルブタゾン、ケトロラク、セレコキシブ、コデイン、ヒドロコデイン、プロポキシフェン、フェンタニル、ヒドロモルホン、レボファノール、メペリジン、メサドン、モルヒネ、オキシコドン、オキシモルホン、ブプレノルフィン、ブトルファノール、ナルブフィン、ペンタゾシン等の鎮痛剤、プロポフォール等のラジカルスカベンジャー、抗炎症剤、セロトニン、ノルエピネフリン、NSAID、イチョウ葉エキス等が挙げられる。また、本発明の神経障害治療剤の投与とともに、低体温療法、脳低温療法、脳低体温療法等の低温療法を合わせて行う事もできる。 The therapeutic agent for neuropathy of the present invention may be administered together with one or more other agents. Other agents include any agent that can be used as a therapeutic agent for neuropathy, such as anti-Parkinson's disease agents such as levodopa, amantadine, carvidopa, and dopaminergic agents such as bromocryptin, purgoride, ropinilol, pramipexol. Drugs, selective B-type monoamine oxidase inhibitors (MAO-B) such as selegiline and lasalidine, catechol-O-methyltransferase (COMT) inhibitors such as enteracapon and tolucapon, and anticholinergic agents such as benztropon and trihexyphenidyl. Antihistamines such as diphenhydramine and orphenadrine, cholinesterase inhibitors such as donepezil, rivastigmin, galantamine and taclin, N-methyl-D-aspartate receptor antagonists such as memantin, haloperidol, thioridine, thiothixene, olanzapine, lisperidone, Antipsychotics such as quetiapine and clozapine, β-blockers such as propranolol, sedatives such as benzodiazepine, anticoagulants such as heparin, low molecular weight heparin, and warfarin, anticonvulsants such as carbamatepine, gabapentin, phenitoin, pregavalin, valprosan, and lamotridine. Drugs, tricyclics, antidepressants such as benrafaxin, bupropion, amitriptilin, desipramine, paroxetine, central α-2 adrenergic agonists such as chronidine and tizanidine, corticosteroids such as dexamethasone and prednisolone, amantadine and dextri NMDA receptor antagonists such as metrophans, local anesthetics such as codeine, mexiretin, capsaicin, etodrac, indomethasine, thrin consent, tolmetatin, nabmeton, pyroxicum, acetaminophen, phenobipron, flurubipron, ibuprofen, ketoprofen, naproxene, naproxene sodium , Oxaprosin, aspirin, cholinemagnesium trisalicylic acid, diflunisal, meclophenamate, mephenamic acid, phenylbutazone, ketrolac, selecoxib, codeine, hydrocodeine, propoxyphene, fentanyl, hydromorphone, levofanol, meperidine, mesadone, morphine, oxycodon, oxymorphone , Buprenorfin, butorphanol, nalbufin, pentazocin and other analgesics, propofol and other radical scavengers, anti-inflammatory agents, serotonin, norepinephrine, NSAID, ginkgo leaf extract and the like. In addition to the administration of the neuropathy therapeutic agent of the present invention, hypothermia therapy such as hypothermia therapy, cerebral hypothermia therapy, and cerebral hypothermia therapy can be combined.

本発明の間葉系幹細胞は様々な神経障害に用いることができるが、具体的疾患としては、自律神経障害、ホルネル症候群、多系統委縮症、純粋自律神経不全等の自律神経系障害、慢性疼痛、神経障害性疼痛、複合性局所疼痛症候群等の疼痛、虚血性脳卒中、一過性脳虚血発作、低酸素−虚血、脳内出血・脳室内出血などの頭蓋内出血、クモ膜下出血等の脳卒中(脳血管事故)、アルツハイマー病、脳血管性認知症、レーヴィ体認知症、HIV関連認知症、前頭側頭型認知症等の認知症、痙攣性症候群、アテトーゼまたは運動異常症候群及び失調性症候群等の脳性麻痺症候群、低血糖、高ナトリウム血症、低ナトリウム血症、低マグネシウム血症、先天代謝異常等による、新生児痙攣性疾患、多発性硬化症等の脱髄性疾患、キランーバレー症候群、遺伝性ニューロパシー、筋萎縮性側索硬化症(ALS)を含む運動ニューロン疾患、重症筋無力症、モノニューロパシー、多発ニューロパシー、神経叢障害等の末梢神経系障害、急性横断性脊髄炎、動静脈奇形、脊髄梗塞(虚血性脊髄障害)等の脊髄障害、脊髄小脳失調症、脊髄小脳変性症等の小脳疾患、脳腫瘍、脳炎、髄膜炎、パーキンソン病等が挙げられる。また、新生児を対象とした、周産期脳障害、新生児脳症及び脳性麻痺等に用いることもできる。これらのうち、虚血性脳卒中、一過性脳虚血発作、低酸素−虚血、脳内出血・脳室内出血などの頭蓋内出血、クモ膜下出血等の脳卒中(脳血管事故)、周産期脳障害、新生児脳症及び脳性麻痺等が好ましい。 The mesenchymal stem cells of the present invention can be used for various neuropathy, and specific diseases include autonomic neuropathy, Hornel syndrome, multisystem ataxia, autonomic neuropathy such as pure autonomic neuropathy, and chronic pain. , Neuropathic pain, pain such as complex local pain syndrome, ischemic stroke, transient cerebral ischemic attack, hypoxia-ischemia, intracranial bleeding such as intracerebral bleeding / intraventricular bleeding, subepithelial bleeding, etc. Stroke (cerebellar vascular accident), Alzheimer's disease, cerebrovascular dementia, Levi body dementia, HIV-related dementia, dementia such as frontotemporal dementia, spasmodic syndrome, atetose or dyskinesia syndrome and ataxia syndrome Cerebral paralysis syndrome, hypoglycemia, hypernatremia, hyponatremia, hypomagnesemia, congenital metabolic disorders, etc., neonatal spasmodic disease, demyelinating disease such as multiple sclerosis, Kiran-Valley syndrome, inheritance Sexual neuropathy, motor neuron disease including muscular atrophic lateral sclerosis (ALS), severe myasthenia, mononeuropathy, multiple neuropathy, peripheral nervous system disorders such as neuropathy, acute transverse myelitis, arteriovenous malformations, Examples thereof include spinal disorders such as spinal cord infarction (ischemic spinal disorder), cerebral diseases such as spinocerebellar ataxia and spinocerebellar degeneration, brain tumors, encephalitis, meningitis, Parkinson's disease and the like. It can also be used for perinatal encephalopathy, neonatal encephalopathy, cerebral palsy, etc. for newborns. Of these, ischemic stroke, transient cerebral ischemic attack, hypoxia-ischemia, intracranial hemorrhage such as intracerebral hemorrhage / intraventricular hemorrhage, stroke (cerebrovascular accident) such as subarachnoid hemorrhage, and perinatal brain Disorders, neonatal encephalopathy, cerebral palsy and the like are preferred.

以下に、実施例及び試験例を挙げて本発明を詳細に説明するが、本発明はこれらの実施例等によって限定されるものではない。 Hereinafter, the present invention will be described in detail with reference to Examples and Test Examples, but the present invention is not limited to these Examples and the like.

[臍帯由来間葉系幹細胞の調製及び培養]
臍帯由来細胞は、Cytotherapy, 18, 229-241, 2016に記載の方法で採取した。簡潔には、東京大学医科学研究所の倫理委員会の承認を得た上で、提供者の同意を得て採取された臍帯を1から2mmの断片に細断し、培養皿上へ播種し、セルアミーゴ(株式会社 椿本チエイン)を被せ、10% fetal bovine serum(FBS)と抗生物質を添加したα−minimal essential medium(MEM-α)中で培養する改良エクスプラント法により、臍帯由来間葉系幹細胞(以下「UCMSC」という)を得た。なお、臍帯組織から前述の改良エクスプラント法により得られた細胞を第一継代細胞(P1)とし、継代を行う事により継代数が進み、第二継代細胞(P2)のように記載する。
[Preparation and culture of umbilical cord-derived mesenchymal stem cells]
Umbilical cord-derived cells were collected by the method described in Cytotherapy, 18, 229-241, 2016. Briefly, with the approval of the Institutional Review Board of the Institute of Medical Sciences, the University of Tokyo, the umbilical cord collected with the consent of the donor is cut into 1 to 2 mm 3 pieces and seeded on a culture dish. Then, cover with cell amigo (Tsubakimoto Chain Co., Ltd.) and incubate in α-minimal essential medium (MEM-α) supplemented with 10% fetal bovine serum (FBS) and antibiotics. Leaf stem cells (hereinafter referred to as "UCMSC") were obtained. In addition, the cells obtained from the umbilical cord tissue by the above-mentioned improved explant method are designated as the first passage cells (P1), and the number of passages increases by performing the passage, and the cells are described as the second passage cells (P2). To do.

得られたUCMSCを、トリプシン(TrypLE Select (1X))を用いて剥離し、遠沈管に移し、400×gで5分間、遠心分離し細胞の沈殿を得た。上清を除去した後、細胞凍結保存液(STEM-CELLBANKER(ゼノアック社))を適量加え懸濁した。当該細胞懸濁溶液を、クライオチューブに分注し、フリーザー内で−80℃にて保存した。その後、液体窒素上の気相に移し、保存を継続した。 The obtained UCMSC was exfoliated using trypsin (TrypLE Select (1X)), transferred to a centrifuge tube, and centrifuged at 400 × g for 5 minutes to obtain a cell precipitate. After removing the supernatant, an appropriate amount of cell cryopreservation solution (STEM-CELLBANKER) was added and suspended. The cell suspension solution was dispensed into cryotubes and stored in a freezer at −80 ° C. After that, it was transferred to the gas phase on liquid nitrogen and storage was continued.

[mRNA発現]
UCMSCをP2からP4までそれぞれ間葉系幹細胞用無血清培地(Rohto社、無血清培地)、PromoCell培地(Mesenchymal Stem Cell Growth Medium 2(PromoCell社製、C-28009, Lot.435M415)にSupplement mix(PromoCell社製, C-39809, Lot.435M126 )を添加)及びMEM-α培地(Thermo Fisher社製、#12571-063、Lot.18997009に血清を10%となるよう添加)でそれぞれ培養し、凍結ストックを作製した。線維芽細胞はMEM-α培地で培養後、凍結ストックを作製した。それぞれのP4細胞と、線維芽細胞(Fibroblast)を起眠し、6ウェルプレートに15,000cells/cmで播種し、3種類それぞれの培地で1日間培養した後、Total RNAを回収した。HGF, SHH, OLIG2, VEGFA, NEUROG1, GRPR, IL1R1, CRHR2, CCKAR, APOEのmRNA発現を、それぞれ定量PCRを用いて検出した。
[MRNA expression]
Supplement mix of UCMSC from P2 to P4 to serum-free medium for mesenchymal stem cells (Rohto, serum-free medium) and PromoCell medium (Mesenchymal Stem Cell Growth Medium 2 (PromoCell, C-28009, Lot.435M415), respectively. (Promot Cell, C-39809, Lot.435M126) was added) and MEM-α medium (Thermo Fisher, # 12571-063, Lot.18997009 was added with serum to 10%), and frozen. Stock was made. Fibroblasts were cultured in MEM-α medium, and then frozen stock was prepared. Each P4 cell and fibroblast were put to sleep , seeded on a 6-well plate at 15,000 cells / cm 2 , and cultured in each of the three media for 1 day, and then Total RNA was collected. The mRNA expressions of HGF, SHH, OLIG2, VEGFA, NEUROG1, GRPR, IL1R1, CRHR2, CCKAR and APOE were detected by quantitative PCR, respectively.

MEM-α培地に比べ、無血清培地で培養した細胞は、HGF, SHH, OLIG2, VEGFA, NEUROG1, GRPR, IL1R1, CRHR2, CCKAR, APOEのmRNA発現が有意に高いことが分かった。また、PromoCell培地で培養した細胞は、MEM-α培地で培養した細胞に比べ、有意に VEGFA, NEUROG1, GRPR, CRHR2, CCKARの mRNA発現量が高いことがわかった(図1)。 It was found that the mRNA expression of HGF, SHH, OLIG2, VEGFA, NEUROG1, GRPR, IL1R1, CRHR2, CCKAR, APOE was significantly higher in the cells cultured in the serum-free medium than in the MEM-α medium. It was also found that the cells cultured in PromoCell medium had significantly higher mRNA expression levels of VEGFA, NEUROG1, GRPR, CRHR2, and CCKAR than the cells cultured in MEM-α medium (Fig. 1).

[浮遊培養細胞(SUS)と平面培養細胞(ADH)の調製]
前述の臍帯組織由来凍結細胞を起眠し、細胞培養フラスコに播種して間葉系幹細胞用無血清培地(Rohto社)を用いて培養した。培養4日目の細胞を回収して、必要細胞数を含んだ細胞懸濁液とマイクロキャリアを混合し、培養槽に添加、攪拌培養を開始した。3もしくは4日目に細胞/マイクロキャリア浮遊液を一部分取し、新しいマイクロキャリアを加えて3もしくは4日間培養する継代培養を2週間行い、浮遊培養細胞(以下、「SUS」と言う)を得た。前述の臍帯組織由来凍結細胞を起眠し、細胞培養フラスコにて播種して間葉系幹細胞用無血清培地(Rohto社)を用いて培養した。3もしくは4日に一度継代を行い、合計で2週間培養し、平面培養細胞(以下、「ADH」と言う)を得た。
[Preparation of suspended cultured cells (SUS) and planar cultured cells (ADH)]
The above-mentioned frozen cells derived from umbilical cord tissue were put to sleep, seeded in a cell culture flask, and cultured in a serum-free medium for mesenchymal stem cells (Rohto). The cells on the 4th day of culture were collected, a cell suspension containing the required number of cells and microcarriers were mixed, added to a culture tank, and stirring culture was started. On the 3rd or 4th day, a part of the cell / microcarrier suspension is taken, a new microcarrier is added, and subculture is carried out for 3 or 4 days for 2 weeks. Obtained. The above-mentioned frozen cells derived from umbilical cord tissue were put to sleep, seeded in a cell culture flask, and cultured in a serum-free medium for mesenchymal stem cells (Rohto). Subculture was performed once every 3 or 4 days, and the cells were cultured for a total of 2 weeks to obtain planar cultured cells (hereinafter referred to as "ADH").

[平面培養細胞(ADH)と浮遊培養細胞(SUS)の比較]
上記方法により得られた平面培養細胞(ADH)、または浮遊培養細胞(SUS)の凍結ストックからTotal RNAを回収した。PAX3, PAX5, EGF, CXCL1, GDNF, NRCAM, DLL1, HEYL, BMP2, NTN1, ASCL1, NRP2, VEGFA, APOEのmRNA発現を、それぞれ定量PCRを用いて検出した。
[Comparison of planar cultured cells (ADH) and suspended cultured cells (SUS)]
Total RNA was recovered from the frozen stock of planar cultured cells (ADH) or suspended cultured cells (SUS) obtained by the above method. The mRNA expressions of PAX3, PAX5, EGF, CXCL1, GDNF, NRCAM, DLL1, HEYL, BMP2, NTN1, ASCL1, NRP2, VEGFA and APOE were detected by quantitative PCR.

平面培養細胞(ADH)に比べ、浮遊培養細胞(SUS)で、有意にPAX3, PAX5, EGF, CXCL1, GDNF, NRCAM, DLL1, HEYL, BMP2, NTN1, ASCL1, NRP2, VEGFA, APOEのmRNA発現量が高いことがわかった(図2)。 The mRNA expression levels of PAX3, PAX5, EGF, CXCL1, GDNF, NRCAM, DLL1, HEYL, BMP2, NTN1, ASCL1, NRP2, VEGFA, APOE were significantly higher in suspension-cultured cells (SUS) than in planar cultured cells (ADH). Was found to be high (Fig. 2).

[脂肪由来間葉系幹細胞の調製]
ヒトドナーから同意を得た後、脂肪吸引法で得た皮下脂肪組織を生理食塩液で洗浄した。細胞外基質の破壊、及び細胞の単離を達成するために、コラゲナーゼ(溶媒は生理食塩液)を添加し、37℃で90分間振倒し、分散した。続いて、この上記懸濁液を800gで5分間、遠心分離して間質血管細胞群の沈殿を得た。上記細胞の沈殿に間葉系幹細胞用無血清培地(Rohto社)を加え、当該細胞懸濁液を400gで5分間遠心分離し、上清除去後に間葉系幹細胞用無血清培地(Rohto社)に再懸濁し、フラスコに細胞を播種した。細胞を37℃、5%CO中で数日間培養した。数日後に培養物をPBSで洗浄して、培養液中に含まれていた血球や脂肪組織の残存等を除去し、プラスチック容器に接着している間葉系幹細胞(以下「ADMSC」と言う)を得た。
[Preparation of adipose-derived mesenchymal stem cells]
After obtaining the consent of the human donor, the subcutaneous adipose tissue obtained by the liposuction method was washed with physiological saline. Collagenase (solvent is saline) was added and shaken at 37 ° C. for 90 minutes to disperse to achieve extracellular matrix disruption and cell isolation. Subsequently, the suspension was centrifuged at 800 g for 5 minutes to obtain a precipitate of stromal vascular cells. A serum-free medium for mesenchymal stem cells (Rohto) was added to the above-mentioned cell precipitate, the cell suspension was centrifuged at 400 g for 5 minutes, and after removing the supernatant, a serum-free medium for mesenchymal stem cells (Rohto) was used. The cells were seeded in a flask after resuspension. Cells were cultured at 37 ° C. in 5% CO 2 for several days. A few days later, the culture was washed with PBS to remove residual blood cells and adipose tissue contained in the culture medium, and the mesenchymal stem cells adhered to the plastic container (hereinafter referred to as "ADMSC"). Got

[脂肪組織由来間葉系幹細胞の凍結保存]
得られたADMSCを、トリプシンを用いて剥離し、遠沈管に移し、400×gで5分間、遠心分離し細胞の沈殿を得た。上清を除去した後、細胞凍結保存液(STEM-CELLBANKER(ゼノアック社))を適量加え懸濁した。当該細胞懸濁溶液を、クライオチューブに分注し、フリーザー内で−80℃にて保存した。その後、液体窒素上の気相に移し、保存を継続した。
[Cyropreservation of adipose tissue-derived mesenchymal stem cells]
The obtained ADMSC was exfoliated with trypsin, transferred to a centrifuge tube, and centrifuged at 400 × g for 5 minutes to obtain a cell precipitate. After removing the supernatant, an appropriate amount of cell cryopreservation solution (STEM-CELLBANKER) was added and suspended. The cell suspension solution was dispensed into cryotubes and stored in a freezer at −80 ° C. After that, it was transferred to the gas phase on liquid nitrogen and storage was continued.

[ラット一過性脳虚血モデル(小泉モデル)を用いた治療効果の確認]
ラット(Wistar、日本チャールズリバー社)の中大脳動脈(MCA)閉塞して、23.3〜27.8時間後にADMSC、ADH及びSUSを、HBSS(Hank's Balanced Salt solution)に懸濁して、尾静脈内から投与した(8×10cells/kg)。なお、比較対象として、細胞を投与せずHBSSのみを投与した動物を設けた。手術前、及び手術14日後に、体重、及び神経症状の観察と、ステップテスト、及びテープ剥がしテストを行った。
[Confirmation of therapeutic effect using rat transient ischemic model (Koizumi model)]
The middle cerebral artery (MCA) of a rat (Wistar, Charles River, Japan) was occluded, and ADMSC, ADH and SUS were suspended in HBSS (Hank's Balanced Salt solution) 23.3 to 27.8 hours later, and the tail vein. It was administered from the inner (8 × 10 6 cells / kg ). As a comparison target, an animal to which only HBSS was administered without administering cells was provided. Before and 14 days after surgery, body weight and neurological symptoms were observed, and a step test and a tape peeling test were performed.

手術前および手術14日後にラットの体幹、後肢および右前肢を保定して持ち上げ、左前肢のみが実験台に触れるようにして、水平面を逆手方向に約5秒間で30cm移動させるステップテストを実施した。その際の左前肢の歩数を記録した。3回繰り返して行い、歩数の平均値を測定値とした(ステップテスト)。また、木全らの報告(薬理と治療,1991;19:4491-4503)を参考に神経症状の観察を行った。 Before and 14 days after surgery, a step test was conducted in which the rat's trunk, hind limbs, and right forelimb were held and lifted, and only the left forelimb touched the laboratory table, and the horizontal plane was moved 30 cm in the opposite direction in about 5 seconds. did. The number of steps on the left forelimb at that time was recorded. It was repeated 3 times, and the average value of the number of steps was used as the measured value (step test). In addition, neurological symptoms were observed with reference to the report by Kimata et al. (Pharmacology and Treatment, 1991; 19: 4491-4503).

Leongらによって報告された測定方法(Leong et al. Stem Cells Translational Medicine、2012;1:177-187)に基づきテープ剥がしテストを行った。右(麻痺側)前肢の裏面に15mmのテープをはり、ケージ内に入れテープを剥がそうとするまでの時間を計測した(Cut offは120秒、テープ剥がしテスト)。A tape peeling test was performed based on the measurement method reported by Leong et al. (Leong et al. Stem Cells Translational Medicine, 2012; 1: 177-187). A 15 mm 2 tape was applied to the back of the right (paralyzed side) forelimb, placed in a cage, and the time until the tape was to be peeled off was measured (Cut off was 120 seconds, tape peeling test).

ADMSC、ADH及びSUSを投与することにより体重の減少度が軽減することが明らかとなった(図3)。また、ADMSC、ADH及びSUSを投与することにより神経症状スコアが減少することが明らかとなった(図4)。ADMSC、ADH、及びSUSを投与することによりステップ回数が上昇することが明らかとなった(図5)。ADMSC、ADH、及びSUSを投与することによりテープを剥がすまでの時間が短縮されることが明らかとなった(図6)。 It was revealed that administration of ADMSC, ADH and SUS reduced the degree of weight loss (Fig. 3). It was also revealed that administration of ADMSC, ADH and SUS reduced the neurological symptom score (Fig. 4). It was revealed that the number of steps was increased by administering ADMSC, ADH, and SUS (Fig. 5). It was revealed that administration of ADMSC, ADH, and SUS shortened the time required to remove the tape (Fig. 6).

以上の結果より、脳血管障害後の神経傷害に対する改善効果が示された。 From the above results, the improvement effect on nerve injury after cerebrovascular accident was shown.

[神経細胞と間葉系幹細胞の相互作用]
Vybrant DiO Cell-labeling solution (Thermo Fisher社製、#V22886)でDiD 蛍光染色を行ったSH-SY5Y(ヒト由来神経芽細胞腫;ECACC製、Lot.16E028、Acc Nc:94030304)を12 well plateに播種して、10% FBS(Thermo Fisher社製、#10437-028、Lot.1658423)及びPen-Strepを含むDMEM/F12(Thermo Fisher、#11320-033、Lot.1930023)培地で、37℃、O:20%、CO:5%条件下にて培養した。培養1日後、各well内の細胞をD-PBS(-)で2回洗浄し、Pen-Strepを含むDMEM, no glucose(Thermo Fisher, # 11966025)培地で、37℃、N:95%、O:5%条件下で2時間培養した。その後、上記と同様に調製して間葉系幹細胞用無血清培地(Rohto社)で培養してDiO 蛍光染色したUCMSCを12 well plateに添加し、10% FBS (Thermo Fisher社製、#10437-028、Lot.1658423)及びPen-Strepを含むDMEM/F12(Thermo Fisher、#11320-033、Lot.1930023)培地で、37℃、O:20%、CO:5%条件下で培養して、各時間経過後、タイムラプス画像を撮影した。なお、それぞれの写真においては、緑色はUCMSC(左下向きの矢印(柄の部分は点線)で指し示した細胞)を示し、赤色はSH-SY5Y(右上向きの矢印で指し示した細胞)を示している(図7)。
[Interaction between nerve cells and mesenchymal stem cells]
SH-SY5Y (human-derived neuroblastoma; ECACC, Lot.16E028, Acc Nc: 94030304) subjected to DiD fluorescence staining with Vybrant DiO Cell-labeling solution (Thermo Fisher, # V22886) into a 12-well plate. Seed in DMEM / F12 (Thermo Fisher, # 11320-033, Lot.1930023) medium containing 10% FBS (Thermo Fisher, # 10437-028, Lot.1658423) and Pen-Strep at 37 ° C. The cells were cultured under O 2 : 20% and CO 2 : 5% conditions. One day after culturing, the cells in each well were washed twice with D-PBS (-), and in DMEM, no glucose (Thermo Fisher, # 11966025) medium containing Pen-Strep, 37 ° C., N 2 : 95%, O 2 : Incubated for 2 hours under 5% conditions. Then, UCMSC prepared in the same manner as above, cultured in serum-free medium for mesenchymal stem cells (Rohto), and DiO fluorescently stained UCMSC was added to a 12-well plate, and 10% FBS (Thermo Fisher, # 10437-). Incubate in DMEM / F12 (Thermo Fisher, # 11320-033, Lot.1930023) medium containing 028, Lot.1658423) and Pen-Strep under the conditions of 37 ° C., O 2 : 20%, CO 2: 5%. After each time, a time-lapse image was taken. In each photograph, green indicates UCMSC (cells pointed by the arrow pointing downward to the left (dotted line in the handle)), and red indicates SH-SY5Y (cells indicated by the arrow pointing to the upper right). (Fig. 7).

UCMSC及びSH-SY5Yが相互に突起を伸長して、細胞間相互作用することが観測された。 It was observed that UCMSC and SH-SY5Y mutually extend protrusions and interact with each other.

SH-SY5Y(ヒト由来神経芽細胞腫;ECACC製、Lot.16E028、Acc Nc:94030304)を96 well plateに播種して、5% FBS(Thermo Fisher社製、#10437-028、Lot.1658423)及びPen-Strepを含むDMEM/F12(Thermo Fisher、#11320-033、Lot.1930023)培地で、37℃、O:20%、CO:5%条件下にて培養した。培養1日後、Well内の培地を除去し、Pen-Strepを含むDMEM, No glucose(Thermo Fisher, # 11966025)培地で、37℃、N:95%、O:5%条件(以下、OGD条件)下で24時間培養した(OGD処理群)。同様に、培養1日後、SH-SY5Y細胞を培養しているWell内の培地を除去し、前述の方法で調整した臍帯由来間葉系幹細胞を間葉系幹細胞用無血清培地(Rohto社)で培養したUCMSCを1,000cells/wellで添加して、OGD条件下で24時間培養した(MSC群)。なお、比較対象として、同様に、培養1日後、Well内の培地を除去し、新たな5% FBS(Thermo Fisher社製、#10437-028、Lot.1658423)及びPen-Strepを含むDMEM/F12(Thermo Fisher、#11320-033、Lot.1930023)培地で、37℃、O:20%、CO:5%条件下にて24時間培養したものを対照群(Control群)とした。培養後、各培養上清中の乳酸脱水素酵素(LDH)活性を測定した(図8)。SH-SY5Y (human-derived neuroblastoma; ECACC, Lot.16E028, Acc Nc: 94030304) was seeded on a 96-well plate and 5% FBS (Thermo Fisher, # 10437-028, Lot.1658423). And Pen-Strep were cultured in DMEM / F12 (Thermo Fisher, # 11320-033, Lot.1930023) medium at 37 ° C. under the conditions of O 2 : 20% and CO 2: 5%. One day after culturing, the medium in Well was removed, and the medium was DMEM, No glucose (Thermo Fisher, # 11966025) containing Pen-Strep at 37 ° C., N 2 : 95%, O 2 : 5% (hereinafter, OGD). Cultivated under (Conditions) for 24 hours (OGD treatment group). Similarly, one day after culturing, the medium in the Well where the SH-SY5Y cells are cultured is removed, and the umbilical cord-derived mesenchymal stem cells prepared by the above method are used in a serum-free medium for mesenchymal stem cells (Rohto). The cultured UCMSC was added at 1,000 cells / well and cultured for 24 hours under OGD conditions (MSC group). For comparison, similarly, 1 day after culturing, the medium in Well was removed, and DMEM / F12 containing new 5% FBS (Thermo Fisher, # 10437-028, Lot.1658423) and Pen-Strep was added. (Thermo Fisher, # 11320-033, Lot.1930023) A control group (Control group) was obtained by culturing in a medium under the conditions of 37 ° C., O 2 : 20%, and CO 2: 5% for 24 hours. After culturing, the lactate dehydrogenase (LDH) activity in each culture supernatant was measured (FIG. 8).

OGD(Oxygen and glucose deprivation)条件下で培養することにより、SH-SY5Y細胞のLDH活性が上昇するが、UCMSCと共培養することにより、LDH活性の上昇が抑制され、虚血による神経細胞死が抑制されることが確認された。 By culturing under OGD (Oxygen and glucose deprivation) conditions, the LDH activity of SH-SY5Y cells increases, but by co-culturing with UCMSC, the increase in LDH activity is suppressed and nerve cell death due to ischemia occurs. It was confirmed that it was suppressed.

SH-SY5Y(ヒト由来神経芽細胞腫;ECACC製、Lot.16E028、Acc Nc:94030304)を12 well plateにぞれぞれ0.038×10cells/wellずつ播種して、10% FBS(Thermo Fisher社製、#10437-028、Lot.1658423)及びPen-Strepを含むDMEM/F12(Thermo Fisher、#11320-033、Lot.1930023)培地で、37℃、O:20%、CO:5%条件下にて培養した。培養1日後、各well内の細胞をD-PBS(-)で2回洗浄後、前述と同様のOGD条件下で2時間培養した(細胞無投与群)。同様に、培養1日後、Well内の培地を除去し、前述の方法で調整した浮遊培養細胞(SUS)を、播種済みSH-SY5Y細胞の半量(半量群)、同量(同量群)及び2倍量(倍量群)播種したtranswell insertを載せ、OGD条件下で2時間培養した(MSC群)。その後、10% FBS(Thermo Fisher社製、#10437-028、Lot.1658423)及びPen-Strepを含むDMEM/F12(Thermo Fisher、#11320-033、Lot.1930023)培地で、37℃、O:20%、CO:5%条件下にて48時間培養した。なお、比較対象として、同様に、培養1日後、各well内の細胞をD-PBS(-)で2回洗浄後、新たな10% FBS (Thermo Fisher社製、#10437-028、Lot.1658423)及びPen-Strepを含むDMEM/F12(Thermo Fisher、#11320-033、Lot.1930023)培地で、37℃、O:20%、CO:5%条件下にて48時間培養した(Control群)。培養後、各SH-SY5Yの細胞活性をWST-8(Cell Counting Kit-8)にて評価した(図9)。SH-SY5Y (human neuroblastoma; ECACC made, Lot.16E028, Acc Nc: 94030304) were seeded by 12 well plate Nizorezore 0.038 × 10 6 cells / well and, 10% FBS ( DMEM / F12 (Thermo Fisher, # 11320-033, Lot.1930023) medium containing Thermo Fisher # 10437-028, Lot.1658423) and Pen-Strep at 37 ° C, O 2 : 20%, CO 2 : Cultured under 5% conditions. One day after culturing, the cells in each well were washed twice with D-PBS (-) and then cultured for 2 hours under the same OGD conditions as described above (cell-free group). Similarly, one day after culturing, the medium in Well was removed, and the suspension cultured cells (SUS) prepared by the above method were used in half the amount (half amount group), the same amount (same amount group), and the same amount of seeded SH-SY5Y cells. A transwell insert seeded in a double amount (double amount group) was placed and cultured for 2 hours under OGD conditions (MSC group). Then, in DMEM / F12 (Thermo Fisher, # 11320-033, Lot.1930023) medium containing 10% FBS (Thermo Fisher, # 10437-028, Lot.1658423) and Pen-Strep, 37 ° C., O 2 The cells were cultured for 48 hours under the conditions of: 20% and CO 2: 5%. Similarly, as a comparison target, after 1 day of culturing, the cells in each well were washed twice with D-PBS (-), and then a new 10% FBS (manufactured by Thermo Fisher, # 10437-028, Lot.1658423). ) And Pen-Strep in DMEM / F12 (Thermo Fisher, # 11320-033, Lot.1930023) medium under the conditions of 37 ° C., O 2 : 20%, CO 2 : 5% (Control). group). After culturing, the cell activity of each SH-SY5Y was evaluated by WST-8 (Cell Counting Kit-8) (Fig. 9).

OGD条件下で培養することにより、神経細胞の細胞活性は有意に下がるが、MSCと共培養することにより、細胞活性の低下を抑制できることが明らかとなった。 It was clarified that the cell activity of nerve cells was significantly reduced by culturing under OGD conditions, but the decrease of cell activity could be suppressed by co-culturing with MSC.

SH-SY5Y(ヒト由来神経芽細胞腫;ECACC製、Lot.16E028、Acc Nc:94030304)を12 well plateにそれぞれ0.038×10cells/wellずつ播種して、10% FBS(Thermo Fisher社製、#10437-028、Lot.1658423)及びPen-Strepを含むDMEM/F12(Thermo Fisher、#11320-033、Lot.1930023)培地で、37℃、O:20%、CO:5%条件下にて培養した。培養1日後、各well内の細胞をD-PBS(-)で2回洗浄後、前述と同様のOGD条件下で2時間培養した(細胞無投与群)。同様に、培養1日後、Well内の培地を除去し、前述の方法で調整した浮遊培養細胞(SUS、SUS群)、平面培養細胞(ADH、ADH群)及び線維芽細胞(Fibroblast、Fibroblast群)をSH-SY5Yと同量播種したtranswell insertを載せ、OGD条件下で2時間培養した。その後、10% FBS (Thermo Fisher社製、#10437-028、Lot.1658423)及びPen-Strepを含むDMEM/F12(Thermo Fisher、#11320-033、Lot.1930023)培地で、37℃、O:20%、CO:5%条件下にて48時間培養した。なお、比較対象として、同様に、培養1日後、各well内の細胞をD-PBS(-)で2回洗浄後、新たな10% FBS (Thermo Fisher社製、#10437-028、Lot.1658423)及びPen-Strepを含むDMEM/F12(Thermo Fisher、#11320-033、Lot.1930023)培地で、37℃、O:20%、CO:5%%条件下にて48時間培養した(Control群)。培養後、各SH-SY5Yの細胞活性をWST-8(Cell Counting Kit-8)にて評価した(図10)。SH-SY5Y (human neuroblastoma; ECACC made, Lot.16E028, Acc Nc: 94030304) were seeded by each 0.038 × 10 6 cells / well in 12 well plate the, 10% FBS (Thermo Fisher Inc. Made in DMEM / F12 (Thermo Fisher, # 11320-033, Lot.1930023) medium containing # 10437-028, Lot.1658423) and Pen-Strep, 37 ° C, O 2 : 20%, CO 2 : 5%. It was cultured under the conditions. One day after culturing, the cells in each well were washed twice with D-PBS (-) and then cultured for 2 hours under the same OGD conditions as described above (cell-free group). Similarly, one day after culturing, the medium in Well was removed, and suspended cultured cells (SUS, SUS group), planar cultured cells (ADH, ADH group) and fibroblasts (Fibroblast, Fibroblast group) prepared by the above method were removed. Was cultivated for 2 hours under OGD conditions with a transwell insert seeded in the same amount as SH-SY5Y. Then, in DMEM / F12 (Thermo Fisher, # 11320-033, Lot.1930023) medium containing 10% FBS (Thermo Fisher, # 10437-028, Lot.1658423) and Pen-Strep, 37 ° C., O 2 The cells were cultured for 48 hours under the conditions of: 20% and CO 2: 5%. Similarly, as a comparison target, after 1 day of culturing, the cells in each well were washed twice with D-PBS (-), and then a new 10% FBS (manufactured by Thermo Fisher, # 10437-028, Lot.1658423). ) And Pen-Strep in DMEM / F12 (Thermo Fisher, # 11320-033, Lot.1930023) medium under 37 ° C., O 2 : 20%, CO 2 : 5%% conditions for 48 hours ( Control group). After culturing, the cell activity of each SH-SY5Y was evaluated by WST-8 (Cell Counting Kit-8) (Fig. 10).

MSCと共培養することにより、OGD条件下で培養することによる神経細胞の細胞活性低下を抑制できるが、線維芽細胞(Fibroblast)によってはこの効果がなく、MSC特異的な効果であることが明らかとなった。 By co-culturing with MSC, it is possible to suppress the decrease in cell activity of nerve cells by culturing under OGD conditions, but it is clear that this effect is not present depending on fibroblasts and is an MSC-specific effect. It became.

SH-SY5Y(ヒト由来神経芽細胞腫;ECACC製、Lot.16E028、Acc Nc:94030304)を12 well plateにそれぞれ0.038×10cells/wellずつ播種して、10% FBS (Thermo Fisher社製、#10437-028、Lot.1658423)及びPen-Strepを含むDMEM/F12(Thermo Fisher、#11320-033、Lot.1930023)培地で、37℃、O:20%、CO:5%条件下にて培養した。培養1日後、各well内の細胞をD-PBS(-)で2回洗浄後、前述と同様のOGD条件下で2時間培養した(細胞無投与群)。同様に、培養1日後、Well内の培地を除去し、臍帯由来間葉系幹細胞(PromoCell社製)を間葉系幹細胞用無血清培地(Rohto社、無血清群)、PromoCell培地(Mesenchymal Stem Cell Growth Medium 2(PromoCell社製、C-28009、Lot.435M415)にSupplement mix(PromoCell社製、C-39809、Lot.435M126)を添加)、PromoCell群)及びMEM-α(Thermo Fisher社製、#12571-063、Lot.18997009、MEM-α群)でそれぞれ培養したUCMSC及び線維芽細胞(Fibroblast、Fibroblast群)をSH-SY5Yと同量培養したtranswell insertを載せ、OGD条件下で2時間培養した。その後、10% FBS (Thermo Fisher社製、#10437-028、Lot.1658423)及びPen-Strepを含むDMEM/F12(Thermo Fisher、#11320-033、Lot.1930023)培地で、37℃、O:20%、CO:5%条件下にて48時間培養した。なお、比較対象として、同様に、培養1日後、各well内の細胞をD-PBS(-)で2回洗浄後、新たな10% FBS (Thermo Fisher社製、#10437-028、Lot.1658423)及びPen-Strepを含むDMEM/F12(Thermo Fisher、#11320-033、Lot.1930023)培地で、37℃、O:20%、CO:5%条件下にて48時間培養した(Control群)。培養後、各SH-SY5Yの細胞活性をWST-8(Cell Counting Kit-8)にて評価した(図11)。SH-SY5Y (human neuroblastoma; ECACC made, Lot.16E028, Acc Nc: 94030304) were seeded by each 0.038 × 10 6 cells / well in 12 well plate the, 10% FBS (Thermo Fisher Inc. Made in DMEM / F12 (Thermo Fisher, # 11320-033, Lot.1930023) medium containing # 10437-028, Lot.1658423) and Pen-Strep, 37 ° C, O 2 : 20%, CO 2 : 5%. It was cultured under the conditions. One day after culturing, the cells in each well were washed twice with D-PBS (-) and then cultured for 2 hours under the same OGD conditions as described above (cell-free group). Similarly, 1 day after culturing, the medium in Well was removed, and umbilical cord-derived mesenchymal stem cells (manufactured by PromoCell) were used as serum-free medium for mesenchymal stem cells (Rohto, serum-free group) and PromoCell medium (Mesenchymal Stem Cell). Supplement mix (PromoCell, C-39809, Lot.435M126) added to Growth Medium 2 (PromoCell, C-28009, Lot.435M415), PromoCell group) and MEM-α (Thermo Fisher, # UCMSC and fibroblasts (Fibroblast, Fibroblast group) cultured in 12571-063, Lot.18997009, MEM-α group, respectively, were placed on a transwell insert cultured in the same amount as SH-SY5Y, and cultured for 2 hours under OGD conditions. .. Then, in DMEM / F12 (Thermo Fisher, # 11320-033, Lot.1930023) medium containing 10% FBS (Thermo Fisher, # 10437-028, Lot.1658423) and Pen-Strep, 37 ° C., O 2 The cells were cultured for 48 hours under the conditions of: 20% and CO 2: 5%. Similarly, as a comparison target, after 1 day of culturing, the cells in each well were washed twice with D-PBS (-), and then a new 10% FBS (manufactured by Thermo Fisher, # 10437-028, Lot.1658423). ) And Pen-Strep in DMEM / F12 (Thermo Fisher, # 11320-033, Lot.1930023) medium under the conditions of 37 ° C., O 2 : 20%, CO 2 : 5% (Control). group). After culturing, the cell activity of each SH-SY5Y was evaluated by WST-8 (Cell Counting Kit-8) (Fig. 11).

MSCと共培養することにより、OGD条件下で培養することによる神経細胞の細胞活性低下を抑制できるが、無血清培地で培養したMSCでその効果がより顕著であることが明らかとなった。 By co-culturing with MSC, it is possible to suppress the decrease in cell activity of nerve cells by culturing under OGD conditions, but it was clarified that the effect is more remarkable in MSC cultured in serum-free medium.

SH-SY5Y(ヒト由来神経芽細胞腫;ECACC製、Lot.16E028、Acc Nc:94030304)を12 well plateにそれぞれ0.038×10cells/wellずつ播種して、10% FBS(Thermo Fisher社製、#10437-028、Lot.1658423)及びPen-Strepを含むDMEM/F12(Thermo Fisher、#11320-033、Lot.1930023)培地で、37℃、O:20%、CO:5%条件下にて培養した。培養1日後、各well内の細胞をD-PBS(-)で2回洗浄後、前述と同様のOGD条件下で2時間培養した(細胞無投与群)。同様に、培養1日後、Well内の培地を除去し、臍帯由来間葉系幹細胞(PromoCell社製、C-12971、Lot.427Z021)を間葉系幹細胞用無血清培地(Rohto社、無血清群)、PromoCell培地(Mesenchymal Stem Cell Growth Medium 2(PromoCell社製、C-28009, Lot.435M415)にSupplement mix(PromoCell社製、C-39809、Lot.435M126)を添加、PromoCell群)及びMEM-α(Thermo Fisher社製、#12571-063、Lot.18997009、MEM-α群)でそれぞれ培養したUCMSC及び線維芽細胞(Fibroblast、線維芽細胞群)をSH-SY5Yと同量培養したtranswell insertを載せ、OGD条件下で2時間培養した。その後、10% FBS(Thermo Fisher社製、#10437-028、Lot.1658423)及びPen-Strepを含むDMEM/F12(Thermo Fisher、#11320-033、Lot.1930023)培地で、37℃、O:20%、CO:5%条件下にて24〜48時間培養した。48H培養後、細胞画像を取得した(図12)。なお、死細胞の染色はEthD-IIIを使用した。各画像の死細胞の数を計測後、死細胞率を「死細胞数/WST-8の吸光度」にて算出した(図13)。SH-SY5Y (human neuroblastoma; ECACC made, Lot.16E028, Acc Nc: 94030304) were seeded by each 0.038 × 10 6 cells / well in 12 well plate the, 10% FBS (Thermo Fisher Inc. Made in DMEM / F12 (Thermo Fisher, # 11320-033, Lot.1930023) medium containing # 10437-028, Lot.1658423) and Pen-Strep, 37 ° C, O 2 : 20%, CO 2 : 5%. It was cultured under the conditions. One day after culturing, the cells in each well were washed twice with D-PBS (-) and then cultured for 2 hours under the same OGD conditions as described above (cell-free group). Similarly, 1 day after culturing, the medium in Well was removed, and umbilical cord-derived mesenchymal stem cells (PromoCell, C-12971, Lot.427Z021) were used as serum-free medium for mesenchymal stem cells (Rohto, serum-free group). ), Supplement mix (PromoCell, C-39809, Lot.435M126) added to PromoCell medium (Mesenchymal Stem Cell Growth Medium 2 (PromoCell, C-28009, Lot.435M415), PromoCell group) and MEM-α UCMSC and fibroblast (fibroblast, fibroblast group) cultured in (Thermo Fisher, # 12571-063, Lot.18997009, MEM-α group) were placed in the same amount as SH-SY5Y. , Cultivated for 2 hours under OGD conditions. Then, in DMEM / F12 (Thermo Fisher, # 11320-033, Lot.1930023) medium containing 10% FBS (Thermo Fisher, # 10437-028, Lot.1658423) and Pen-Strep, 37 ° C., O 2 The cells were cultured for 24-48 hours under the conditions of: 20% and CO 2: 5%. After culturing for 48H, cell images were acquired (Fig. 12). EthD-III was used for staining of dead cells. After measuring the number of dead cells in each image, the dead cell rate was calculated by "number of dead cells / absorbance of WST-8" (FIG. 13).

無血清群では、細胞無投与群、線維芽細胞群、PromoCell群及びMEM-α群に比べ、細胞増殖活性が高いことが明らかとなった。また、無血清群では、細胞無投与群、PromoCell群群及びMEM-α群に比べ、全細胞数に対する、死細胞数が少ないことが明らかとなった。 It was revealed that the cell proliferation activity was higher in the serum-free group than in the cell-free group, the fibroblast group, the PromoCell group and the MEM-α group. In addition, it was revealed that the number of dead cells in the serum-free group was smaller than the total number of cells in the cell-free group, the PromoCell group, and the MEM-α group.

本発明によると、神経障害の新規治療剤を提供することができる。 According to the present invention, it is possible to provide a novel therapeutic agent for neuropathy.

Claims (5)

HGF, SHH, OLIG2, VEGFA, NEUROG1, GRPR, IL1R1, CRHR2, CCKAR, APOE, PAX3, PAX5, EGF, CXCL1, GDNF, NRCAM, DLL1, HEYL, BMP2, NTN1, ASCL1, NRP2のいずれか一つ、または二つ以上が高発現であることを特徴とする、間葉系幹細胞。 HGF, SHH, OLIG2, VEGFA, NEUROG1, GRPR, IL1R1, CRHR2, CCKAR, APOE, PAX3, PAX5, EGF, CXCL1, GDNF, NRCAM, DLL1, HEYL, BMP2, NTN1, ASCL1, NRP2, or Mesenchymal stem cells characterized by high expression of two or more. 他家由来である、請求項1に記載の間葉系幹細胞。 The mesenchymal stem cell according to claim 1, which is derived from an allogeneic family. 臍帯組織もしくは脂肪組織由来である、請求項1又は2に記載の間葉系幹細胞。 The mesenchymal stem cell according to claim 1 or 2, which is derived from umbilical cord tissue or adipose tissue. 浮遊培養法により調製される、請求項1から3のいずれか1項に記載の間葉系幹細胞。 The mesenchymal stem cell according to any one of claims 1 to 3, which is prepared by a suspension culture method. 請求項1から4のいずれか1項に記載の間葉系幹細胞を含有する神経障害治療剤。 A neuropathy therapeutic agent containing mesenchymal stem cells according to any one of claims 1 to 4.
JP2020530111A 2018-07-09 2019-06-28 Mesenchymal stem cells and neuropathy therapeutic agents Pending JPWO2020012991A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
JP2023215423A JP2024023760A (en) 2018-07-09 2023-12-21 Mesenchymal stem cells and therapeutic agents for neurological disorders

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
JP2018130205 2018-07-09
JP2018130205 2018-07-09
PCT/JP2019/025928 WO2020012991A1 (en) 2018-07-09 2019-06-28 Mesenchymal stem cell and therapeutic agent for neuropathy

Related Child Applications (1)

Application Number Title Priority Date Filing Date
JP2023215423A Division JP2024023760A (en) 2018-07-09 2023-12-21 Mesenchymal stem cells and therapeutic agents for neurological disorders

Publications (1)

Publication Number Publication Date
JPWO2020012991A1 true JPWO2020012991A1 (en) 2021-07-15

Family

ID=69142832

Family Applications (2)

Application Number Title Priority Date Filing Date
JP2020530111A Pending JPWO2020012991A1 (en) 2018-07-09 2019-06-28 Mesenchymal stem cells and neuropathy therapeutic agents
JP2023215423A Pending JP2024023760A (en) 2018-07-09 2023-12-21 Mesenchymal stem cells and therapeutic agents for neurological disorders

Family Applications After (1)

Application Number Title Priority Date Filing Date
JP2023215423A Pending JP2024023760A (en) 2018-07-09 2023-12-21 Mesenchymal stem cells and therapeutic agents for neurological disorders

Country Status (3)

Country Link
JP (2) JPWO2020012991A1 (en)
CN (1) CN112368370A (en)
WO (1) WO2020012991A1 (en)

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2021192670A1 (en) * 2020-03-27 2021-09-30 ロート製薬株式会社 Mesenchymal stem cells and medium for mesenchymal stem cells
CN112608892B (en) * 2020-12-25 2023-11-14 深圳博雅感知药业有限公司 Method for serum-free separation and subculturing of umbilical cord mesenchymal stem cells by using platelet lysate

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100098739A1 (en) * 2008-10-20 2010-04-22 University Of Virginia Patent Foundation Compositions and methods for modular soft tissue repair
EP2850178A4 (en) * 2012-05-18 2015-10-28 Agency Science Tech & Res Umbilical cord mesenchymal stem cell exosomes
WO2017141831A1 (en) * 2016-02-16 2017-08-24 ロート製薬株式会社 Therapeutic agent containing fat-tissue derived stromal cells for ischemic disease and manufacturing method therefor
JP7108541B2 (en) * 2016-10-18 2022-07-28 ロート製薬株式会社 Kit for preparing therapeutic agent for disease, therapeutic agent for disease, and method for preparing therapeutic agent for disease

Also Published As

Publication number Publication date
CN112368370A (en) 2021-02-12
WO2020012991A1 (en) 2020-01-16
JP2024023760A (en) 2024-02-21

Similar Documents

Publication Publication Date Title
JP6371286B2 (en) Prevention and treatment of pre-eclampsia
JP7082372B2 (en) Pulmonary fibrosis treatment agent, PTPRR expression promoter and pulmonary fibrosis treatment kit
JP2024023760A (en) Mesenchymal stem cells and therapeutic agents for neurological disorders
WO2018159432A1 (en) Mesenchymal stem cells and pharmaceutical composition
JPWO2017204231A1 (en) Therapeutic agents for brain disorders including umbilical cord-derived cells
JP7246569B2 (en) Composition for promoting stem cell-derived exosome production and enhancing stem cell potency
KR20190063453A (en) Composition For Promoting Production of Stem Cell-derived Exosomes
JP6960120B2 (en) Liver disease therapeutic agents and methods for treating liver disease
WO2018116732A1 (en) Therapeutic agent for non-alcoholic steatohepatitis, and kit for treatment of non-alcoholic steatohepatitis
JP2019156739A (en) Mesenchymal stem cell, disease therapeutic agent, and microparticle
JP2017119646A (en) Sex hormone secretion promoter and germ cell proliferation promoter
JP2023024665A (en) Mesenchymal stem cells and therapeutic agent for liver disease
TWI510626B (en) Method of washing adherent cell using trehalose-containing cell-washing solution
JP7136700B2 (en) Cell pharmaceutical composition, disease treatment kit and cell suspension solution
JP7189659B2 (en) Cell pharmaceutical composition for treating disease, kit for treating disease, method for preparing cell pharmaceutical composition
JP2022522460A (en) How to improve the angiogenic potential of mesenchymal stem cells
JP2022179749A (en) Cell pharmaceutical composition, disease treatment kit, and cell suspension solution

Legal Events

Date Code Title Description
A621 Written request for application examination

Free format text: JAPANESE INTERMEDIATE CODE: A621

Effective date: 20220531

A131 Notification of reasons for refusal

Free format text: JAPANESE INTERMEDIATE CODE: A131

Effective date: 20230516

A521 Request for written amendment filed

Free format text: JAPANESE INTERMEDIATE CODE: A523

Effective date: 20230713

A02 Decision of refusal

Free format text: JAPANESE INTERMEDIATE CODE: A02

Effective date: 20230926

A521 Request for written amendment filed

Free format text: JAPANESE INTERMEDIATE CODE: A523

Effective date: 20231221

A911 Transfer to examiner for re-examination before appeal (zenchi)

Free format text: JAPANESE INTERMEDIATE CODE: A911

Effective date: 20231228

A912 Re-examination (zenchi) completed and case transferred to appeal board

Free format text: JAPANESE INTERMEDIATE CODE: A912

Effective date: 20240216