US20130195991A1 - Composition for Treatment of Damaged Part - Google Patents

Composition for Treatment of Damaged Part Download PDF

Info

Publication number
US20130195991A1
US20130195991A1 US13/637,107 US201113637107A US2013195991A1 US 20130195991 A1 US20130195991 A1 US 20130195991A1 US 201113637107 A US201113637107 A US 201113637107A US 2013195991 A1 US2013195991 A1 US 2013195991A1
Authority
US
United States
Prior art keywords
cells
damaged part
conditioned medium
treatment composition
dental pulp
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US13/637,107
Other languages
English (en)
Inventor
Minoru Ueda
Yoichi Yamada
Katsumi Ebisawa
Akihito Yamamoto
Kiyoshi Sakai
Kohki Matsubara
Hisashi Hattori
Masahiko Sugiyama
Takanori Inoue
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.)
University of Tokushima NUC
Original Assignee
Nagoya University NUC
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 Nagoya University NUC filed Critical Nagoya University NUC
Priority to US13/637,107 priority Critical patent/US20130195991A1/en
Assigned to NATIONAL UNIVERSITY CORPORATION NAGOYA UNIVERSITY reassignment NATIONAL UNIVERSITY CORPORATION NAGOYA UNIVERSITY ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: EBISAWA, KATSUMI, HATTORI, HISASHI, INOUE, TAKANORI, MATSUBARA, KOHKI, SAKAI, KIYOSHI, SUGIYAMA, MASAHIKO, UEDA, MINORU, YAMADA, YOICHI, YAMAMOTO, AKIHITO
Publication of US20130195991A1 publication Critical patent/US20130195991A1/en
Assigned to TOKUSHIMA UNIVERSITY reassignment TOKUSHIMA UNIVERSITY ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: NATIONAL UNIVERSITY CORPORATION NAGOYA UNIVERSITY
Abandoned 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
    • 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
    • A61K35/30Nerves; Brain; Eyes; Corneal cells; Cerebrospinal fluid; Neuronal stem cells; Neuronal precursor cells; Glial cells; Oligodendrocytes; Schwann cells; Astroglia; Astrocytes; Choroid plexus; Spinal cord tissue
    • 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
    • A61K35/32Bones; Osteocytes; Osteoblasts; Tendons; Tenocytes; Teeth; Odontoblasts; Cartilage; Chondrocytes; Synovial membrane
    • 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
    • A61K35/54Ovaries; Ova; Ovules; Embryos; Foetal cells; Germ cells
    • A61K35/545Embryonic stem cells; Pluripotent stem cells; Induced pluripotent stem cells; Uncharacterised stem cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/18Growth factors; Growth regulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/18Growth factors; Growth regulators
    • A61K38/1833Hepatocyte growth factor; Scatter factor; Tumor cytotoxic factor II
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/18Growth factors; Growth regulators
    • A61K38/1841Transforming growth factor [TGF]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/18Growth factors; Growth regulators
    • A61K38/1858Platelet-derived growth factor [PDGF]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/18Growth factors; Growth regulators
    • A61K38/1858Platelet-derived growth factor [PDGF]
    • A61K38/1866Vascular endothelial growth factor [VEGF]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/22Hormones
    • A61K38/30Insulin-like growth factors, i.e. somatomedins, e.g. IGF-1, IGF-2
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/02Stomatological preparations, e.g. drugs for caries, aphtae, periodontitis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/02Drugs for dermatological disorders for treating wounds, ulcers, burns, scars, keloids, or the like
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • A61P25/16Anti-Parkinson drugs
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0652Cells of skeletal and connective tissues; Mesenchyme
    • C12N5/0662Stem cells
    • C12N5/0664Dental pulp stem cells, Dental follicle stem cells

Definitions

  • the present invention relates to a composition for treatment of a damaged part, and a treatment method using the same.
  • stem cells include embryonic stem cells (ES cells), induced pluripotent stem cells (iPS cells) and somatic stem cells.
  • ES cells embryonic stem cells
  • iPS cells induced pluripotent stem cells
  • somatic stem cells mesenchymal stem cells (MSCs) isolated from various tissues including bone marrow, adipose tissue, skin, umbilical cord and placenta have been used in particular in clinical applications in skin regeneration.
  • MSCs mesenchymal stem cells isolated from various tissues including bone marrow, adipose tissue, skin, umbilical cord and placenta have been used in particular in clinical applications in skin regeneration.
  • bone marrow aspiration is an invasive and painful procedure for the donor.
  • BMSCs bone marrow stem cells
  • a neurological disorder particularly an intractable neurological disorder such as spinal cord injury, is one of the diseases to which therapy by regenerative medicine is expected to be applied.
  • Transplantation therapy of an intractable neurological disorder using neural stem cells from human embryos or ES cells is recognized as a realistic research target, but has a serious problem in terms of morality and safety. Therefore, practical “stem cell source” is still searched for (for example, Keirstead et al., Human embryonic stem cell - derived oligodendrocyte progenitor cell transplants remyelinate and restore locomotion after spinal cord injury , Journal of Neuroscience (2005) vol. (19) pp. 4694; Okano et al., Neural stem cells and regeneration of injured spinal cord , Kidney international (2005), Vol. 68, pp.
  • stem cells in a living organism include stem cells derived from bone marrow or adipose tissue (for example, International Publication 02/086108 pamphlet). These stem cells have shortcomings such as (1) reduction with age in the number of stem cells that can be obtained, (2) difficulty in terms of ensuring the safety of transplanted stem cells due to accumulation of genetic mutations with age, (3) low proliferative capacity of the cells and (4) severe body invasion accompanying the collection of stem cells (for example, Gronthos et al., Postnatal human dental pulp stem cells ( DPSCs ) in vitro and in vivo , Proc Natl Acad Sci USA (2000) vol. 97 (25) pp.
  • DPSCs Postnatal human dental pulp stem cells
  • SHED stem cells from human exfoliated deciduous teeth , Proceedings of the National Academy of Sciences (2003) Vol. 100, 5807-5812; Arthur et al., Adult human dental pulp stem cells differentiate toward functionally active neurons under appropriate environmental cues , Stem Cells (2008) vol. 26 (7) pp. 1787-1795). Since SHED and DPSCs are self-derived tissue stem cells, safety in the case of transplantation is high, and hardly any moral problem is involved.
  • DPSCs have a potential to be employein cell-based treatment for systemic disorders such as neuronal disorders and cardiac diseases, and that DPSCs ameliorate ischemic disorders (Arthur et al., Adult human dental pulp stem cells differentiate toward functionally active neurons under appropriate environmental cues , Stem Cells (2008) vol. 26 (7) pp.
  • MSCs can contribute to skin repair. Further, wound healing by external application of various growth factors is widely studied. However, the result of the use of growth factors in single administration or multiple administrations, or the result of the use of multiple growth factors in combination with a view to obtaining synergistic effects, has not been clinically confirmed.
  • UVB ultraviolet rays
  • HDF human dermal fibroblasts
  • VEGF vascular endothelial growth factor
  • HGF hepatocyte growth factor
  • IGF insulin-like growth factor
  • PDGF platelet-derived growth factor
  • TGF- ⁇ transforming growth factor ⁇
  • cytokines are reported as important functions of MSCs, and a wide variety of pharmaceutical activities of MSCs has been demonstrated in, particularly, skin biology (Jettanacheawchankit S, Sasithanasate S, Sangvanich P, Banlunara W, Thunyakitpisal P., Acemannan stimulates gingival fibroblast proliferation; expressions of keratinocyte growth factor -1 , vascular endothelial growth factor, and type I collagen; and wound healing , J Pharmacol Sci. 2009 April; 109(4): 525-531; Miura et al., SHED: stem cells from human exfoliated deciduous teeth , Proceedings of the National Academy of Sciences (2003) Vol.
  • dental pulp stem cells such as SHED or DPSCs can be medically applied, and specific target diseases thereof are not known at all.
  • An object of the present invention is to provide a novel therapeutic means that utilizes dental pulp stem cells.
  • the present invention encompasses the following aspects:
  • a damaged part treatment composition for repairing a damaged part of a target tissue including a stem cell-conditioned medium obtained by culturing stem cells.
  • the stem cell-conditioned medium includes at least two cytokines selected from the group consisting of vascular endothelial growth factor (VEGF), hepatocyte growth factor (HGF), insulin-like growth factor (IGF), platelet-derived growth factor (PDGF) and transforming growth factor ⁇ (TGF- ⁇ ).
  • VEGF vascular endothelial growth factor
  • HGF hepatocyte growth factor
  • IGF insulin-like growth factor
  • PDGF platelet-derived growth factor
  • TGF- ⁇ transforming growth factor ⁇
  • CNS central nervous system
  • the damaged part treatment composition according to any one of [1] to [9], wherein the treatment of a damaged part includes treatment of a CNS disease, and the CNS disease is a disease or disorder selected from the group consisting of a spinal cord injury, a neurodegenerative disorder, degeneration or loss of neuronal cells and a retinal disease involving a neuronal cell disorder.
  • the CNS disease is a disease or disorder selected from the group consisting of a spinal cord injury, a neurodegenerative disorder, degeneration or loss of neuronal cells and a retinal disease involving a neuronal cell disorder.
  • a damaged part treatment method for repairing a damaged part of a target tissue including administering the damaged part treatment composition of any one of [1] to [10] to a patient having the target tissue for the damaged part treatment composition, in an amount effective for repairing the damaged part of the target tissue.
  • a method of treating cerebral infarction including administering the damaged part treatment composition of any one of [1] to [10] to a cerebral infarction patient, in an amount therapeutically effective for repairing a damaged part of the brain.
  • a method of treating a CNS disease including administering the damaged part treatment composition of any one of [1] to [10] as a CNS disease treatment composition to a CNS disease patient, in a therapeutically effective amount.
  • dental pulp stem cell is an undifferentiated dental pulp stem cell that has not been subjected to differentiation-inducing treatment after obtainment thereof, or a differentiation-induced dental pulp stem cell that has been induced to differentiate into a neural cell after obtainment thereof.
  • a novel treatment means utilizing a dental pulp stem cell specifically a damaged part treatment composition and a production method thereof, and a damaged part treatment method using the damaged part treatment composition, can be provided.
  • FIG. 1 is a diagram illustrating an experimental design using hairless mice. Wrinkle was induced by UVB irradiation.
  • FIG. 2 is a view showing the morphology, immunological analysis and proliferation rates of various types of cells.
  • A) to (C) respectively represent (A) BMSC, (B) DPSC and (C) SHED ( ⁇ 40).
  • D) to (F) represent immunofluorescence staining images of the stem cell marker STRO-1.
  • D) BMSC, (E) DPSC and (F) SHED were positive for STRO-1 (green fluorescence).
  • DAPI was used to visualize the nuclei (blue fluorescence).
  • G The proliferation rates of SHED, DPSCs and BMSCs were assessed using BrdU. Bar: standard deviation. Significance: *P ⁇ 0.05.
  • FIG. 3 is a view showing evaluation of wrinkles by replica analysis after SH-CM injection.
  • A a group to be treated
  • B a group treated with 100% SH-CM.
  • FIG. 4 is a diagram demonstrating amelioration of wrinkles in natural level of SH-CM- and SHED-injected group.
  • FIG. 5 is a view showing Hematoxylin-Eosin staining images.
  • A SH-CM-treated group.
  • B SHED-injected group.
  • FIG. 6 is a graph comparing the dermal thicknesses.
  • FIG. 7 is a diagram showing an effect of SH-CM on the proliferation of HDF.
  • FIG. 8 shows a western blotting analysis demonstrating an effect of SH-CM on collagen type I and MMP-1.
  • FIG. 9 is a conceptual diagram illustrating the mechanism of bone regeneration using the composition of the invention.
  • FIG. 10 is an explanatory diagram illustrating the experimental method according to Example 3 of the invention.
  • FIG. 11 is a view explaining the calculation of the BIC value employed in Example 3 of the invention.
  • FIG. 12 shows staining images obtained as a result of Example 3 of the invention.
  • FIG. 13 is a diagram showing the results of Example 3 of the invention.
  • FIG. 14 is a photograph demonstrating the results of Example 3 of the invention.
  • FIG. 15 is an X-ray photograph showing the results of the clinical case of Example 3 of the invention.
  • FIG. 16 is a view explaining the experimental model of Example 4 of the invention.
  • FIG. 17 is a photograph explaining the treatment modalities employed in Example 4 of the invention.
  • FIG. 18 is a photograph explaining the treatment modalities employed in Example 4 of the invention.
  • FIG. 19 is a diagram explaining the treatment modalities employed in Example 4 of the invention.
  • FIG. 20 shows staining images showing the regeneration state of the cementum obtained as a result of Example 4 of the invention.
  • the upper photographs show the case of using GF, and the lower photographs show the case of using PRP.
  • FIG. 21 is a graph showing the results of Example 4 of the invention (N 2 —NC).
  • FIG. 22 is a graph showing the results of Example 4 of the invention (N 1 -JE).
  • FIG. 23 is a photograph showing the pretreatment in the clinical case of Example 4 of the invention.
  • FIG. 24 is a photograph explaining the manner of treatment in the clinical case of Example 4 of the invention.
  • FIG. 25 is a photograph showing the results of the clinical case of Example 4 of the invention.
  • FIG. 26 is a diagram illustrating the induction of cerebral infarction according to Example 5 of the invention.
  • FIG. 27 is a graph showing changes in disability score after starting the nasal administration to Group I, Group II and Group III in Example 5 of the invention.
  • FIG. 28 is a graph showing the infarct volumes on day 16 after starting the nasal administration to Group I, Group II and Group III in Example 5 of the invention.
  • FIG. 29 is a conceptual diagram explaining a preparation method of the conditioned medium.
  • hSHED stem cell from human exfoliated deciduous teeth.
  • hDPSC permanent teeth dental pulp stem cell.
  • hBMSC human bone marrow mesenchymal stem cell.
  • hFibroblast human fibroblast.
  • FIG. 30 is a photograph showing the results of a neurite outgrowth experiment (phase-contrast microscopic image).
  • FIG. 31 shows the results of a neurite outgrowth experiment.
  • the graph shows the proportion of cells of which neurites were observed (left) and neurite length (right).
  • FIG. 32 is a photograph showing the results of a neurite outgrowth experiment (phase-contrast microscopic image).
  • FIG. 33 shows the result of a neurite outgrowth experiment.
  • the graph shows the proportion of cells of which neurites were observed (left) and neurite length (right).
  • FIG. 34 is a photograph showing the results of an apoptosis inhibition experiment (TUNEL assay).
  • FIG. 35 shows the result of an apoptosis inhibition experiment (TUNEL assay).
  • the graph shows statistically-processed apoptosis inhibiting effects of the conditioned medium from dental pulp stem cells.
  • the left graph is a graph in which apoptosis inhibiting effects in the presence of CSPG are checked, and the right graph is a graph in which apoptosis inhibiting effects in the presence of MAG are checked.
  • FIG. 36 is a graph showing the results of an experiment using an animal model of spinal cord crush injury.
  • SHED-CM dental pulp stem cell-conditioned medium administered group.
  • BMSC-CM bone marrow mesenchymal stem cell-conditioned medium administered group.
  • Control PBS administered group.
  • FIG. 37 shows the results of an experiment using an animal model of spinal cord crush injury. Comparison between the control group and the SHED-CM group is shown in terms of the bone marrow state (upper photograph) and the spinal cord weight (lower graph).
  • FIG. 38 shows the results of an experiment using an animal model of spinal cord crush injury.
  • SHED-CM dental pulp stem cell-conditioned medium administered group.
  • Control PBS administered group.
  • FIG. 39 shows the results of an experiment using an animal model of spinal cord crush injury.
  • SHED-CM dental pulp stem cell-conditioned medium administered group.
  • Control PBS administered group.
  • the damaged part treatment composition of the invention is a damaged part treatment composition for repairing a damaged part of a target tissue, the composition including a stem cell-conditioned medium obtained by culturing stem cells.
  • the damaged part treatment method of the invention is a damaged part treatment method for repairing or restoring a damaged part of a target tissue, the method including administering the damaged part treatment composition to a patient having the target tissue for the damaged part treatment composition, in an amount effective for repairing the damaged part of the target tissue.
  • SHED growth factor derived from stem cells from human exfoliated deciduous teeth
  • HDFs human dermal fibroblasts
  • SHED affect HDFs by enhancing collagen synthesis and activating the growth and migration activity of HDFs.
  • SHED or SHED-derived conditioned medium (SH-CM) can be used for treatment of photo-aging.
  • SHED and SH-CM should be structurally suitable for treatment of photo-aging.
  • SHED contribute to enhancement of the wound healing activity of HDFs, mainly with a secreted growth factor or extracellular matrix protein.
  • deciduous teeth naturally exfoliate during infancy, and are usually disposed of as they are. Therefore, utilization of stem cells from human exfoliated deciduous teeth has a great advantage in terms of the absence of invasiveness of the obtainment thereof and morality problem for utilization.
  • a stem cell-conditioned medium obtained by culturing stem cells is used as an active ingredient for a damaged part treatment composition.
  • the stem cell-conditioned medium which contains a cytokine mixture
  • the stem cell-conditioned medium induces cell growth in the damaged part, as a result of which the tissue having the damaged part can be repaired.
  • the mixture of cytokines in the stem cell-conditioned medium used in the invention serves as an inductive signal for endogenous stem cells in the target tissue, and, therefore, the endogenous stem cells can differentiate and proliferate.
  • the proliferation of cells, generation of extracellular matrix, etc. may occur in the damaged part of the target tissue. From these, it is thought that a tissue having a damaged part can be repaired based on such regenerative ability of endogenous stem cells in the target tissue.
  • the term “damaged part” means a part in a tissue that became unable to perform its original function due to occurrence of physical or physiological defect in the tissue, and the concept thereof encompasses external injury as well as a injured part, dysfunctional part or diseased part caused by physical or physiological defect of the tissue.
  • “repair” means that some or all of the functions that was lost due to damage to the target tissue are maintained or recovered as compared to the functions of the damaged part at the time of damaging, and broadly encompasses recovery of the functions of the tissue as well as regeneration as a functional tissue.
  • the assessment for the maintenance or recovery of the functions may be carried out based on, for example, an assay usually employed for the assessment of the appearance and the degree of the function of interest, although the assessment varies depending on the damaged tissue.
  • somatic stem cells used in the invention include, but are not limited to, stem cells from the dermal system, the digestive system, the bone marrow system, the nervous system, etc.
  • somatic stem cells in the dermal system include epidermal stem cells, hair follicle stem cells, etc.
  • somatic cells in the digestive system include pancreatic (common) stem cells, hepatic stem cells, etc.
  • somatic cells in the bone marrow system include hematopoietic stem cells, mesenchymal stem cells, etc.
  • somatic stem cells in the nervous system include neural stem cells, retinal stem cells, etc. Somatic cells used in the invention may be naturally-occurring or genetically-modified as long as they can achieve the intended treatment.
  • the origins of stem cells are classified into ectoderm, endoderm and mesoderm.
  • Stem cells of ectodermal origin are present mainly in the brain, and include neural stem cells.
  • Stem cells of endodermal origin are present mainly in the bone marrow, and include blood vessel stem cells, hematopoietic stem cells, mesenchymal stem cells, etc.
  • Stem cells of mesoderm origin are present mainly in organs, and include hepatic stem cells, pancreatic stem cells, etc.
  • somatic stem cells which may be derived from any mesenchyme, more preferably somatic stem cells derived from dental pulp, and most preferably somatic stem cells derived from human exfoliated deciduous teeth.
  • Somatic stem cells from mesenchyme may produce various cytokines such as vascular endothelial growth factor (VEGF), hepatocyte growth factor (HGF), insulin-like growth factor (IGF), platelet-derived growth factor (PDGF), transforming growth factor- ⁇ (TGF- ⁇ )-1 and -3, TGF- ⁇ , KGF, HBEGF and SPARC.
  • VEGF vascular endothelial growth factor
  • HGF hepatocyte growth factor
  • IGF insulin-like growth factor
  • PDGF platelet-derived growth factor
  • TGF- ⁇ transforming growth factor- ⁇ -1 and -3
  • TGF- ⁇ KGF
  • HBEGF HBEGF
  • SPARC transforming growth factor- ⁇
  • the stem cell-conditioned medium preferably includes at least two cytokines, and more preferably includes a combination of two or more selected from the group consisting of vascular endothelial growth factor (VEGF), hepatocyte growth factor (HGF), insulin-like growth factor (IGF), platelet-derived growth factor (PDGF) and transforming growth factor ⁇ (TGF- ⁇ ).
  • VEGF vascular endothelial growth factor
  • HGF hepatocyte growth factor
  • IGF insulin-like growth factor
  • PDGF platelet-derived growth factor
  • TGF- ⁇ transforming growth factor ⁇
  • the mixture of cytokines for use in the invention may be used as a part of the stem cell-conditioned medium or as a mixture of cytokines that has been isolated from the stem cell-conditioned medium.
  • a part of the cytokines may be replaced with one or more known corresponding cytokine.
  • the stem cell-conditioned medium for use in the invention is preferably obtained from a culture of somatic stem cells derived from the same individual as that having the target tissue, in order to avoid rejection.
  • the target tissue may be the same as or different from a tissue from which the somatic stem cell used to obtain the stem cell-conditioned medium is derived.
  • a stem cell-conditioned medium used in the invention is not limited to a stem cell-conditioned medium obtained from culturing somatic stem cells, and may contain a stem cell-conditioned medium obtained from culturing embryonic stem cells (ES cells), induced pluripotent stem cells (iPS cells), embryonal carcinoma cells (EC cells) or the like.
  • ES cells embryonic stem cells
  • iPS cells induced pluripotent stem cells
  • EC cells embryonal carcinoma cells
  • the somatic stem cell-conditioned medium is a medium obtained by culturing somatic stem cells, and does not include the cells themselves.
  • the conditioned medium that can be used in the invention can be obtained by, for example, removing cell components by separation after culturing.
  • the conditioned medium may be subjected to various treatments (such as centrifugation, concentration, solvent substitution, dialysis, freezing, drying, freeze-drying, dilution, desalting or storage), as appropriate, before use.
  • the stem cells for obtaining the stem cell-conditioned medium can be selected by an ordinary method, and can be selected based on the size and morphology of cells, or as adhesive cells.
  • the stem cells can be selected as adhesive cells from dental pulp cells obtained from exfoliated deciduous teeth or permanent teeth, or as subcultured cells thereof, as described below.
  • the later-described method of producing a CNS disease treatment composition can preferably be used as a method of producing the damaged part treatment composition.
  • the dental pulp stem cell-conditioned medium to be used may be a conditioned medium obtained by culturing the selected stem cells.
  • the stem cell-conditioned medium can be obtained after obtaining target stem cells from a tissue that may contain the target stem cells in a similar manner.
  • the “stem cell-conditioned medium” is defined as a medium that is obtained by culturing stem cells, and that does not include cells themselves.
  • the composition of the invention includes the “stem cell-conditioned medium” as an active ingredient.
  • the composition as a whole does not include any cells (regardless of the type of cells).
  • the composition according to this aspect is clearly distinguished from the stem cells themselves as a matter of course, and from various compositions that contain stem cells, due to the feature described above.
  • a typical example of this aspect is a composition that does not include any stem cells, and that consists only of the stem cell-conditioned medium.
  • a basal medium or a medium obtained by adding serum or the like to a basal medium, can be used for the stem cell culture medium.
  • a serum-free “dental pulp stem cell-conditioned medium” it is preferable to use a serum-free medium throughout the entire process or to use a serum-free medium at subculturing for the last passage, or for the last few passages.
  • DMEM Iscove's Modified Dulbecco's Medium
  • HamF12 Ham's F12 medium (HamF12) (Sigma-Aldrich Corporation, GIBCO Corporation, etc.
  • RPMI1640 medium etc.
  • Two or more basal media may be used in combination.
  • a mixed medium is a medium formed by mixing equivalent amounts of IMDM and HamF12 (commercially available as, for example, IMDM/HamF12 (tradename, GIBCO Corporation)).
  • ingredients that can be added to the medium include serums (such as fetal bovine serum, human serum and sheep serum), serum replacements (knockout serum replacement (KSR), etc.), bovine serum albumin (BSA), antibiotics, various vitamins and various minerals.
  • stem cell-conditioned medium For the cultivation of stem cells, usually-employed conditions can be applied as they are.
  • the method for producing a stem cell-conditioned medium may be the same as the later-described method of producing a CNS disease treatment composition, except for appropriately modifying the step of isolation and selection of stem cells in accordance with the type of stem cells. Those skilled in the art would be able to appropriately carry out the isolation and selection of stem cells in accordance with the type of stem cells.
  • the target tissue in the invention is not particularly limited, and examples thereof include skin, bone, periodontal tissue, brain, etc.
  • the composition of the invention is effective for repairing such target tissues.
  • FIG. 9 shows a conceptual diagram of the mechanism of bone regeneration using the composition of the invention.
  • the composition of the invention is also effective for the treatment of disorders related to tissue damage.
  • disorders include cerebral infarction, periodontal disease, spinal cord injury, skin ulceration, osteoporosis, etc.
  • the composition of the invention is a composition for treatment of cerebral infarction, periodontal disease, spinal cord injury, skin ulceration, osteoporosis, etc., and includes a stem cell-conditioned medium obtained by culturing somatic stem cells.
  • the damaged part treatment composition of the invention is used as a composition for treatment of a damaged part, such as treatment of a damage to skin, periodontal tissue or bone, treatment of cerebral infarction or treatment of CNS disease.
  • the dosage of the damaged part treatment composition may be any therapeutically effective amount.
  • the damaged part treatment composition which includes the stem cell-conditioned medium as an active ingredient
  • the damaged part treatment composition may be used after concentrating the active ingredient as described below.
  • ingredients may additionally be used in the composition of the invention in accordance with the state of the subject to which the composition is applied, as long as the expected therapeutic effect is maintained.
  • ingredients that can additionally be used in the invention include the following:
  • Hyaluronic acid, collagen, fibrinogen (for example, BOLHEAL (registered trademark)), etc. may be used as organic bioabsorbable materials.
  • Gelling materials for use preferably have high bioaffinity, and hyaluronic acid, collagen, fibrin adhesive or the like may be used.
  • hyaluronic acids and collagens may be selected and used, and it is preferable to adopt those suitable for the purpose of application of the composition of the invention (the tissue to which the composition is to be applied).
  • Collagens to be used are preferably soluble (acid-soluble collagens, alkali-soluble collagens, enzyme-solubilized collagens, etc.).
  • Other pharmaceutically-acceptable ingredients may be contained.
  • Lactose, starch, sorbitol, D-mannitol, white sugar, etc. may be used as excipients.
  • Starch, carboxymethylcellulose, calcium carbonate, etc. may be used as disintegrants.
  • Phosphoric acid salts, citric acid salts, acetic acid salts, etc. may be used as buffering agents.
  • Gum arabic, sodium alginate, Tragacanth, etc. may be used as emulsifying agents.
  • Glycerin monostearate, aluminum monostearate, methylcellulose, carboxymethylcellulose, hydroxymethylcellulose, sodium lauryl sulfate, etc. may be used as suspending agents.
  • Benzyl alcohol, chlorobutanol, sorbitol, etc. may be used as soothing agents.
  • Propyleneglycol, ascorbic acid, etc. may be used as stabilizers.
  • Phenol, benzalkonium chloride, benzylalcohol, chlorobutanol, methylparaben, etc. may be used as preservatives.
  • Benzalkonium chloride, parahydroxybenzoic acid, chlorobutanol, etc. may be used as antiseptic agents.
  • Antibiotics, pH adjusting agents, growth factors such as epidermal growth factor (EGF), nerve growth factor (NGF) and brain-derived neurotrophic factor (BDNF)
  • growth factors such as epidermal growth factor (EGF), nerve growth factor (NGF) and brain-derived
  • the final form of the composition of the invention is not particularly limited.
  • Examples of the form include liquid forms (such as a purely liquid form and a gel form), and solid forms (such as a powdery form, a fine grain form and a granular form).
  • aspects of the invention include a method of repairing a damaged part of a target tissue and a method of treating a damaged tissue. These methods include administering the stem cell-conditioned medium to the damaged part of the target tissue or the damaged tissue. Due to the administering, the target tissue having the damaged part can effectively be repaired. In particular, in a case in which the target tissue is brain, the methods can preferably applied as a method of treating cerebral infarction.
  • the method and route of the administration of the damaged part treatment composition are not particularly limited.
  • the damaged part treatment composition is preferably administered parenterally, and the parenteral administration may be systemic administration or topical administration.
  • topical administration include injection, application or spraying to the target tissue, etc.
  • the method of administering the damaged part treatment composition include intravenous administration, intraarterial administration, intraportal administration, intradermal administration, subcutaneous administration, intramuscular administration, intraperitoneal administration, intranasal administration, etc.
  • intranasal administration or the like is preferable due to its low invasiveness.
  • the dosage regimen may be, for example, from once to several times a day, once every two days, once every three days, or the like.
  • the dosage regimen may be prepared in consideration of the sex, age, weight, pathological condition, etc. of the subject (recipient).
  • the selection of the administration method may be carried out by a person skilled in the art, based on the type of target tissue, the type of disease to be treated, etc.
  • application of intranasal administration or the like is particularly preferable for, for example, the treatment of a disorder or repair of a damaged tissue of which target tissue is located in the brain, because the intranasal administration is less invasive and free from the need to consider the passage through the blood-brain barrier.
  • intranasal administration may preferably be applied in a case in which the target tissue is brain.
  • Intranasal administration may preferably be applied to treatment of cerebral infarction.
  • the subject to which the damaged part treatment composition is administered is typically a human patient having damage in the target tissue.
  • mammals other than human including pet animals, farm animals and laboratory animals, specific examples of which include mice, rats, guinea pigs, hamsters, monkeys, cattle, pigs, goats, sheep, dogs, cats, etc.
  • mammals other than human including pet animals, farm animals and laboratory animals, specific examples of which include mice, rats, guinea pigs, hamsters, monkeys, cattle, pigs, goats, sheep, dogs, cats, etc.
  • the method of treating cerebral infarction of the invention includes intranasally administering the stem cell-conditioned medium, to repair a damaged part of the brain. According to this treatment method, a region that was damaged by cerebral infarction can effectively be restored with less invasiveness.
  • aspects of the invention encompass, particularly, a CNS disease treatment composition and a method of treating a CNS disease.
  • dental pulp stem cells are a unique group of cells that coexpress all neural lineage markers including neural stem cell markers, differentiated neural cell markers, astrocyte markers and oligodendrocyte markers, and that dental pulp stem cells highly express brain-derived neurotrophic factor (BDNF), and have also demonstrated, through animal experiments, that dental pulp stem cells induce nerve regeneration (see Japanese Patent Application No. 2010-92585).
  • BDNF brain-derived neurotrophic factor
  • the inventors thus far looked for the potential capacity of dental pulp stem cells (SHED, DPSCs), and studied the utility thereof as cells from various viewpoints.
  • SHED dental pulp stem cells
  • DPSCs dental pulp stem cells
  • the inventors have drastically changed their viewpoint, and carried out various experiments focusing on a dental pulp stem cell-conditioned medium.
  • peripheral nerves easily regenerate after being damaged, but central nerves (brain, spinal cord) rarely regenerate.
  • the biggest reason why the central nerve regeneration does not occur is the presence of various factors that inhibit outgrowth of regenerated axons in the CNS after being damaged.
  • CSPG Activated astrocyte-derived chondroitin sulfate proteoglycan
  • MAG myelin-associated glycoprotein
  • nerve regeneration inhibitory factors have thus far been identified as nerve regeneration inhibitory factors. These inhibitory substances inhibit neuronal axon outgrowth via activation of intracellular protein Rho or ROCK, and induce apoptosis. No agent has been found which inhibits apoptosis even in the presence of nerve regeneration inhibitory factor, and which exerts axon elongation effect. Analysis by the inventors revealed a surprising fact that a dental pulp stem cell-conditioned medium inhibits the action of nerve regeneration inhibitory substances (cancels the inhibition), promotes outgrowth of neurites, and suppresses apoptosis even in the environment of a damaged CNS (i.e., the environment in which substances that inhibit outgrowth of neurites and induce apoptosis are present).
  • the inventors further studied the activity of the dental pulp stem cell-conditioned medium using model animals with injured spinal cord, as a result of which the administration of the dental pulp stem cell-conditioned medium remarkably improved the motor function of hindlimbs. Further, as a result of histological evaluation, the administration of the dental pulp stem cell-conditioned medium suppressed morphological alteration of the spinal cord and enlargement of nerve injury. As discussed above, excellent regenerative and therapeutic effects of the dental pulp stem cell-conditioned medium were confirmed also by animal experiments.
  • the dental pulp stem cell-conditioned medium is quite effective for regeneration and healing of the CNS.
  • the invention as discussed below is mainly based on this finding.
  • the dental pulp stem cell-conditioned medium is more advantageous than a case in which dental pulp stem cells themselves are used, in terms of advance preparation and storage, and the dental pulp stem cell-conditioned medium is particularly suitable for the treatment of the acute or subacute phase of CNS diseases.
  • the utility of the dental pulp stem cell-conditioned medium is quite high also in the sense that the dental pulp stem cell-conditioned medium does not include any cellular components and is capable of overcoming the immune rejection problem.
  • the present aspect of the invention includes the following:
  • a CNS disease treatment composition including a dental pulp stem cell-conditioned medium.
  • the CNS disease treatment composition according to any one of [1] to [9], wherein the conditioned medium is a conditioned medium obtained by culturing adhesive cells in dental pulp cells or subcultured cells thereof
  • the CNS disease treatment composition according to any one of [1] to [9], wherein the CNS disease is a disease or disorder selected from the group consisting of neurodegenerative diseases such as spinal cord injury, amyotrophic lateral sclerosis, Alzheimer's disease, Parkinson's disease, progressive supranuclear palsy, Huntington's disease, multiple system atrophy and spinocerebellar ataxia, degeneration or loss of neuronal cells caused by cerebral ischemia, intracerebral hemorrhage or cerebral infarction and a retinal disease involving a neuronal cell disorder.
  • neurodegenerative diseases such as spinal cord injury, amyotrophic lateral sclerosis, Alzheimer's disease, Parkinson's disease, progressive supranuclear palsy, Huntington's disease, multiple system atrophy and spinocerebellar ataxia
  • degeneration or loss of neuronal cells caused by cerebral ischemia, intracerebral hemorrhage or cerebral infarction and a retinal disease involving a neuron
  • a method of producing a CNS disease treatment composition including the following steps (1) to (3):
  • the CNS disease is a disease or disorder selected from the group consisting of spinal cord injury, neurodegenerative diseases such as amyotrophic lateral sclerosis, Alzheimer's disease, Parkinson's disease, progressive supranuclear palsy, Huntington's disease, multiple system atrophy and spinocerebellar ataxia, degeneration or loss of neuronal cells caused by cerebral ischemia, intracerebral hemorrhage or cerebral infarction and a retinal disease involving a neuronal cell disorder.
  • neurodegenerative diseases such as amyotrophic lateral sclerosis, Alzheimer's disease, Parkinson's disease, progressive supranuclear palsy, Huntington's disease, multiple system atrophy and spinocerebellar ataxia
  • degeneration or loss of neuronal cells caused by cerebral ischemia intracerebral hemorrhage or cerebral infarction
  • a retinal disease involving a neuronal cell disorder.
  • a method of treating a CNS disease including a step of administering the CNS disease treatment composition of any one of [1] to [10] to a CNS disease patient, in a therapeutically effective amount.
  • dental pulp stem cell is an undifferentiated dental pulp stem cell that has not been subjected to differentiation-inducing treatment after obtainment thereof, or a differentiation-induced dental pulp stem cell that has been induced to differentiate into a neural cell after obtainment thereof
  • a method of determining whether or not a prepared dental pulp stem cell-conditioned medium is effective as an active ingredient of the CNS disease treatment composition comprising the following step (a) and/or (b):
  • the CNS disease treatment composition of the invention includes a dental pulp stem cell-conditioned medium.
  • Dental pulp stem cells are roughly classified into two types—dental pulp stem cells from deciduous teeth and permanent teeth dental pulp stem cells.
  • dental pulp stem cells from deciduous teeth are abbreviated to SHED
  • dental pulp stem cells from DPSCs are abbreviated to DPSCs, in accordance with customary practices.
  • SHED-conditioned medium and DPSC-conditioned medium can be used as a conditioned medium for forming the CNS disease treatment composition.
  • the CNS disease treatment composition can be characterized by the feature—“exhibiting a neurite outgrowth activity in the presence of nerve regeneration inhibitory substance”.
  • nerve regeneration inhibitory substances are present in the CNS. This is an important point when CNS disease therapy (mainly nerve regeneration) is planned, and needs consideration.
  • CNS disease treatment composition having the feature described above allows for suppression of the action of nerve regeneration inhibitory substances, and promotion of nerve regeneration.
  • the nerve regeneration inhibitory substances are chondroitin sulfate proteoglycan (CSPG) and myelin-associated glycoprotein (MAG).
  • CSPG chondroitin sulfate proteoglycan
  • MAG myelin-associated glycoprotein
  • CSPG or MAG nerve regeneration inhibitory substance
  • a test composition is confirmed to have the above-described feature if neurite outgrowth is observed when the neuronal cells are cultured in the coexistence of the test composition and CSPG or MAG
  • the CNS disease treatment composition can alternatively be characterized by the feature—“exhibiting an apoptosis inhibiting activity toward neuronal cells”. Whether or not the CNS disease treatment composition has this feature can be confirmed by, for example, an in vitro experimentation using neuronal cells (see later-described Examples with respect to the details of the experimentation). A test composition is confirmed to have this feature if cell death due to apoptosis is suppressed when the neuronal cells are cultured in the presence of the test composition. In a preferable aspect, the CNS disease treatment composition has both of this feature and the above-described feature (exhibiting a neurite outgrowth activity in the presence of a nerve regeneration inhibitory substance).
  • the term “dental pulp stem cell-conditioned medium” refers to a medium that is obtained by culturing dental pulp stem cells, and that does not include cell components (i.e., dental pulp cells and dental pulp stem cells). Therefore, a conditioned medium that can be used in the invention can be obtained by, for example, removing cell components by separation after culturing.
  • the conditioned medium may be subjected to various treatments (such as centrifugation, concentration, solvent substitution, dialysis, freezing, drying, freeze-drying, dilution, desalting or storage), as appropriate, before use. Details of treatment methods for the conditioned medium are described later.
  • Dental pulp stem cells can be selected as adhesive cells in dental pulp cells. Therefore, a conditioned medium obtained by culturing adhesive cells in dental pulp cells collected from exfoliated deciduous teeth or permanent teeth, or subcultured cells thereof, can be used as the dental pulp stem cell-conditioned medium. Details of the method of preparing the dental pulp stem cell-conditioned medium are described later.
  • the dental pulp stem cell-conditioned medium is defined as a medium that is obtained by culturing dental pulp stem cells and that does not include cell components.
  • the CNS disease treatment composition includes the dental pulp stem cell-conditioned medium as an active ingredient, and, in one aspect thereof, the composition as a whole does not include any cells (regardless of the type of cells).
  • the composition according to this aspect is clearly distinguished from the dental pulp stem cells themselves as a matter of course, and from various compositions that contain dental pulp stem cells, based on the feature described above.
  • a typical example of this aspect is a composition consisting only of the dental pulp stem cell-conditioned medium.
  • One embodiment of the present aspect has characteristics in that the dental pulp stem cell-conditioned medium and the dental pulp stem cells are used in combination.
  • dental pulp stem cells from deciduous teeth are used in consideration of their higher cell proliferation capacity compared to permanent teeth dental pulp stem cells (DPSCs).
  • SHED are considered to have higher differentiation capacity.
  • a high BDNF expression level of SHED (see, Japanese Patent Application No. 2010-92585), which may provide higher therapeutic effects, is another advantage of using SHED.
  • SHED also has an advantage in that SHED can be easily obtained.
  • compositions of the above-described embodiment in which the dental pulp stem cell-conditioned medium and the dental pulp stem cells are used in combination is literally and practically distinguished from compositions or agents in which the dental pulp stem cells are used as active ingredients with a focus on the utility of dental pulp stem cells themselves, based on the point that the composition of the above-described embodiment includes the dental pulp stem cell-conditioned medium as an essential active ingredient.
  • the embodiment described above is characterized by combined use of the dental pulp stem cell-conditioned medium and the dental pulp stem cells.
  • the expression “combined use” as used herein means that the dental pulp stem cell-conditioned medium and the dental pulp stem cells are used together.
  • the CNS disease treatment composition is provided as a combination preparation in which the dental pulp stem cell-conditioned medium and the dental pulp stem cells are mixed.
  • dental pulp stem cells that have not been subjected to induction of differentiation after obtainment thereof (i.e., dental pulp stem cells that remain undifferentiated; also referred to as “undifferentiated dental pulp stem cells” herein).
  • the CNS disease treatment composition exerts strong nerve protection activity, and is thus suitable to, particularly, application in the acute or subacute phase of CNS diseases (for example, intractable neural diseases involving severe loss or degeneration of neuronal cells, such as spinal cord injury or cerebral infarction).
  • CNS diseases for example, intractable neural diseases involving severe loss or degeneration of neuronal cells, such as spinal cord injury or cerebral infarction.
  • the dental pulp stem cells used in this embodiment are positive for the neural stem cell marker Nestin, positive for the neural stem cell marker Doublecortin, positive for the neuronal call marker ⁇ -III tubulin, positive for the neuronal call marker NeuN, positive for the astrocyte marker GFAP, and positive for the oligodendrocyte marker CNPase, and highly express BDNF (see, Japanese Patent Application No. 2010-92585).
  • the CNS disease treatment composition may also be provided in the form of, for example, a kit composed of a first constituent element containing the dental pulp stem cell-conditioned medium and a second constituent element containing the dental pulp stem cells.
  • the second constituent element is administered simultaneously with the administration of the first constituent element or after the administration of the first constituent element.
  • a regimen in which the first constituent element and the second constituent element are simultaneously administered is particularly suitable for application in the acute or subacute phase of CNS diseases.
  • the term “simultaneously” does not require exact simultaneity. Accordingly, the concept of “simultaneously” encompasses a case in which both elements are administered with no time lag such as administration to the subject after mixing of both constituent elements, as well as a case in which both constituent elements are administered with substantially no time lag such as administration of one of the constituent elements immediately after the administration of the other one of the constituent elements.
  • the first constituent element is administered in the acute or subacute phase, and the second constituent element is administered thereafter (for example, 3 days to 1 week after the administration of the first constituent element), continuous and comprehensive therapeutic effects can be expected.
  • this regimen it is preferable to use dental pulp stem cells that have been induced to differentiate into neural cells (here also referred to as “differentiation-induced dental pulp stem cells”) as an active ingredient of the second constituent element.
  • neural cells encompasses motor neurons, dopamine-producing cells, various CNS cells, astrocytes, oligodendrocytes and Schwann cells.
  • the type of neural cells into which the dental pulp stem cells are to be induced to differentiate may be determined in consideration of the disease and pathological condition of the subject to be treated. For example, for the treatment of spinal cord injury, dental pulp stem cells that have been induced to differentiate into mature nerve cells, oligodendrocytes or Schwann cells may be used in the second constituent element. An example of a method of inducing neural differentiation is described below.
  • a method composed of the following two steps may be used for induction of differentiation into dopamine-producing neuronal cells.
  • dental pulp stem cells are cultured for 2 to 3 days in, for example, a DMED medium that contains 12.5 U/mL Nystatin, N2 supplement, 20 ng/mL bFGF and 20 ng/mL EGF, using a dish coated with poly-L-lysine.
  • a DMED medium that contains 12.5 U/mL Nystatin, N2 supplement, 20 ng/mL bFGF and 20 ng/mL EGF
  • the cells after the first step are cultured for 6 to 7 days in, for example, a NeurobasalTM medium that contains B27 supplement, 1 mM db-cAMP, 0.5 mM IBMX, 200 ⁇ M ascorbic acid and 50 ng/mL BDNF.
  • the induced dopamine-producing neuronal cells can be confirmed by immunostaining using an anti-tyrosine hydroxylase antibody.
  • various methods that have been reported as methods for inducing differentiation of neural stem cells or embryonic stem cells into dopamine-producing neuronal cells such as a method of culturing in the presence of bFGF followed by floating culture of aggregates (Studer, L. et al.: Nat.
  • Neurosci., 1: 290-295, 1998) a method of culturing in the presence of bFGF and glia cell-conditioned medium (Daadi, M. M. and Weiss, S. J.: Neuroscience, 19: 4484-4497, 1999), a method utilizing FGF8, Shh, bFGF, ascorbic acid, etc. (Lee, S. H. et al.: Nat. Biotechnol., 18: 675-679, 2000), and a method of co-culturing with bone marrow stromal cells (Kawasaki, H. et al.: Neuron, 28: 31-40, 2000), may be utilized after appropriate modification thereof, if necessary.
  • a method composed of the following two steps may be used for induction of astrocyte differentiation.
  • dental pulp stem cells are cultured for four days in, for example, a DMEM/F12 medium that contains N2 supplement and 10 ng/mL bFGF, using a dish doubly coated with poly-L-ornithine and fibronectin.
  • the cells are cultured for three days in the medium further added with 80 ng/mL LIF and 80 ng/mL BMP2.
  • the differentiation induced astrocytes can be confirmed by immunostaining using an anti-GFAP antibody.
  • a method composed of the following two steps may be used for induction of oligodendrocyte differentiation. Similar to the induction of astrocyte differentiation, in the first step, dental pulp stem cells are cultured for four days in, for example, a DMEM/F12 medium that contains N2 supplement, 10 ng/mL bFGF and 0.5% FCS, using a dish doubly coated with poly-L-ornithine and fibronectin. As a result of this step, the dental pulp stem cells are induced into oligodendrocyte progenitor cells.
  • the cells are cultured for four days in a DMEM/F12 medium that contains 20 ng/mL T3 (Triiodothyronine), 20 ng/mL T4 (Thyroxine) and N2 supplement.
  • T3 Triiodothyronine
  • T4 Thyroxine
  • N2 supplement 20 ng/mL T3 (Triiodothyronine), 20 ng/mL T4 (Thyroxine) and N2 supplement.
  • the differentiation induced oligodendrocytes can be confirmed using an anti-04 antibody.
  • pluripotent stem cells that have been induced to differentiate into neural cells may be used in addition to, or in place of, the differentiation induced dental pulp stem cells.
  • pluripotent stem cells include induced pluripotent stem cells (iPS cells) and embryonic stem cells (ES cells).
  • the “induced pluripotent stem cells (iPS cells)” are cells having pluripotency (multipotency), and proliferative capacity that are produced by reprogramming somatic cells by introduction of reprogramming factors.
  • the induced pluripotent stem cells exhibit properties similar to those of ES cells.
  • the iPS cells can be produced by various iPS cell production methods that have been reported thus far. Of course, application of iPS cell production methods that will be developed in the future is also contemplated, as a matter of course.
  • the most basic technique among iPS cell production methods is a method of introducing the four transcriptional factors of Oct3/4, Sox2, KIF4 and c-Myc into a cell using a virus (Takahashi K, Yamanaka S: Cell 126 (4), 663-676, 2006; Takahashi, K, et al: Cell 131 (5), 861-72, 2007).
  • Establishment of human iPS cells by introducing the four factors of Oct4, Sox2, Lin28 and Nonog is also reported (Yu J, et al: Science 318 (5858), 1917-1920, 2007).
  • Establishment of iPS cells by introducing the three factors other than c-Myc (Nakagawa M, et al: Nat. Biotechnol.
  • Cells in which transformation into iPS cells, i.e., reprogramming, has occurred can be selected using, for example, expression of a pluripotent stem cell marker (undifferentiated marker) such as Fbox15, Nanog, Oct/4, Fgf-4, Esg-1 or Cript as an indicator.
  • a pluripotent stem cell marker such as Fbox15, Nanog, Oct/4, Fgf-4, Esg-1 or Cript as an indicator.
  • the selected cells are collected as iPS cells.
  • mouse ES cells include ES-E14TG2a cells (ATCC), ES-D3 cells or the like (ATCC), H1 cells (Riken BioResource Center, Tsukuba-city, Japan), B6G-2 cells (Riken BioResource Center, Tsukuba-city, Japan), R1 cells (Samuel Lunenfeld Research Institute, Toronto, Canada), mouse ES cells (129SV, catalogue number R-CMTI-1-15, R-CMTI-1A) (Dainippon Sumitomo Pharma Co., Ltd., Osaka, Japan) and mouse ES cells (C57/BL6, catalogue number R-CMTI-2A (Dainippon Sumitomo Pharma Co., Ltd., Osaka, Japan).
  • Monkey ES cells are available from, for example, Stem Cell Research Center, Institute for Frontier Medical Sciences, Kyoto University. Human ES cells are available from, for example, Stem Cell Research Center, Institute for Frontier Medical Sciences, Kyoto University, WiCell Research Institute (Madison, USA), and ES Cell International Pte Ltd (Singapore). Methods for establishing ES cells have been achieved, and part thereof has been practice routinely. Therefore, one can himself establish desired ES cells using ordinary methods. For example, Nagy. A. et al.
  • Yodosha Co., Ltd. may be referenced with respect to method for establishing mouse ES cells.
  • Suemori H, Tada T, Torii R, et al., Dev Dyn 222, 273-279, 2001, etc. may be referenced.
  • Method for establishing human ES cells Wassarman, P. M. et al.: Methods in Enzymology, Vol. 365 (2003), etc. may be referenced.
  • the CNS disease treatment composition is preferably free from serum.
  • the absence of serum in the CNS disease treatment composition improves the safety of the composition.
  • a serum-free conditioned medium can be prepared by culturing dental pulp stem cells in a medium that does not contain any serum (serum-free medium).
  • serum-free medium can be obtained by carrying out subculturing for the last passage, or for the last few passages, in a serum-free medium.
  • a serum-free conditioned medium can be obtained also by removing serum from a collected conditioned medium, using, for example, solvent substitution by dialysis or column.
  • the CNS disease treatment composition is utilized for treatment of diseases of central nerves (brain and spinal cord).
  • CNS diseases to which the CNS disease treatment composition can be applied include spinal cord injury, neurodegenerative diseases such as amyotrophic lateral sclerosis, Alzheimer's disease, Parkinson's disease, progressive supranuclear palsy, Huntington's disease, multiple system atrophy and spinocerebellar ataxia, degeneration or loss of neuronal cells caused by cerebral ischemia, intracerebral hemorrhage or cerebral infarction, and a retinal disease involving a neuronal cell disorder.
  • CNS disease treatment composition promotes regeneration and healing of CNS tissues due to its neurite outgrowth effects and/or its apoptosis inhibitory effects toward neuronal cells.
  • Any disease or disorder to which treatment based on this mechanism is effective can be the target disease of the invention, regardless of the type or cause (for example, primary cause such as external injury or cerebral infarction or secondary cause such as infection or tumor) of the disease or disorder.
  • Spinal cord injury refers to a state in which the spinal cord is damaged by an external impact or by an internal factor such as a spinal tumor or hernia, and is classified according to complete-type (a state in which the spinal cord is completely severed at a certain point) and incomplete-type (a state in which the function of the spinal cord is partially maintained although the spinal cord is damaged or compressed), based on the degree of the damage.
  • complete-type a state in which the spinal cord is completely severed at a certain point
  • incomplete-type a state in which the function of the spinal cord is partially maintained although the spinal cord is damaged or compressed
  • Spinal cord injury is one of the diseases to which regenerative medicine is expected to be applied, and use of bone marrow, neural stem cells, embryonic stem cells, artificial pluripotent stem cells, etc. is under investigation.
  • a decisive treatment technique has not been realized owing to various problems. Under such a circumstance, the CNS disease treatment composition provides a treatment method that is expected to provide a high therapeutic effect, and the significance and value
  • Cerebral ischemia is a state in which blood supply to the brain is insufficient, and oxygen and nutrients are not sufficiently supplied to the brain. Cerebral ischemia causes the death of neuronal cells and cerebral edema, and serves as a cause of cerebral infarction.
  • the composition of the invention can be applied also to the treatment of destruction of neuronal cells due to cerebral ischemia or the like, or various diseases that accompany the destruction of neuronal cells.
  • Parkinson's disease, spinocerebellar ataxia, Alzheimer's disease, Huntington's disease, multiple system atrophy and progressive supranuclear palsy are intractable neural diseases caused by region-specific neuronal loss in the cerebrum, midbrain and cerebellum regions.
  • the CNS disease treatment composition is able to exert a therapeutic effect by suppressing the neuronal degeneration and loss of in these diseases.
  • the CNS disease treatment composition can also be applied to retinal diseases accompanied by neuronal cell disorders.
  • neuronal cells photoreceptor cells (cone photoreceptor cells, rod photoreceptor cells), bipolar cells, horizontal cells, amacrine cells and ganglion cells—are present in retina.
  • the CNS disease treatment composition exerts a therapeutic effect by suppressing the neuronal death and loss in retinal diseases caused by damage to one type, or two or more types, selected from these neuronal cells present in retina, as well as in retinal diseases with pathological conditions exhibiting damage to one type, or two or more types, selected from these neuronal cells, example of which include traumatic retinal detachment, retinal tear, concussion of retina, optic canal fracture, diabetic retinopathy, age-related macular degeneration, retinitis pigmentosa, glaucoma, choroideremia, Leber's hereditary optic neuropathy, cone dystrophy, familial drusen, central areolar choroidal dystrophy and autosomal dominant optic atrophy.
  • ingredients may additionally be used in the composition of the invention, as long as the expected therapeutic effect is maintained.
  • Ingredients that can additionally be used in the invention include those listed below.
  • Hyaluronic acid, collagen, fibrinogen (for example, BOLHEAL (registered trademark)), etc. may be used as organic bioabsorbable materials.
  • Gelling materials for use preferably have high bioaffinity, and hyaluronic acid, collagen or fibrin adhesive or the like may be used.
  • hyaluronic acids and collagens may be selected and used, and it is preferable to adopt those suitable for the purpose of application of the composition of the invention (the tissue to which the composition is to be applied).
  • Collagens to be used are preferably soluble (acid-soluble collagens, alkali-soluble collagens, enzyme-solubilized collagens, etc.).
  • Other pharmaceutically-acceptable ingredients may be contained.
  • Lactose, starch, sorbitol, D-mannitol, white sugar, etc. may be used as excipients.
  • Starch, carboxymethylcellulose, calcium carbonate, etc. may be used as disintegrants.
  • Phosphoric acid salts, citric acid salts, acetic acid salts, etc. may be used as buffering agents.
  • Gum arabic, sodium alginate, Tragacanth, etc. may be used as emulsifying agents.
  • Glycerin monostearate, aluminum monostearate, methylcellulose, carboxymethylcellulose, hydroxymethylcellulose, sodium lauryl sulfate, etc. may be used as suspending agents.
  • Benzyl alcohol, chlorobutanol, sorbitol, etc. may be used as soothing agents.
  • Propyleneglycol, ascorbic acid, etc. may be used as stabilizers.
  • Phenol, benzalkonium chloride, benzylalcohol, chlorobutanol, methylparaben, etc. may be used as preservatives.
  • Benzalkonium chloride, parahydroxybenzoic acid, chlorobutanol, etc. may be used as antiseptic agents.
  • Antibiotics, pH adjusting agents, growth factors (such as nerve growth factor (NGF), brain-derived neurotrophic factor (BDNF)), etc. may also be contained.
  • the final form of the CNS disease treatment composition is not particularly limited.
  • Examples of the form include liquid forms (such as a purely liquid form and a gel form), and solid forms (such as a powdery form, a fine grain form and a granular form).
  • Methods for producing the CNS disease treatment composition are not particularly limited.
  • a production method that includes the following steps (1) to (3) is preferable:
  • dental pulp stem cells which are adhesive cells, are selected from dental pulp cells.
  • the dental pulp cells may be prepared by isolating from a living organism in advance. This selection step may include preparing dental pulp cells. A specific example of a procedure of a series of operations from the preparation of dental pulp cells to the selection of dental pulp stem cells is described below.
  • a naturally-exfoliated deciduous tooth (or extracted deciduous tooth or permanent tooth) is disinfected using a chlorhexidine or ISODINE solution, and, thereafter, the tooth crown part is divided, and a dental pulp tissue is collected using a dental reamer.
  • the collected dental pulp tissue is suspended in a basal medium (Dulbecco's Modified Eagle's Medium containing 10% bovine serum and an antibiotic), and treated with 2 mg/mL collagenase and DISPASE at 37° C. for 1 hour.
  • the dental pulp cells after the enzymatic treatment are collected by centrifugation (5,000 rpm) for 5 minutes.
  • Cell separation using a cell strainer should basically not be carried out since it decreases the collection efficiency of neural stem cell fraction such as SHED or DPSC.
  • the cells are re-suspended in 4 cc of the basal medium, and seeded in a culture dish for adhesive cells having a diameter of 6 cm.
  • a medium for example, Dulbecco's Modified Eagle's Medium (DMEM) containing 10% FCS
  • the cells are cultured in an incubator maintained at 5% CO 2 and 37° C. for about two weeks.
  • the cells are washed with, for example, PBS for once or a few times.
  • This operation may be replaced by harvesting adhesive cells (dental pulp stem cells) that have formed colonies.
  • treatment with 0.05% trypsin-EDTA is carried out at 37° C. for 5 minutes, and cells that have detached from the dish are harvested.
  • step (2) following step (1) the selected adhesive cells are cultured.
  • the cells are seeded in a culture dish for adhesive cells, and cultured in an incubator maintained at 5% CO 2 and 37° C.
  • Subculture is carried out, if necessary.
  • subconfluence the state in which cells cover about 70% of the surface area of the culture vessel
  • confluence the state in which cells cover about 70% of the surface area of the culture vessel
  • the cells are detached from the culture vessel and harvested, and seeded again into a culture vessel filled with a culture medium.
  • Subculture may be carried out repeatedly.
  • subculture may be carried out for one to eight passages, thereby allowing the cells to proliferate to the required cell number (for example, about 1 ⁇ 10 7 cells/mL).
  • the detachment of cells from the culture vessel can be carried out using an ordinary method such as treatment with trypsin.
  • the cells may be harvested and stored (in which the storage condition may be, for example, ⁇ 198° C.).
  • Cells collected from various donors may be stored in the form of a dental pulp stem cell bank.
  • the medium may be, for example, a basal medium or a basal medium supplemented with serum or the like.
  • a serum-free medium may be used throughout the entire process or at subculturing for the last passage or for the last few passages.
  • DMEM Iscove's Modified Dulbecco's Medium
  • HamF12 Ham's F12 medium (HamF12) (Sigma-Aldrich Corporation, GIBCO Corporation, etc.)
  • RPMI1640 medium etc.
  • Two or more basal media may be used in combination.
  • a mixed medium is a medium formed by mixing equivalent amounts of IMDM and HamF 12 (commercially available as, for example, IMDM/HamF12 (tradename, GIBCO Corporation)).
  • ingredients that can be added to the medium include serums (such as fetal bovine serum, human serum and sheep serum), serum replacements (knockout serum replacement (KSR), etc.), bovine serum albumin (BSA), antibiotics, various vitamins and various minerals.
  • step (3) following step (2) the conditioned medium from the dental pulp stem cells selected and cultured by the above-described method is collected.
  • the conditioned medium can be collected by suctioning the culture medium using a dropper of a pipette.
  • the collected conditioned medium is used as an active ingredient of the composition of the invention, directly or after being subjected to one or more treatments.
  • the treatments include centrifugation, concentration, solvent substitution, dialysis, freezing, drying, freeze-drying, dilution, desalting and storage (for example, 4° C. or ⁇ 80° C.).
  • the dental pulp stem cell-conditioned medium exhibited the expected activity (neurite outgrowth activity and apoptosis inhibitory activity) even without complex high purification, as shown in the later-described Examples.
  • the absence of the necessity for complex purification step is advantageous also in that a decrease in activity caused by purification can be avoided.
  • the collected conditioned medium may be subjected to the following step (a) or step (b), or both.
  • a conditioned medium that exhibited a positive result in step (a) is expected to provide an excellent therapeutic effect by its neurite outgrowth activity.
  • a conditioned medium that exhibited a positive result in step (b) is expected to provide an excellent therapeutic effect by its apoptosis inhibitory activity toward neuronal cells. It is preferable to carry out both of step (a) and step (b), and use a conditioned medium that exhibited a positive result in both steps as an active ingredient of the composition of the invention.
  • Methods for checking in steps (a) and (b) are as described above (in the section discussing the first aspect of the invention).
  • the quality of a collected, prepared or stored conditioned medium can also be checked through steps (a) and (b). Therefore, it is understood that these steps themselves have high utility and value as a method of determining the quality of a dental pulp stem cell-conditioned medium (i.e., as a means for determining the suitability as an active ingredient for CNS disease treatment).
  • physiologically active substances contained in the stem cell-conditioned medium can be formulated as a drug.
  • a nerve regenerative active substance to be formulated as a drug.
  • methods usually employed for this purpose may be applied. Examples of the concentration method include the following two methods.
  • the conditioned medium is concentrated (up to 75-fold) using an AMICON ULTRA CENTRIFUGAL FILTER UNITS-10K (manufactured by Millipore Corporation). Specific operation procedure thereof is as described below.
  • the conditioned medium is concentrated (up to 10-fold) using an ethanol precipitation method.
  • Specific protocol thereof is as follows.
  • the stem cell-conditioned medium in the composition of the invention may be freeze-dried. This provides excellent storage stability.
  • the method of freeze-drying the stem cell-conditioned medium may be any method usually employed for this purpose. Examples of the freeze-drying method include the following method:
  • a further aspect of the invention provides a method of treating a CNS disease which includes a step of administering a therapeutically effective amount of the CNS disease treatment composition to a CNS disease patient.
  • the administration route of the composition of the invention is not particularly limited as long as the composition is delivered to the target tissue.
  • the composition may be applied, for example, by topical administration. Examples of the topical administration include injection into the target tissue or application to the target tissue.
  • the composition of the invention may be administered by intravenous administration, intraarterial administration, intraportal administration, intradermal administration, subcutaneous administration, intramuscular administration or intraperitoneal administration.
  • the dosage regimen may be, for example, from once to several times a day, once every two days, once every three days, or the like.
  • the dosage regimen may be prepared in consideration of the sex, age, weight, pathological condition, etc. of the subject (recipient).
  • the subject to which the composition of the invention is administered is typically a human patient suffering from a CNS disease.
  • mammals other than human including pet animals, farm animals and laboratory animals, specific examples of which include mice, rats, guinea pigs, hamsters, monkeys, cattle, pigs, goats, sheep, dogs, cats, etc.
  • the composition of the invention is administered preferably to a subject in the acute or subacute phase, so that the effects of the composition of the invention are most exerted.
  • dental pulp stem cells may be administered to the same subject, thereby providing a complex or continuous effect, according to an embodiment of the invention.
  • undifferentiated dental pulp stem cells that have not been subjected to differentiation inducing treatment after obtainment thereof, or differentiation-induced dental pulp stem cells that have been induced to differentiate into a neural cell after obtainment thereof, may be used as the dental pulp stem cells.
  • differentiation-induced dental pulp stem cells that have been induced to differentiate into neural cells. It is also possible to use pluripotent stem cells (such as iPS cells or ES cells) that have been induced to differentiate into neural cells in addition to, or in place of, the differentiation-induced dental pulp stem cells.
  • pluripotent stem cells such as iPS cells or ES cells
  • the pulp was gently removed and digested in a solution of 3 mg/mL collagenase type I and 4 mg/mL dispase at 37° C. for 1 hour.
  • DMEM Dulbecco's Modified Eagle Medium
  • mesenchymal cell growth supplement Lonza Inc., Walkersville, Md.
  • antibiotics 100 U/mL penicillin, 100 mg/mL streptomycin and 0.25 mg/mL amphotericin B; GIBCO
  • the cells were subcultured at about 1 ⁇ 10 4 cells/cm 2 . Cells passaged from once to three times were used in the experiments.
  • Human BMMSCs were purchased from Lonza Inc., and cultured according to the manufacturer's instructions.
  • SHED, DPSCs and BMSCs were fixed with 3% paraformaldehyde, and then rinsed twice with phosphate-buffered saline, and treated with 100 mM glycine for 20 minutes. Cells were then permeabilized with 0.2% Triton-X (Sigma-Aldrich, St. Louis, Mo.) for 30 minutes, and subsequently incubated in a mixture of 5% donkey serum and 0.5% bovine serum albumin for 20 minutes.
  • Triton-X Sigma-Aldrich, St. Louis, Mo.
  • the cells were incubated with a mouse anti-human STRO-1 antibody (1:100; R&D, Minneapolis, Minn.) as a primary antibody for 1 hour, incubated for 30 minutes with a goat anti-mouse immunoglobulin M-FITC antibody (1:500; Southern Biotech, Birmingham, Ala.) as a secondary antibody, and mounted using Vectashield with DAPI (Vector Laboratories Inc, Burlingame, Calif.).
  • mice Five-week-old female hairless mice (Hos: HR-1) were provided from SLC Inc. (Shizuoka, Japan). All mice were housed in climate-controlled quarters (22 ⁇ 1° C. at 50% humidity) with a 12/12-hour light/dark cycle. Animals were allowed free access to water and a chow diet, and were observed daily. The mice were irradiated dorsally using a UVB-emitting system RMX-3W (Handok Biotech, Seoul, Korea) for eight weeks, five times a week.
  • RMX-3W UVB-emitting system
  • a bank of 10 Toshiba SE lamps was used without any filtering for UVB (peak of emission being about 312 nm, and the irradiance between 290 and 320 nm corresponding to 55% of the total amount of UVB).
  • the distance from the lamps to the animals' backs was 89 cm.
  • the irradiation dose was 1 MED (minimal erythemal dose; 60 mJ/cm 2 ) in the first two weeks, 2 MEDs (120 mJ/cm 2 ) in the third week, 3 MEDs (180 mJ/cm 2 ) in the forth week, and 4 MEDs (240 mJ/cm 2 ) in the fifth through eight weeks.
  • the total UVB dose was approximately 115 MEDs (6.9 J/cm 2 ).
  • SH-CM 100% was subcutaneously injected into the restricted area of the mice.
  • PBS-suspended SHED 4 ⁇ 10 5
  • the dermis was treated by PBS only.
  • SHED 4 ⁇ 10 5 cells were cultured in DMEM/F12 (Invitrogen-Gibco-BRL, Grand Island, N.Y.) serum-free medium. Conditioned medium of SHED was collected after 72 hours of culture, centrifuged at 300 ⁇ g for 5 min, and filtered using a 0.22 mm syringe filter.
  • Dorsal skins (1 cm ⁇ 1 cm) were fixed with a 10% formalin neutral buffered solution, embedded in polyester wax, and sectioned at 6 mm. The sections were subjected to Hematoxylin & Eosin (H&E) staining and Masson's trichrome staining.
  • H&E Hematoxylin & Eosin
  • HDFs were cultured in a DMEM supplemented with 10% fetal bovine serum, 100 U/mL penicillin and 100 mg/mL streptomycin at 37° C. with 5% CO 2 . After starvation with serum-free medium for 24 hours, cells were washed with PBS, and exposed to UVB with 3 to 4 drops of PBS. UVB irradiation was carried out using a UV light source (Waldmann, Schwenningen, Germany). Immediately after the irradiation, the PBS was aspirated, and replaced with a complete medium. UVB irradiation doses were varied in the range of from 50 to 250 mJ/cm 2 during the test, and finally fixed to 70 mJ/cm 2 for further experimentation.
  • HDFs were plated at a density of 5 ⁇ 10 3 cells/well in 96-well plates, and the proliferation of HDFs was measured using a CCK-8 Kit (Dojindo, Gaithersburg, Md.). After starvation for 24 hours in a serum-free medium, the cells were continuously cultured for 24 hours with or without SH-CM, and were exposed to UVB (70 mJ/cm 2 ) for 90 seconds. Then, UVB-irradiated cells were cultured in a complete medium for 24 hours and harvested. HDFs were added to 10 mL of the CCK-8 solution, and incubated for 3 hours. The absorbance was measured at 450 nm using a microplate reader (TECAN, Gro dig, Austria). OD values of each well were converted into their relative cell numbers based on a comparative standard curve.
  • HDFs (2 ⁇ 10 4 cells/well) were seeded in 24-well plates, and pretreated as described above. Then, the cells were lysed in a RIPA buffer (50 mM Tris-HCl, 0.15 M NaCl, 1 mM EDTA, 1% Triton X-100, 1% SDS, 50 mM NaF, 1 mM Na 3 VO 4 , 5 mM dithiothreitol, 1 mg/mL leupeptin and 20 mg/mL PMSF, pH 7.4). Fifty micrograms of proteins were separated on an 8% SDS-polyacrylamide gel by electrophoresis. The proteins were transferred to a PVDF membrane.
  • RIPA buffer 50 mM Tris-HCl, 0.15 M NaCl, 1 mM EDTA, 1% Triton X-100, 1% SDS
  • 50 mM NaF 1 mM Na 3 VO 4
  • 5 mM dithiothreitol 1 mg/m
  • the membrane was incubated with an anti-collagen type I antibody (Santa Cruz, Saint Louis, Mo.) and an anti-matrix metalloproteinase-1 (MMP-1) antibody (Calbiochem, Darmstadt, Germany). Then, the membrane was washed, and incubated with horseradish peroxidase-conjugated anti-goat IgG antibody (1:10,000, Santa Cruz, Saint Louis, Mo.). The blots were reacted with an immunoglobulin western reagent, and were exposed to X-ray film.
  • MMP-1 anti-matrix metalloproteinase-1
  • SHED and DPSCs displayed a fibroblastic morphology resembling BMSCs ( FIG. 2A-C ).
  • Immunofluorescence analysis indicated that SHED, DPSCs and BMSCs contained STRO-1 positive cells ( FIG. 2D-F ).
  • the proliferation rate of SHED was significantly higher than those of DPSCs and BMSCs ( FIG. 2G ).
  • FIG. 3 and FIG. 4 show that repeated SH-CM treatment alleviated the fine wrinkles induced by UVB irradiation.
  • the SHED injected group showed the same tendency as that of the SH-CH group.
  • FIG. 5 shows the histological measurements of the dermal thickness of the hairless mouse skin by H&E staining. Collagen fibers are stained as shown in FIG. 5 , and the degree of staining is remarkably high in the SH-CM treated group (A) and the SHED injected group (B). Measurement of the dermal thickness showed significant increases in the SHED injected group and the SH-CM treated group ( FIG. 6 ). Further, a marked increase of collagen bundles was observed in both groups, but was not observed in the control group ( FIG. 5 ).
  • SH-CM contains diverse growth factors, and in a case in which a unique characteristic of the growth factors is their ability to initiate mitosis of quiescent cells, it is possible that enhanced proliferation by SH-CM in this experiment is mediated by the growth factors secreted from SHED.
  • SHED stem cell-based therapy.
  • STRO-1 positive cells were found in SHED, DPSCs and BMMSCs. STRO-1 is known to recognize a trypsin-resistant cell-surface antigen present on a subpopulation of bone marrow cells, including a predominant proportion of skeletal stem cell having high growth and differentiation potential, and colony forming unit fibroblastic populations. High proliferative capacity is one of the most critical characteristics of postnatal somatic stem cells.
  • SHED shows the highest population rate among SHED, DPSCs and BMSCs. It was previously reported that micro array analysis revealed that SHED expresses multiple growth factors such as FGF, TGF-b, CTGF, NGF and BMP associated with this pathway, at high levels (S. Nakamura, Y. Yamada et al., Stem Cell Proliferation Pathways Comparison between Human Exfoliated Deciduous Teeth and Dental Pulp Stem Cells by Gene Expression Profile from Promising Dental Pulp , JOE, Vol. 35, (11), 1536-1542, 2009).
  • FGF2 was reported as a cytokine that acts to promote the proliferation of numerous kinds of cells and control extracellular matrix generation during tissue regeneration and wound healing.
  • Paracrine factors such as VEGF, KGF or FGF
  • stem cell transplantation is also a “cell-based” cytokine therapy.
  • conditioned media containing growth factors can be used in order to avoid the negative effect of UVB on HDFs.
  • the concept of paracrine effects mediating at least part of the effects of stem cell therapy is not inconsistent with previous data.
  • Cell-based cytokine therapy can provide benefits in wound healing. Keratinocyte differentiation by SHED-derived growth factors may contribute to re-epithelialization in wound closure. Further, SHED-derived growth factors can provide benefits in wound healing, tissue remodeling and skin graft genesis.
  • Photoaging is a complex process having pathologic similarities to skin wounds.
  • MSCs play a key role in this process, and interact with keratinocytes, fat cells and mast cells. MSCs are also a source of extracellular matrix proteins, of which fibrillar type I and type III collagens are significantly reduced in the papillary dermis, and their reduction has been shown to correlate well with the clinical severity of photoaging. This reduction results from a combination of reduced procollagen biosynthesis and increased enzymatic breakdown via the actions of MMP. Fisher et al.
  • UV irradiation induced the synthesis of MMP in human skin in vivo (Phipps R P, Borrello M A, Blieden T M., Fibroblast heterogeneity in the periodontium and other tissues , J Periodontal Res. 1997 January; 32(1 Pt 2):159-165; Fisher G J, Datta S C, Talwar H S, Wang Z Q, Varani J, Kang S, et al., Molecular basis of sun - induced premature skin ageing and retinoid antagonism . Nature 1996; 379: 335-339).
  • MMP-1, MMP-13 and membrane-type MMP-14 display collagenolytic activity
  • MMP-2 and MMP-9 were reported to be true elastases.
  • MMP-mediated collagen and elastin destruction accounts for a large part of the connective tissue damage that occurs in photodamaged skin (Tsukahara K, Nakagawa H, Moriwaki S, Takema Y, Fujimura T, Imokawa G., Inhibition of ultraviolet - B - induced wrinkle formation by an elastase - inhibiting herbal extract: implication for the mechanism underlying elastase - associated wrinkles , Int J Dermatol 2006; 45: 460-468).
  • SHED exerts effects on HDFs by causing an increase in collagen synthesis and by activating the proliferation and migration activity of HDFs, suggesting that SHED or SH-CM can be used for the treatment of photoaging and wound healing.
  • SHED is more suitable for dermal wound healing compared with MSCs, in terms of properties thereof.
  • SHED contributes to enhance wound healing potential of HDFs.
  • Immortalized human mesenchymal stem cells (MSCs: Ronza Co., Ltd, USA) were used to prepare a growth factor (GF) admixture.
  • the cells were cultured for 2 to 8 passages using 10% FSC-containing DMEM.
  • the supernatant (culture medium: CM) was sampled.
  • the CM was concentrated by spinning (4° C., 1500 rpm, 15 min). The remaining CM was washed with 90% ethanol at ⁇ 20° C., and then spun again.
  • the concentrated CM was freeze-dried, thereby obtaining a growth factor powder.
  • Each growth factor in the powder was analyzed by the Western-blotting method.
  • the detected growth factors are as follows: PDGF, VEGF, IGF, KGF, HGF and TGF.
  • a titanium implant (3.75 mm in diameter) was inserted into the center of each defect.
  • the spaces around the implant were filled with graft materials, such as (1) PRP, (2) 100% GF, (3) MSCs (1 ⁇ 1,000,000) and (4) empty defect (control) ( FIG. 10 ).
  • graft materials such as (1) PRP, (2) 100% GF, (3) MSCs (1 ⁇ 1,000,000) and (4) empty defect (control) ( FIG. 10 ).
  • growth factors derived from a mesenchymal stem cell have similar abilities to a living stem cell with respect to bone regeneration.
  • the sinus lift procedure with installation of two implants was performed at a post molar region in maxilla.
  • a 100% cell-based GF with b-TCP ( ⁇ -tricalcium phosphate) granules was grafted into the sinus cavity. Eight weeks later, the grafted portion was successfully filled with new bone, and osteointegration between the implants and the bone was confirmed by X-ray observation ( FIGS. 14 and 15 ).
  • a two-wall type periodontal defect was made in the distal portion of molar teeth in the dog mandible ( FIG. 16 ).
  • the defect in each dog was treated by the following method or procedure ( FIGS. 17 and 18 ).
  • the dog mandible with molar teeth and gingiva was dissected, and a histological specimen thereof was made.
  • the depth of the pocket N 1 -JE; length of the epithelium down growth
  • the length of new cementum N 2 -NC
  • FIG. 19 These parameters were useful for the evaluation of the amelioration of periodontal disease.
  • the length of new cementum was much longer in GF and MSC than in GTR and the control. Further, the GF and MSC groups showed remarkable improvement in the depth of pocket, compared with the GTR and control groups ( FIGS. 20 to 22 ).
  • the MSC-derived growth factor has similar capacity to MSC themselves in terms of periodontal tissue regeneration.
  • the medial portion of the lower right canine had a deep periodontal defect having a depth of 7 mm.
  • the aterocollagen sponge with 100% GF was filled into the defect ( FIGS. 23 and 24 ).
  • the defect seemed to be clinically repaired with a newly formed periodontal tissue ( FIG. 25 ).
  • cytokine therapies have an advantage over stem cell therapies in terms of safety, stability, easy manipulation, easy preservation, easy transportation and low cost.
  • a 4-0 monofilament nylon suture (Shirakawa, Tokyo, Japan) with the tip rounded by flame heating and silicone (KE-200, Shin-Etsu Chemical, Tokyo, Japan) was advanced from the external carotid artery into the internal carotid artery until it blocked the origin of the MCA.
  • the regional cerebral blood flow of the MCA territory was measured using a laser-Doppler flowmeter (Omega FLO-N1: Omega Wave Inc, Tokyo, Japan) after occlusion. The response was considered positive and included only if the reduction in regional cerebral blood flow was greater than 70%.
  • SH-CM which had been prepared in the same manner as in Example 1, was used in this experiment.
  • the rats were laid on their backs, their neck were elevated by rolled-up 4 cm ⁇ 4 cm gauze, and a total of 100 ⁇ l per rat was administered in the olfactory pathway using a Hamilton microsyringe, 10 ⁇ l at a time, alternating the nostrils, with an interval of 2 min between each administration. During these procedures, the mouth and the opposite nostril were shut. Intranasal administration was performed everyday during a period from day 3 to day 15.
  • a blind test on the rats was carried out on days 1, 3, 6, 9, 12 and 15 using a standardized motor disability scale with slight modifications.
  • the rat was given 1 point for each of the following parameters: flexion of the forelimb contralateral to the stroke when instantly hung by the tail; extension of the contralateral hindlimb when pulled from the table; and rotation to the paretic side against resistance.
  • 1 point was given for circling motion to the paretic side when trying to walk, 1 point was given for failure to walk out of a circle of 50 cm in diameter within 10 seconds, 2 points were given for failure to leave the circle, within 20 seconds, and 3 points were scored for inability to exit the circle within 60 seconds.
  • 1 point each was given for inability of the rat to extend the paretic forepaw when pushed against the table from above, laterally, or sideways.
  • the evaluation according to the motor disability scale was performed 3 times per animal time-point.
  • cryosections obtained from samples on day 16 were stained with Hematoxylin and Eosin.
  • Image J National Institutes of Health, ML was used to determine each infarct area in 20 coronal sections (20 mm-thick) at 1.00-mm intervals. The entire infarct area was covered by these 12 coronal sections.
  • Regional infarct volumes were calculated by summing the infarct areas and multiplying these areas by the distance between sections (1.00 mm), followed by remediation for brain edema.
  • cytokine therapy has an excellent restorative effect toward cerebral infarct areas, and is useful for treatment of cerebral infarction. Similar results were also confirmed in other rats.
  • Conditioned media from dental pulp stem cells were prepared according to the following procedure (see, FIG. 29 ), and were used in an experiment for verifying the nerve regeneration effect.
  • PC12 cells a cell line derived from an immortalized rat adrenal pheochromocytoma, were used as neural cells. It is known that the addition of nerve growth factor (NGF), one of neurotrophic factors, to PC 12 cells induces the outgrowth of axon-like processes and differentiation into neuron-like cells. Thus, PC12 cells are used as model cells for various in vitro experiments on nervous system.
  • NGF nerve growth factor
  • the neurite outgrowth effect and apoptosis inhibitory effect of the dental pulp stem cell-conditioned medium were examined in the presence or absence of a nerve regeneration inhibitory substance (neurite outgrowth inhibitory factor).
  • CSPG and MAG were used as nerve regeneration inhibitory substances.
  • the protocol of the experiment is as described below.
  • P12 cells are seeded in the plates coated with the nerve regeneration inhibitory substance, and are cultured with the dental pulp stem cell-conditioned medium for 24 hours.
  • the apoptosis of the cells is evaluated according to the TUNEL assay.
  • a serum-free medium, a fibroblast-conditioned medium and a bone marrow mesenchymal stem cell-conditioned medium are used as comparative groups.
  • the dental pulp stem cell-conditioned medium exhibited a stronger neurite outgrowth effect ( FIGS. 30 to 33 ) and apoptosis inhibitory effect ( FIGS. 34 and 35 ) as compared to other groups (comparative groups).
  • the dental pulp stem cell-conditioned medium exhibited a strong neurite outgrowth effect even without the addition of NGF, which is essential for PC12 cells to differentiate into neuron-like cells (i.e., the dental pulp stem cell-conditioned medium exhibited a strong neurite outgrowth effect by itself).
  • culturing PC12 neuron-like cells with the dental pulp stem cell-conditioned medium results in outgrowth of neurites (i.e., the dental pulp stem cell-conditioned medium exhibits neurite outgrowth activity), even in the dish coated with a nerve regeneration inhibitory substance CSPG (see FIG. 30 ).
  • Addition of the bone marrow mesenchymal stem cell-conditioned medium or the skin-derived fibroblast-conditioned medium alone, or addition of Y27632 alone, which inhibits ROCK activation, does not exhibit such outgrowth activity.
  • the dental pulp stem cell-conditioned medium increases the proportion of cells exhibiting neurite outgrowth, and promotes the formation of longer neurites, even under conditions in which a nerve regeneration inhibitory substance CSPG is present.
  • culturing PC12 neuron-like cells with the dental pulp stem cell-conditioned medium results in outgrowth of neurites (i.e., the dental pulp stem cell-conditioned medium exhibits neurite outgrowth activity), even in the dish coated with a nerve regeneration inhibitory substance MAG.
  • Addition of the bone marrow mesenchymal stem cell-conditioned medium or the dermal fibroblast-conditioned medium alone, or addition of Y27632 alone, which inhibits ROCK activation, does not exhibit such outgrowth activity.
  • the dental pulp stem cell-conditioned medium increases the proportion of cells exhibiting neurite outgrowth, and promotes the formation of longer neurites, even under conditions in which a nerve regeneration inhibitory substance MAG is present.
  • PLL poly-L-lysine coat
  • PLL+NGF poly-L-lysine coat and addition of nerve growth factor (NGF)
  • PLL/CSPG poly-L-lysine coat and CSPG coat
  • PLL/CSPG Y27632 poly-L-lysine coat
  • PLL/CSPG SHED-CM poly-L-lysine coat, CSPG coat and culturing with SHED-conditioned medium
  • PLL/CSPG DPSC-CM poly-L-lysine coat, CSPG coat and culturing with DPSC-conditioned medium
  • PLL/CSPG BMSC-CM poly-L-lysine coat
  • PLL/CSPG Fibro-CM poly-L-lysine coat
  • the symbols in FIG. 31 represent the following: PLL: poly-L-lysine coat, PLL+NGF: poly-L-lysine coat and addition of nerve growth factor (NGF), PLL/CSPG: poly-L-lysine coat and CSPG coat, PLL/CSPG+NGF: poly-L-lysine coat, CSPG coat and addition of NGF, PLL/CSPG+SHED-CM: poly-L-lysine coat, CSPG coat and culturing with SHED-conditioned medium, PLL/CSPG+NGF+SHED-CM: poly-L-lysine coat, CSPG coat, addition of NGF and culturing with SHED-conditioned medium, PLL/CSPG+DPSC-CM: poly-L-lysine coat, CSPG coat and culturing with DPSC-conditioned medium, PLL/CSPG+NGF+DPSC-CM: poly-L-lysine coat, CSPG coat, addition of NGF and culturing with DPSC-conditioned
  • PLL poly-L-lysine coat
  • PLL+NGF poly-L-lysine coat and addition of nerve growth factor (NGF)
  • PLL/MAG poly-L-lysine coat and MAG coat
  • PLL/MAG Y27632 poly-L-lysine coat, MAG coat and addition of Y27632
  • PLL/MAG SHED-CM poly-L-lysine coat, MAG coat and culturing with SHED-conditioned medium
  • PLL/MAG DPSC-CM poly-L-lysine coat, MAG coat and culturing with DPSC-conditioned medium
  • PLL/MAG BMSC-CM poly-L-lysine coat, MAG coat and culturing with bone marrow mesenchymal stem cell-conditioned medium
  • PLL/MAG Fibro-CM poly-L-lysine coat, MAG coat and culturing with fibroblast-conditioned medium.
  • PLL poly-L-lysine coat
  • PLL+NGF poly-L-lysine coat and addition of nerve growth factor (NGF)
  • PLL/MAG poly-L-lysine coat and MAG coat
  • PLL/MAG+NGF poly-L-lysine coat
  • PLL/MAG+SHED-CM poly-L-lysine coat
  • PLL/MAG+NGF+SHED-CM poly-L-lysine coat
  • MAG coat addition of NGF and culturing with SHED-conditioned medium
  • PLL/MAG+DPSC-CM poly-L-lysine coat, MAG coat and culturing with DPSC-conditioned medium
  • PLL/MAG+NGF+DPSC-CM poly-L-lysine coat, MAG coat, addition of NGF and culturing with DPSC-conditioned medium
  • PLL/MAG+BMSC-CM poly-L-lysine coat,
  • the symbols in FIG. 34 represent the following: PLL: poly-L-lysine coat, PLL/CSPG: poly-L-lysine coat and CSPG coat, PLL/CSPG SHED-CM: poly-L-lysine coat, CSPG coat and culturing with SHED-conditioned medium, PLL/CSPG DPSC-CM: poly-L-lysine coat, CSPG coat and culturing with DPSC-conditioned medium, PLL/CSPG BMSC-CM: poly-L-lysine coat, CSPG coat and culturing with bone marrow mesenchymal stem cell-conditioned medium, PLL/CSPG Fibro-CM: poly-L-lysine coat, CSPG coat and culturing with fibroblast-conditioned medium, PLL/CSPG+Y27632: poly-L-lysine coat, CSPG coat and addition of Y27632, PLL: poly-L-lysine coat, PLL/MAG: poly-L-lysine coat and
  • PLL poly-L-lysine coat
  • PLL/CSPG poly-L-lysine coat and CSPG coat
  • PLL/CSPG SHED-CM poly-L-lysine coat
  • PLL/CSPG DPSC-CM poly-L-lysine coat
  • PLL/CSPG BMSC-CM poly-L-lysine coat
  • PLL/CSPG Fibro-CM poly-L-lysine coat
  • PLL/CSPG+Y27632 poly-L-lysine coat
  • PLL poly-L-lysine coat
  • PLL/MAG poly-L-lysine coat and MAG
  • the dental pulp stem cell-conditioned medium suppresses the action of nerve regeneration inhibitory substances, promotes neurite outgrowth, and suppresses apoptosis.
  • the dental pulp stem cell-conditioned medium is quite effective for CNS regeneration and the treatment of CNS diseases.
  • 10th thoracic vertebrae were removed from 8-week-old female SD rats under general anesthesia with pentobarbital sodium, and crush injury damage was induced by applying a 200 kilodyn force from outside the dura mater using an IH impactor, to obtain a model of spinal cord crush injury.
  • SHED-CM dental pulp stem cell-conditioned medium
  • BMSC-CM bone marrow mesenchymal stem cell-conditioned medium
  • PBS control
  • BBB Basso-Beattie-Bresnahan
  • the ameliorating effects of the hindlimb motor function was compared based on the BBB scores.
  • the evaluation results are shown in FIG. 36 .
  • the group administred with the dental pulp stem cell-conditioned medium (SHED-CM) exhibited surprising improvement and recovery of the hindlimb motor function as represented by score 15 (forelimb-hindlimb coordination, occasional (5% to 50%) steps with heel lifting, and external rotation of paw position).
  • the group administered with the bone marrow mesenchymal stem cell-conditioned medium (BMSC-CM) also exhibited a certain degree of improvement, the improvement effect thereof is far lower than that of the SHED-CM group.
  • the rats were perfusion fixed with paraformaldehyde. Subsequently, a length of the spinal cord including the injury site and extending 5 mm rostrally and 5 mm caudally from the injury site was dissected, and taken out. The weights of the spinal cords were compared between the Sham group, the control group, the BMSC-CM group and the SHED-CM group.
  • the morphological alteration of the spinal cord 8 weeks after the initiation of the administration was assessed.
  • the states of the spinal cords taken out are shown in the upper panel of FIG. 37 .
  • the comparison of the weights (masses) of the spinal cords is shown in the lower panel of FIG. 37 .
  • the SHED-CM treated group the atrophy of the spinal cord caudal to the injury site (injury epicenter) was suppressed (the upper panel of FIG. 37 ).
  • the morphological alteration of the injured spinal cord was suppressed by the administration of SHED-CM.
  • the SHEM-CM group also exhibited an increase in the weight of the spinal cord (the lower panel of FIG. 37 ).
  • the rats were perfusion fixed with paraformaldehyde.
  • a length of the spinal cord including the injury site and extending 5 mm rostrally and 5 mm caudally from the injury site was dissected, and taken out.
  • the spinal cord was embedded and frozen in O.C.T compound, and frozen section slides of the spinal cord were prepared.
  • the frozen section slides of the spinal cord were immunostained with an anti-serotonin (5-HT) antibody and with an antibody against neuroaxis (anti-Neurofilament-M (NF-M) antibody).
  • 5-HT anti-serotonin
  • NF-M anti-Neurofilament-M
  • the damaged portion and a neighbourhood thereof were histologically examined 8 weeks after the initiation of the administration.
  • the results of the immunostaining are shown in FIG. 38 .
  • Continuous administration of a small amount of SHED-CM maintained the number of the total neurofilaments (NF-M) in the region caudal to the injury site.
  • the number of serotonin fibers projecting from the raphe nuclei of the brain stem to the spinal cord was also maintained. It was revealed that the loss of neurofilaments is suppressed by the administration of SHED-CM, and that a neurotransmitter serotonin produced in the upper areas and the brain stem was transported to the areas lower than the injury site.
  • control group and the SHED-CM group were perfusion fixed with paraformaldehyde 24 hours after the spinal cord crush injury and one week after the spinal cord crush injury.
  • a length of the spinal cord including the injury site and extending 5 mm rostrally and 5 mm caudally from the injury site was dissected, and taken out. Then, the spinal cord taken out was embedded and frozen in O.C.T compound, and frozen section slides of the spical cord were prepared.
  • the frozen section slides of the spical cord were double-immunostained with TUNEL, which specifically reacts with fragmentalized DNAs, and an anti-GFAP antibody specific for astrocytes, an anti-NeuN antibody specific for neuronal cells or an anti-CNPase antibody specific for oligodendrocytes, to compare the cell death of neuronal cells and glia cells.
  • the cell death of the neuronal cells in the damaged portion and in the neighbourhood thereof was evaluated 24 hours after the spinal cord crush injury and one week after the spinal cord crush injury. The results are shown in FIG. 39 .
  • Continuous administration of a small amount of SHED-CM suppressed apoptotic cell death of astrocytes, neurons and oligodendrocytes that occurred immediately after the nerve injury.
  • apoptotic cell death of oligodendrocytes are observed in a larger area one week after the injury (enlargement of secondary damage). It was revealed that SHED-CM also suppresses this apoptotic cell death, thereby suppressing the enlargement of neural damage.
  • neurodegenerative diseases such as amyotrophic lateral sclerosis, Alzheimer's disease, Parkinson's disease, progressive supranuclear palsy, Huntington's disease, multiple system atrophy and spinocerebellar ataxia, degeneration or loss of neuronal cells caused by cerebral ischemia, intracerebral hemorrhage or cerebral infarction and a retinal disease involving a neuronal cell disorder are contemplated as diseases to which the CNS disease treatment composition according to the invention can be applied.
  • neurodegenerative diseases such as amyotrophic lateral sclerosis, Alzheimer's disease, Parkinson's disease, progressive supranuclear palsy, Huntington's disease, multiple system atrophy and spinocerebellar ataxia
  • degeneration or loss of neuronal cells caused by cerebral ischemia intracerebral hemorrhage or cerebral infarction
  • a retinal disease involving a neuronal cell disorder are contemplated as diseases to which the CNS disease treatment composition according to the invention can be applied
  • a stem cell-conditioned medium that is obtained by culturing stem cells and that contains a mixture of cytokines is used, endogenous stem cells in the target tissue is allowed to differentiate and proliferate. As a result, the target tissue is repaired and regenerated through the proliferation of cells in the damaged part, the generation of extracellular matrix, etc.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Chemical & Material Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Medicinal Chemistry (AREA)
  • Zoology (AREA)
  • Immunology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Epidemiology (AREA)
  • Developmental Biology & Embryology (AREA)
  • Biomedical Technology (AREA)
  • Cell Biology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Biotechnology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Virology (AREA)
  • Organic Chemistry (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Neurology (AREA)
  • Neurosurgery (AREA)
  • Reproductive Health (AREA)
  • Rheumatology (AREA)
  • Genetics & Genomics (AREA)
  • Wood Science & Technology (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Ophthalmology & Optometry (AREA)
  • Vascular Medicine (AREA)
  • Molecular Biology (AREA)
  • Endocrinology (AREA)
  • Diabetes (AREA)
  • Hematology (AREA)
  • Gynecology & Obstetrics (AREA)
  • General Engineering & Computer Science (AREA)
US13/637,107 2010-03-26 2011-03-25 Composition for Treatment of Damaged Part Abandoned US20130195991A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US13/637,107 US20130195991A1 (en) 2010-03-26 2011-03-25 Composition for Treatment of Damaged Part

Applications Claiming Priority (9)

Application Number Priority Date Filing Date Title
US31771310P 2010-03-26 2010-03-26
US41037010P 2010-11-05 2010-11-05
JP2010267962 2010-12-01
JP2010-267962 2010-12-01
US201161437697P 2011-01-31 2011-01-31
JP2011-037028 2011-02-23
JP2011037028 2011-02-23
US13/637,107 US20130195991A1 (en) 2010-03-26 2011-03-25 Composition for Treatment of Damaged Part
PCT/JP2011/057412 WO2011118795A1 (ja) 2010-03-26 2011-03-25 損傷部治療用組成物

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP2011/057412 A-371-Of-International WO2011118795A1 (ja) 2010-03-26 2011-03-25 損傷部治療用組成物

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US16/043,395 Division US11135243B2 (en) 2010-03-26 2018-07-24 Composition for treatment of damaged part

Publications (1)

Publication Number Publication Date
US20130195991A1 true US20130195991A1 (en) 2013-08-01

Family

ID=44673335

Family Applications (2)

Application Number Title Priority Date Filing Date
US13/637,107 Abandoned US20130195991A1 (en) 2010-03-26 2011-03-25 Composition for Treatment of Damaged Part
US16/043,395 Active US11135243B2 (en) 2010-03-26 2018-07-24 Composition for treatment of damaged part

Family Applications After (1)

Application Number Title Priority Date Filing Date
US16/043,395 Active US11135243B2 (en) 2010-03-26 2018-07-24 Composition for treatment of damaged part

Country Status (7)

Country Link
US (2) US20130195991A1 (ko)
EP (1) EP2554175B1 (ko)
JP (2) JPWO2011118795A1 (ko)
KR (1) KR20130008594A (ko)
CN (1) CN103037872B (ko)
ES (1) ES2663330T3 (ko)
WO (1) WO2011118795A1 (ko)

Cited By (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9856455B2 (en) 2006-08-24 2018-01-02 Neurofx, Inc. Methods of producing stem cell conditioned media
US9962428B2 (en) 2012-12-21 2018-05-08 National University Corporation Nagoya University Composition having tissue-repairing activity, and use therefor
US10000738B2 (en) 2014-01-27 2018-06-19 Beijing Stomatology Hospital, Capital Medical University Usage of odontogenic stem cells and genetically modified odontogenic stem cells
US20190167724A1 (en) * 2017-12-04 2019-06-06 Caire Medical-Biotechnology International Co. Neuroprotective composition, preparation process thereof and medical uses thereof
US10494606B2 (en) * 2012-03-28 2019-12-03 Quarrymen&Co. Inc. Immortalized stem cell, compositions, preparations and uses thereof
US10709741B2 (en) * 2013-02-13 2020-07-14 National University Corporation Nagoya University Composition for preventing or treating inflammatory disease
US11015171B2 (en) 2015-11-05 2021-05-25 Quarrymen & Co. Inc. Immortalized stem cells and method for producing same
US20210260135A1 (en) * 2018-08-03 2021-08-26 Laboratory Of Cell Applied Technologies, Co Production Method for Culture Supernatant Preparation
US11179420B2 (en) 2016-04-27 2021-11-23 Rohto Pharmaceutical Co., Ltd. Method for treating a disease, comprising administering mesenchymal stem cells or culture supernatant thereof to a subject
US11679122B2 (en) 2017-12-21 2023-06-20 Osaka University Therapeutic agent for nervous system disease
CN117545495A (zh) * 2021-06-21 2024-02-09 帕纳吉株式会社 眼部症状改善剂和眼部症状的改善方法

Families Citing this family (45)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20130195991A1 (en) * 2010-03-26 2013-08-01 National University Corporation Nagoya University Composition for Treatment of Damaged Part
JP5608927B2 (ja) * 2010-04-13 2014-10-22 国立大学法人名古屋大学 歯髄幹細胞を用いた神経疾患治療用組成物
SG195128A1 (en) * 2011-06-03 2013-12-30 Mesoblast Inc Method of treating the effects of stroke
KR20180128097A (ko) * 2012-02-10 2018-11-30 재패닉 코포레이션 비 인간 줄기세포의 배양 상등액을 원료로 하는 화장품 또는 피부 재생 촉진제, 및 단백질의 이온 도입 방법
KR102126331B1 (ko) * 2012-02-23 2020-06-25 주식회사 탑셀바이오 컨디션드 배지 분말의 제조방법
KR101914580B1 (ko) * 2012-03-07 2018-11-06 주식회사 탑셀바이오 줄기세포 컨디션드 배지 분말을 포함하는 뇌신경 또는 척수신경 관련 질환 예방, 개선 또는 치료용 의약 조성물
US11744856B1 (en) 2012-04-11 2023-09-05 Medicell Technologies, Llc Compositions and methods to improve skin quality and appearance, cure skin and tissue damage, and use in therapy
KR20150108843A (ko) * 2012-12-26 2015-09-30 가부시키가이샤 쿠오리맨 고기능 임플란트 재료
JPWO2014126175A1 (ja) * 2013-02-13 2017-02-02 国立大学法人名古屋大学 糖尿病の予防又は治療用組成物
JP2014156410A (ja) * 2013-02-14 2014-08-28 Nagoya Univ 組織形成用組成物及びその利用
WO2014185470A1 (ja) * 2013-05-14 2014-11-20 株式会社再生医療推進機構 神経損傷の治療用移植材の製造方法
JP2014230685A (ja) * 2013-05-30 2014-12-11 国立大学法人 東京医科歯科大学 歯周組織形成用材料
EP3013944B1 (en) * 2013-06-24 2020-03-18 University of Southern California Composition of mesenchymal stem cells
CN106102754B (zh) 2014-01-24 2020-04-17 石匠株式会社 癌治疗用医药组合物以及将该组合物作为有效成分的癌治疗用医药制剂
JP6474549B2 (ja) * 2014-03-03 2019-02-27 国立大学法人徳島大学 幹細胞の培養産物の評価指標及びその利用
JP2016128396A (ja) * 2015-01-09 2016-07-14 上田 実 iPS細胞培養上清を含む医薬組成物およびその製造方法、化粧品およびその製造方法、抗加齢組成物、疾患発症抑制方法、疾患治療方法、組織異常治療方法ならびに美容方法
JP2016128397A (ja) * 2015-01-09 2016-07-14 上田 実 iPS細胞培養上清を含む神経変性疾患治療用組成物およびその製造方法、神経変性疾患発症抑制方法ならびに神経変性疾患治療方法
CN104726406B (zh) * 2015-02-13 2018-05-01 中国医科大学 一种诱导牙髓间充质干细胞分化为神经细胞的方法
CN104711219A (zh) * 2015-03-01 2015-06-17 安徽新生命干细胞科技有限公司 一种牙髓干细胞培养基
JP2016210727A (ja) * 2015-05-08 2016-12-15 株式会社 バイオミメティクスシンパシーズ 培養上清を含有するマイクロニードル剤
CN105749254A (zh) * 2016-02-28 2016-07-13 深圳爱生再生医学科技有限公司 用于治疗血管性痴呆的干细胞制剂及其制备方法和应用
JP6401818B2 (ja) * 2016-04-27 2018-10-10 株式会社金太郎CellsPower 活性化幹細胞製造方法
MY186453A (en) * 2016-04-27 2021-07-22 Kintarocellspower Co Ltd Method for producing activated stem cells
JP7083141B2 (ja) * 2016-05-31 2022-06-10 学校法人愛知学院 筋肉量減少又は筋力低下を改善するための組成物及びその利用
CN109843305B (zh) * 2016-08-14 2023-03-10 雷蒙特亚特特拉维夫大学有限公司 治疗神经障碍的间充质细胞来源的外泌体
CN106511384A (zh) * 2016-11-08 2017-03-22 广州医科大学附属第三医院 一种用于促进糖尿病创面愈合的冻干粉及其制备方法
CN110831694A (zh) * 2017-03-31 2020-02-21 细尔琳生物医学公司 生物相容性调整细胞培养基组合物及其用途
EP3607957A4 (en) * 2017-05-02 2020-12-16 Koji Tanabe PHARMACEUTICAL COMPOSITION AND COSMETIC COMPOSITION
JP6860915B2 (ja) * 2017-10-31 2021-04-21 株式会社日本バイオセラピー研究所 培養上清を生産する方法
GB2568928B (en) * 2017-12-01 2023-03-08 Young Cell Biomedical Tech Inc Neuroprotective composition, preparation process thereof and medical uses thereof
WO2019139137A1 (ja) 2018-01-12 2019-07-18 国立大学法人大阪大学 重層扁平上皮細胞の正常分化・成熟促進剤、上皮疾患治療剤及び重層扁平上皮細胞の正常分化・成熟促進方法
JPWO2019216438A1 (ja) * 2018-05-11 2021-05-20 株式会社ジーンテクノサイエンス 間葉系細胞の培養上清を含む歯周病治療剤
JP6884935B2 (ja) * 2018-12-20 2021-06-09 株式会社システムエッヂ 再生治療用組成物の製造方法
JP6684956B1 (ja) * 2019-05-29 2020-04-22 パナジー株式会社 細胞増殖方法、細胞増殖剤および細胞増殖用培地
KR20220066959A (ko) * 2019-09-26 2022-05-24 가부시키가이샤 투셀 생체 조직 손상 수복제의 제조 방법 및 생체 조직 손상 수복제
JP2021116303A (ja) * 2020-01-23 2021-08-10 医療法人社団サカイクリニック62 歯周組織用製剤
JP6830286B1 (ja) 2020-06-26 2021-02-17 デクソンファーマシューティカルズ株式会社 精巣機能改善剤および精巣機能改善方法
CN115996708A (zh) * 2020-06-30 2023-04-21 国立大学法人高知大学 含有脐带血单个核细胞培养上清的脑神经损伤治疗剂
WO2022014693A1 (ja) 2020-07-17 2022-01-20 デクソンファーマシューティカルズ株式会社 組成物、結合阻害剤、医療機器およびcovid-19の予防方法
WO2022054565A1 (ja) 2020-09-08 2022-03-17 デクソンファーマシューティカルズ株式会社 サイトカインストーム抑制剤、サイトカインストーム抑制剤の使用方法およびスクリーニング方法
JP2022086174A (ja) * 2020-11-30 2022-06-09 ニューロテックメディカル株式会社 神経障害の治療剤
JP7057006B1 (ja) 2021-04-12 2022-04-19 医療法人社団サカイクリニック62 歯周組織用製剤及びそれを含むキット
JP6974892B1 (ja) * 2021-05-27 2021-12-01 デクソンファーマシューティカルズ株式会社 癌悪液質の改善剤および癌悪液質の改善方法
JP7125818B1 (ja) * 2022-04-07 2022-08-25 株式会社再生医学研究所 難治性神経疾患治療用組成物およびその製造方法
JP7468955B1 (ja) 2023-10-17 2024-04-16 セルプロジャパン株式会社 細胞の培養上清を生産する方法

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008020815A1 (en) * 2006-08-15 2008-02-21 Agency For Science, Technology And Research Mesenchymal stem cell conditioned medium
WO2008024996A2 (en) * 2006-08-24 2008-02-28 Johnstone Brian H Production of neural protective and regenerative factors from stem cells and treatment of nervous system conditions therewith
WO2008060374A2 (en) * 2006-10-06 2008-05-22 University Of Virginia Patent Foundation Methods and compositions useful for diabetic wound healing
US20090053182A1 (en) * 2007-05-25 2009-02-26 Medistem Laboratories, Inc. Endometrial stem cells and methods of making and using same
US20110177041A1 (en) * 2008-08-22 2011-07-21 Misako Nakashima Material for treatment of cerebral infarction and brain tissue regeneration method

Family Cites Families (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP3763749B2 (ja) 2001-03-28 2006-04-05 独立行政法人科学技術振興機構 脊髄におけるシナプス形成ニューロンを誘導する中枢神経系前駆細胞
KR100449141B1 (ko) 2001-04-19 2004-09-21 (주)라이프코드 간엽 간세포를 신경세포로 분화시키는 방법
CA2466478A1 (en) * 2001-11-28 2003-06-05 Anges Mg, Inc. Gene therapy agents for neurodegenerative diseases
US9175264B2 (en) * 2003-04-19 2015-11-03 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Postnatal stem cells and uses thereof
CN1882341A (zh) * 2003-11-19 2006-12-20 艾克若克斯Dds有限公司 用于治疗皮肤损伤的方法和组合物
ITNA20040043A1 (it) * 2004-07-28 2004-10-28 Francesco Carinci Tecnica di ingegneria tissutale ottenibile mediante isolamento di una nuova sottopopolazione di cellule staminali mbp-shed ed mbp-dpsc, isolate da polpa di denti decidui e permanenti in grado di produrre in vitro tessuto osseo umano.
US20110002895A1 (en) * 2007-12-05 2011-01-06 Minoru Ueda Composition for autotransplantation or allotransplantation using dental pulp stem cell, and use of the composition
JP4629791B2 (ja) 2009-12-21 2011-02-09 日立コンシューマエレクトロニクス株式会社 デジタルコンテンツ受信装置、デジタルコンテンツ受信方法、およびデジタルコンテンツ送受信方法
US20130195991A1 (en) * 2010-03-26 2013-08-01 National University Corporation Nagoya University Composition for Treatment of Damaged Part

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008020815A1 (en) * 2006-08-15 2008-02-21 Agency For Science, Technology And Research Mesenchymal stem cell conditioned medium
WO2008024996A2 (en) * 2006-08-24 2008-02-28 Johnstone Brian H Production of neural protective and regenerative factors from stem cells and treatment of nervous system conditions therewith
WO2008060374A2 (en) * 2006-10-06 2008-05-22 University Of Virginia Patent Foundation Methods and compositions useful for diabetic wound healing
US20090053182A1 (en) * 2007-05-25 2009-02-26 Medistem Laboratories, Inc. Endometrial stem cells and methods of making and using same
US20110177041A1 (en) * 2008-08-22 2011-07-21 Misako Nakashima Material for treatment of cerebral infarction and brain tissue regeneration method

Non-Patent Citations (7)

* Cited by examiner, † Cited by third party
Title
Gandia et al. "Human dental pulp stem cells improve left ventricular function, induce angiogenesis, and reduce infarct size in rats with acute myocardial infarction." Stem cells 26 (3): 638-645, 2008 *
Iohora et al. "A novel stem cell source for vasculogenesis in ischemia: subfraction of side population cells from dental pulp", Stem Cells 26: 2408-2418, 2008 *
Kim et al. "Antiwrinkle effect of adipose-derived stem cell: activation of dermal fibroblast by secretory factors." Journal of Dermatological Science 53(2): 96-102, 2009 *
Pierdomenico et al. "Multipotent mesenchymal stem cells with immunosuppressive activity can be easily isolated from dental pulp", Transplantation 80: 836-42, 2005 *
Sakai et al. "Human dental pulp-derived stem cells promote locomotor recovery after complete transection of the rat spinal cord by multiple neuro-regenerative mechanisms" The Journal of clinical investigation 122(1): 80-90, 2012 *
Tran-Hung et al. "Quantification of angiogenic growth factors released by human dental cells after injury." Archives of Oral Biology 53 (1): 9-13, 2008 *
What are adult stem cells?. In Stem Cell Information [World Wide Web site]. Bethesda, MD: National Institutes of Health, U.S. Department of Health and Human Services, 2012 [cited Friday, September 19, 2014] Available at *

Cited By (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9856455B2 (en) 2006-08-24 2018-01-02 Neurofx, Inc. Methods of producing stem cell conditioned media
US10683482B2 (en) 2006-08-24 2020-06-16 Neurofx, Inc. Stem cell conditioned media and methods of using the same
US10494606B2 (en) * 2012-03-28 2019-12-03 Quarrymen&Co. Inc. Immortalized stem cell, compositions, preparations and uses thereof
US11000572B2 (en) 2012-12-21 2021-05-11 Tokushima University Composition having tissue-repairing activity, and use therefor
US10507230B2 (en) 2012-12-21 2019-12-17 Tokushima University Composition having tissue-repairing activity, and use therefor
US9962428B2 (en) 2012-12-21 2018-05-08 National University Corporation Nagoya University Composition having tissue-repairing activity, and use therefor
US10709741B2 (en) * 2013-02-13 2020-07-14 National University Corporation Nagoya University Composition for preventing or treating inflammatory disease
US10000738B2 (en) 2014-01-27 2018-06-19 Beijing Stomatology Hospital, Capital Medical University Usage of odontogenic stem cells and genetically modified odontogenic stem cells
US11015171B2 (en) 2015-11-05 2021-05-25 Quarrymen & Co. Inc. Immortalized stem cells and method for producing same
US11179420B2 (en) 2016-04-27 2021-11-23 Rohto Pharmaceutical Co., Ltd. Method for treating a disease, comprising administering mesenchymal stem cells or culture supernatant thereof to a subject
US20190167727A1 (en) * 2017-12-04 2019-06-06 Caire Medical-Biotechnology International Co. Neuroprotective composition, preparation process thereof and medical uses thereof
US20190167724A1 (en) * 2017-12-04 2019-06-06 Caire Medical-Biotechnology International Co. Neuroprotective composition, preparation process thereof and medical uses thereof
US11679122B2 (en) 2017-12-21 2023-06-20 Osaka University Therapeutic agent for nervous system disease
US20210260135A1 (en) * 2018-08-03 2021-08-26 Laboratory Of Cell Applied Technologies, Co Production Method for Culture Supernatant Preparation
CN117545495A (zh) * 2021-06-21 2024-02-09 帕纳吉株式会社 眼部症状改善剂和眼部症状的改善方法

Also Published As

Publication number Publication date
ES2663330T3 (es) 2018-04-12
EP2554175A4 (en) 2013-08-21
JP2016065106A (ja) 2016-04-28
CN103037872A (zh) 2013-04-10
US11135243B2 (en) 2021-10-05
EP2554175B1 (en) 2018-02-28
JPWO2011118795A1 (ja) 2013-07-04
EP2554175A1 (en) 2013-02-06
CN103037872B (zh) 2015-12-02
US20180325946A1 (en) 2018-11-15
WO2011118795A1 (ja) 2011-09-29
KR20130008594A (ko) 2013-01-22
JP6296622B2 (ja) 2018-03-20

Similar Documents

Publication Publication Date Title
US11135243B2 (en) Composition for treatment of damaged part
JP6286689B2 (ja) 非ヒト幹細胞の培養上清を原材料とする化粧品又は皮膚再生促進剤、及びタンパク質のイオン導入方法
Rooney et al. Gene-modified mesenchymal stem cells express functionally active nerve growth factor on an engineered poly lactic glycolic acid (PLGA) substrate
MX2010009540A (es) Composiciones y metodos para usar celulas estromaticas para mejorar el tratamiento de lesiones del sistema nervioso central.
Fuloria et al. Regenerative potential of stem cells derived from human exfoliated deciduous (SHED) teeth during engineering of human body tissues
WO2012156968A2 (en) Use of mesenchymal stem cells for the improvement of affective and cognitive function
Kircelli et al. Neuronal regeneration in injured rat spinal cord after human dental pulp derived neural crest stem cell transplantation
US20200206272A1 (en) Treatment agent for epidermolysis bullosa
JP2016210730A (ja) 医薬組成物及びその製造方法並びに医薬品
WO2017064672A1 (en) A method and composition to differentiate mesenchymal stem cells into melanocytes
Naito et al. Characterization of canine dental pulp cells and their neuroregenerative potential
US11466254B2 (en) Composition containing, as active ingredient, culture of chicken bone marrow-derived osteochondral progenitor cells for promoting osteogenesis or inducing chondrogenic differentiation
CN111821318A (zh) 用于治疗或预防白发的外泌体及其用途和使用方法
US10214725B2 (en) Methods and compositions for somatic cell proliferation and viability
KR20160052377A (ko) C3 또는 C1r 보체를 분비하는 태반 유래 세포 및 이를 포함하는 조성물
US11339196B2 (en) Method using expression of LIN28 for preparing stem cells having excellent renewal ability and therapeutic capacity
Yang et al. Application of human umbilical cord mesenchymal stem cells in spinal cord injury
US20230242873A1 (en) Fibroblast based therapy for treatment of parkinson's disease
KR101224504B1 (ko) 크립토탄신온의 신규 용도
Zadeh et al. Mining Human Clinical Waste as a Rich Source of Stem Cells for Neural Regeneration
Liu et al. Stem Cells from Human Dental Tissue for Regenerative Medicine
Pajer et al. Therapeutic Cells and Stem Cells for Nerve Regeneration
Ahmed et al. Progression of Treating Alzheimer's Disease with Stem Cell-based Therapies
KR20230140802A (ko) 줄기세포의 기능 강화용 조성물 및 이의 용도
WO2016111378A1 (ja) iPS細胞培養上清を含む神経変性疾患治療用組成物およびその製造方法、神経変性疾患発症抑制方法ならびに神経変性疾患治療方法

Legal Events

Date Code Title Description
AS Assignment

Owner name: NATIONAL UNIVERSITY CORPORATION NAGOYA UNIVERSITY,

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:UEDA, MINORU;YAMADA, YOICHI;EBISAWA, KATSUMI;AND OTHERS;REEL/FRAME:029459/0620

Effective date: 20121108

AS Assignment

Owner name: TOKUSHIMA UNIVERSITY, JAPAN

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:NATIONAL UNIVERSITY CORPORATION NAGOYA UNIVERSITY;REEL/FRAME:043704/0307

Effective date: 20170501

STCB Information on status: application discontinuation

Free format text: ABANDONED -- AFTER EXAMINER'S ANSWER OR BOARD OF APPEALS DECISION