WO2008056963A1 - Procédé permettant la prolifération de cellules souches au moyen de leptine - Google Patents

Procédé permettant la prolifération de cellules souches au moyen de leptine Download PDF

Info

Publication number
WO2008056963A1
WO2008056963A1 PCT/KR2007/005670 KR2007005670W WO2008056963A1 WO 2008056963 A1 WO2008056963 A1 WO 2008056963A1 KR 2007005670 W KR2007005670 W KR 2007005670W WO 2008056963 A1 WO2008056963 A1 WO 2008056963A1
Authority
WO
WIPO (PCT)
Prior art keywords
stem cells
leptin
cells
cell
culture medium
Prior art date
Application number
PCT/KR2007/005670
Other languages
English (en)
Inventor
Chanil Moon
Hyunwon Yang
Original Assignee
Chanil Moon
Hyunwon Yang
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 Chanil Moon, Hyunwon Yang filed Critical Chanil Moon
Publication of WO2008056963A1 publication Critical patent/WO2008056963A1/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0607Non-embryonic pluripotent stem cells, e.g. MASC
    • 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
    • C12N5/0634Cells from the blood or the immune system
    • C12N5/0647Haematopoietic stem cells; Uncommitted or multipotent progenitors
    • 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
    • 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
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/30Hormones
    • C12N2501/355Leptin

Definitions

  • the present invention relates, in general, to a method for proliferating stem cells with leptin and, more specifically, to a method for proliferating various bone marrow- or blood-derived stem cells to a great extent in a culture medium supplemented with leptin without an increase in the number of subcultures
  • a stem cell is an undifferentiated somatic cell that is capable either of division to give rise to daughter stem cells, or of differentiating into any specialized cell type given the appropriate signals.
  • In vitro culturing and differentiation of stem cells affords specific cells useful in the treatment of various diseases (Petit-Zeman S., Regenerative medicine. Nat. Biotechnol. 19: 201-206, 2001) .
  • organ transplantation was developed as a predominant promising therapy for replacing recipients' damaged or failing organs with working ones from donors .
  • organ transplantation is problematic in that organs are in very short supply and donors as well as recipients must undergo surgery.
  • stem cell therapy does not suffer from these problems, and can be effectively administered merely by injecting specific cells.
  • stem cells can grow and be transformed into specialized cells having characteristics consistent with cells of various tissues, their use in therapy has been proposed for various incurable diseases.
  • patients suffering from diabetes must be administered with insulin daily or must have the pancreas transplanted from brain-dead patients.
  • Stem cell therapy can be used to treat diabetes simply by injecting stem cells, which then differentiate into insulin-secreting pancreatic cells.
  • Effective medical treatment is also anticipated for Parkinson' s disease by injecting stem cells, which then differentiate into dopamine-secreting cerebral. cells, and for myocardial infarction, merely by injecting stem cells, which differentiate into cardiomyocytes . Further, this medical technology is applicable to cancer treatment.
  • Stem cells can be used for the development of cellular agents involved in apoptosis of cancer.
  • stem cells are administered to reconstruct tissues or organs which to replace the excised tissues or organs in which cancer occurred.
  • tissue engineering which has recently progressed greatly, allows the development of artificial organs (Petit-Zeman S., Regenerative medicine. Nat. Biotechnol. 19: 201-206, 2001, Gussoni E. et al., Nature 401: 390-394, 1999; Kim, Byoung-Soo, Bioindustry. VoI 31, Development of Artificial Organs Stem Cell-Based Tissue Engineering, 2002) .
  • stem cell therapy is a new and promising strategy for the treatment of various incurable diseases. Furthermore, the successful results of stem cell therapy in the recovery of function of damaged organs are noteworthy enough to attract immediate and special attention from patients, scientists and various persons in the medical and pharmaceutical fields. In spite of the positive side thereof, stem cell therapy, however, suffers from many disadvantages, evidenced in some clinical tests using stem cells. Of them, the greatest disadvantages are that it is difficulty to obtain stem cells in an amount sufficient for use in treatment and that cells, after administered, fail to graft after a long engraftment period.
  • Leptin is a cytokine-like protein produced by adipose tissue, which is known to play a key role in regulating energy intake and energy expenditure, including the regulation (decrease) of appetite and (increase) of metabolism.
  • leptin receptors As the expression of leptin receptors (Ob-R) is found in immune cells and hematopoietic stem cells as well as the hypothalamus, which recognizes satiation, leptin is known to be involved in the proliferation and differentiation of hematopoietic stem cells
  • leptin receptors show structural similarity to receptors of gpl30 cytokines
  • leptin is believed to act as a cytokine for hematopoietic stem cells, like Epo, G-CSF and GM-CSF, which play essential roles in the proliferation of stem cells.
  • a method for proliferating stem cells comprising culturing the stem cells in a culture medium supplemented with leptin or an analogue or derivative thereof.
  • the method for proliferating stem cells with leptin in accordance with the present invention has advantages over conventional methods in terms of growth rate, stability and economy. Therefore, it is expected that thanks to the high growth rate and stability thereof, the method featuring the use of leptin in culturing stem cells in accordance with the present invention may be a basic technology for stem cell therapy. Also, the method of the present invention is expected to play an essential role in the development of cell therapeutics for various incurable diseases, thereby making a contribution to the generation of a high value-added industry.
  • FIG. 1 is a graph showing cell counts of the human bone marrow-derived stem cells cultured in serum-free media supplemented or not supplemented with leptin in the presence or absence of a growth factor.
  • FIG. 2 is a graph showing cell counts of the human bone marrow-derived stem cells cultured in serum media supplemented or not supplemented with leptin.
  • FIG. 3 is a graph showing cell counts of the human umbilical cord blood hematopoietic stem cells cultured in serum media supplemented or not supplemented with leptin.
  • FIG. 4 is a graph showing cell counts of the murine bone marrow-derived stem cells cultured in serum-free media supplemented or not supplemented with leptin in the presence or absence of a growth factor.
  • FIG. 5 shows cell counts of the murine bone marrow- derived stem cells cultured in serum-free media not supplemented with any additives ⁇ upper left panel) , and supplemented with leptin (upper right panel) , with G-CSF (lower left panel) and with a combination of leptin and G-CSF (lower right panel) in microscopic photographs.
  • FIG. 6 shows the effect of lejptin on the secretion of cytokines from the murine bone marrow-derived stem cells cultured in culture media in graphs .
  • the present invention pertains to a method for proliferating stem cells, comprising culturing stem cells.
  • Leptin secreted by adipocytes, is known to regulate obesity and appetite. Recent studies indicate that leptin is involved in the proliferation of hematopoietic stem cells. In the present invention, it is revealed that, when supplemented with leptin, cell culture media allow the proliferation of various stem cells in a large quantity without many rounds of sub-culturing. The addition of leptin once to a cell culture medium during the culture of stem cells makes it possible to stably and conveniently increase the number of stem cells without many processes for conventional stem cell proliferation until cell recovery, thereby greatly reducing the production cost.
  • five human hematopoietic stem cells including bone marrow-derived stem cells, bone marrow-derived hematopoietic stem cells, blood stem cells, blood hematopoietic stem cells, and umbilical cord blood hematopoietic stem cells, and murine bone marrow-derived stem cells were isolated and cultured in serum-added or serum-free media supplemented or not supplemented with growth factors in the presence or absence of leptin, followed by counting the cells .
  • Human bone marrow-derived stem cells (bone marrow cells with CD105+, CD166+, CD29+, CD44+, CD14-, CD34- and CD45- markers) were observed to increase four times in counts thereof when cultured in serum-free media in the presence of leptin compared to when cultured in a control, and to a greater extent than when cultured in media supplemented with growth factors (FIG. 1) .
  • FIG. 1 In the case of serum media, far greater effects, e.g., cell counts increased seven times, were observed (FIG. 2) .
  • Human umbilical cord blood hematopoietic stem cells were also observed to increase at least four-fold in number when cultured in the presence of leptin (serum medium) compared to when cultured in the absence of leptin (FIG. 3) .
  • leptin serum medium
  • FIG. 3 Human umbilical cord blood hematopoietic stem cells
  • stem cells useful in the present invention include bone marrow-derived stem cells, bone marrow-derived hematopoietic stem cells, blood stem cells, blood hematopoietic stem cells, umbilical cord blood hematopoietic stem cells, fetal stem cells and adipocyte-derived stem cells, but are not limited thereto.
  • Leptin may be used alone or in concert with conventional proliferation methods, for example, in combination with other growth factors, for example, erythropoietin, G-CSF, GM-CSF, LIF, IL-3, and the like.
  • leptin analogues or leptin derivatives can afford the proliferation effect, as well.
  • any culture medium whether supplemented with serum or not, can be used in the present invention (e.g., Dulbecco's medium or a special medium containing cellulose (e.g., a medium from Stem Cell Technology Inc.).
  • the method of the present invention has an advantage in that stem cells can proliferate to a large quantity within a short period of time even in the absence of additional growth factors or additives.
  • the method of the present invention guarantees approximately eight times as many cells within the same period compared to conventional proliferation methods (after 2 weeks of culture, the number of cells increased 7.17 times : 58.87 times) .
  • the time necessary to reach the same number of stem cells can be reduced approximately 64% by the method of the present invention, compared to conventional methods.
  • stem cells can be grown to an amount sufficient for use in clinical application, thus obviating the repetitive rounds of stem cell extraction processes for cell therapeutic agents.
  • the method of the present invention which can guarantee a sufficient amount of stem cells, is expected to eliminate the problems of immune suppression or infection due to allo- or xenografts.
  • the reduced culture procedure and time period in accordance with the present invention is believed to result in reduced stress to stem cells as well as in the possibility of differences between the original stem cells and the final stem cells due to self-renewal or apoptosis . Therefore, it is anticipated that the proliferation method of stem cells in accordance with the present invention leads to a first economically favorable process, which can be a solution to the prior art problems .
  • human bone marrow-derived stem cells having CD105+, CD166+, CD29+, CD44+, CD14-, CD34- and CD45- markers were used.
  • an Iscove' s Modified Dulbecco' s Medium Gibco-BRL, USA
  • a special medium containing cellulose Stem Cell Technology Inc.
  • insulin transferrin, glutamine, fetal bovine serum and leptin (all from Sigma, USA)
  • G-CSF and erythropoietin both from Gibco, USA
  • the bone marrow was sampled in an amount of 10 ml from persons who were identified to be free of infection and transferred into vacutainer tubes coated with an anticoagulant agent, followed by rotating the vacutainer tubes two or three times to homogenously mix the bone marrow with the anticoagulant agent.
  • the content was homogeneously mixed by shaking the tubes about 20 times prior to centrifugation on a horizontal rotor (swing-out head, Beckman Coulter Inc, USA) at 3000 rpm at 20 0 C for 20 min (within 2 hours after the sampling of the bone marrow) .
  • the upper plasma layer thus separated was removed from the tube and transferred into cryogenic vials in an amount of 0.5 ml per vial.
  • the cell layer and the remaining plasma were well mixed with a pipette, transferred into 50 mL tubes, and diluted to a final volume of 45 ml with saline. After 10 rounds of inversion, 50 ⁇ l of the mixture was used for cell counting while another 50 ⁇ l was stored for subsequent flow cytometry.
  • the tube containing the remainder was centrifuged at 1,500 rpm at room temperature for 5 min, followed by removing the supernatant and suspending the cells in a culture medium.
  • the cell suspension was treated with antibodies (Sigma, USA) to surface markers, CD105, CD166, CD29, CD44, CD14, CD34 and CD45, and the properties thereof were analyzed using a flow cytometry analyzer (Beckman Coulter Inc. USA) , followed by isolation of the cells having surface markers CD105+, CD166+, CD29+, CD44+, CD14-, ,,CD34- and CD45- only. Of them, 50 ⁇ l was stored separately until the measurement of cell count.
  • a culture medium was aliquoted in an amount of 5 ml into 15 ml culture dishes and then supplemented with leptin, serum and other growth factors in predetermined amounts (EPO 3 IU/ml, G-CSF 10 ng/ml, leptin 100 nM) in accordance with test groups (Table 1) , followed by inoculating the cells at a density of 10 6 cells/ml of medium. After 14 days of incubation at 37°C in a 5% CO 2 atmosphere, the cell cultures were centrifuged at 1,500 rpm at room temperature for 5 min. After the removal of the supernatant, centrifugation was again conducted at 1500 rpm at room temperature for 5 min.
  • the cell pellets thus obtained were washed twice with a Hartman Dex solution (Choongwae Pharma Corporation) or Iscove' s MDM, and suspended in 20 ml of a Hartman Dex solution.
  • the cell suspension was filtered through a cell strainer, followed by 10 rounds of inversion 100 ⁇ l of the cell suspension was taken using a micropipette. 50 ⁇ l was used to count the cells while the other 50 ⁇ l was subjected to flow cytometry.
  • EXAMPLE 2 Proliferation of Human Bone Marrow-Derived Hematopoietic Stem cell
  • CD34 (+) cells were isolated from 10 ml of the bone marrow in the same manner as in Example 1 after the pre-blood test (CD34+ progenitor cell isolation kit, MACS, Miltenyi Biotec, Germany) .
  • a blood sample was taken from a suitable site in the arm tied with a tourniquet using a vacuum needle and a scalp needle, both connected to a holder, after which the vacutainer tube was rotated two or three times in order to mix the blood with an anticoagulant agent therein. Centrifugation was conducted within 2 hours after blood sampling. The vacutainer tube was turned upside down about 20 times to homogenously mix the content thereof before centrifugation at 3000 rpm at 20°C for 20 min on a horizontal rotor (swing-out head). The plasma in the upper layer (0.5 cm above the buffy coat layer) was removed using a 15 ml tube and transferred into cryogenic vials at an amount of 0.5 ml per vial.
  • lymphocytes and the remaining plasma in the buffy coat layer were well mixed with a pipette, transferred into 50 iriL tubes and diluted to a final volume of 45 ml with saline. After 10 rounds of inversion, 50 ⁇ l of the mixture was used for cell counting while another 50 ⁇ l was stored for subsequent flow cytometry.
  • the tube containing the remainder was centrifuged at 1,500 rpm at room temperature for 5 min. Following the removal of the supernatant, the cells were suspended in a culture medium and well mixed with Ficoll-Hypaque. Following flotation on Ficoll-Hypaque gradients, stem cells were carefully removed from a layer located 1/3 of the total length from the top thereof?.
  • CD34 (+) cells were isolated from 10 ml of the bone marrow in the same manner as in Example 1 after the pre-blood test (CD34+ progenitor cell isolation kit, MACS, Miltenyi Biotec, Germany) .
  • mice (Charles River, USA) under general anesthesia.
  • the hind legs were dissected, and the tibia were separated from the surrounding soft tissues and cut at both the fore end and the rear end, followed by the injection of a 23G syringe needle thereinto.
  • a culture medium was passed through the 23G syringe needle, the bone marrow was effused.
  • the bone marrow mass thus obtained was loosened into single cells by being passed back and forth in a Pasteur pipette.
  • the culture medium containing the bone marrow was transferred into a 15 ml culture tube and centrifuged at 1300 rpm for 5 min.
  • the culture tube was swung to loosen the cell pellet, which was then suspended with 1 ml of a culture medium. Thereafter, the same procedure as in Example 3 was repeated to isolate and culture the stem cells.
  • the culture tube was centrifuged at 1300 rpm for 5 min when using a general culture medium.
  • the culture tube was allowed to stand for 30 min at 4 0 C after the addition of 5 ml of chilled Iscove' s MDM thereinto.
  • the culture medium was observed to turn from a semi-solid state to a liquid state, it was centrifuged at 1300 rpm for 5 min.
  • Example 3 The supernatant was removed and the cell pellet was dissolved in 5 ml of the culture medium and again centrifuged at 1300 rpm for 5 min, followed by washing the cells twice with a Hartman Dex solution. The same procedure as in Example 3 was repeated to isolate and count the cells .
  • the experimental results after the cell isolation and proliferation are shown in FIGS. 4 and 5.
  • the culture medium was measured for cytokine levels 24 hours after treatment with leptin. The results are shown in FIG. 6.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Biomedical Technology (AREA)
  • Organic Chemistry (AREA)
  • Chemical & Material Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Genetics & Genomics (AREA)
  • Biotechnology (AREA)
  • Wood Science & Technology (AREA)
  • Zoology (AREA)
  • General Engineering & Computer Science (AREA)
  • Biochemistry (AREA)
  • Cell Biology (AREA)
  • General Health & Medical Sciences (AREA)
  • Microbiology (AREA)
  • Developmental Biology & Embryology (AREA)
  • Hematology (AREA)
  • Immunology (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)

Abstract

L'invention porte sur un procédé qui permet la prolifération de cellules souches. La présence de leptine dans un milieu de culture cellulaire permet d'obtenir une prolifération considérable de diverses cellules souches sanguines ou de moëlle osseuse sans augmentation du nombre de sous-cultures. Le procédé précité permet de réduire le temps nécessaire à l'obtention d'une quantité prédéterminée de cellules et, par conséquent, de réaliser des avantages économiques. L'invention permet également la prolifération stable de cellules souches non modifiées.
PCT/KR2007/005670 2006-11-10 2007-11-12 Procédé permettant la prolifération de cellules souches au moyen de leptine WO2008056963A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
KR20060111117 2006-11-10
KR10-2006-0111117 2006-11-10

Publications (1)

Publication Number Publication Date
WO2008056963A1 true WO2008056963A1 (fr) 2008-05-15

Family

ID=39364732

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/KR2007/005670 WO2008056963A1 (fr) 2006-11-10 2007-11-12 Procédé permettant la prolifération de cellules souches au moyen de leptine

Country Status (2)

Country Link
KR (1) KR20080042761A (fr)
WO (1) WO2008056963A1 (fr)

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009155041A2 (fr) * 2008-05-28 2009-12-23 Children's Medical Center Corporation Méthode de modulation de la croissance de cellule souche hématopoïétiquie
US8551782B2 (en) 2006-03-24 2013-10-08 Children's Medical Center Corporation Methods for promoting HSC engraftment
US9051548B2 (en) 2009-02-03 2015-06-09 Children's Medical Center Corporation Methods for enhancing hematopoietic stem/progenitor cell engraftment
US9056085B2 (en) 2009-02-03 2015-06-16 Children's Medical Center Corporation Methods for enhancing hematopoietic stem/progenitor cell engraftment
US9402852B2 (en) 2006-10-20 2016-08-02 Children's Medical Center Corporation Method to enhance tissue regeneration
US9452186B2 (en) 2011-12-02 2016-09-27 Fate Therapeutics, Inc. Enhanced stem cell composition
US10111907B2 (en) 2011-12-02 2018-10-30 Fate Therapeutics, Inc. Methods of treating ischemia
US10851412B2 (en) 2013-03-15 2020-12-01 Fate Therapeutics, Inc. Cell potency assay for therapeutic potential

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR101350144B1 (ko) * 2013-05-03 2014-01-10 재단법인 통합의료진흥원 팔물탕 추출물을 포함하는 골수유래 줄기세포 증식 촉진용 조성물
WO2018226051A2 (fr) 2017-06-07 2018-12-13 주식회사 엑소스템텍 Composition de milieu sans sérum pour la culture de cellules comprenant un exosome dérivé d'une cellule souche humaine

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1998018486A1 (fr) * 1996-10-25 1998-05-07 Icogen Corporation Utilisation de la leptine pour stimuler l'hematopoiese

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1998018486A1 (fr) * 1996-10-25 1998-05-07 Icogen Corporation Utilisation de la leptine pour stimuler l'hematopoiese

Non-Patent Citations (7)

* Cited by examiner, † Cited by third party
Title
BENNETT B.D. ET AL.: "A role for leptin and its cognate receptors in hematopoiesis", CURR. BIOL., vol. 6, no. 9, 1 September 1996 (1996-09-01), pages 1170 - 1180, XP000673008, DOI: doi:10.1016/S0960-9822(02)70684-2 *
CIOFFI J.A. ET AL.: "Novel B219/OB receptor isoforms: possible role of leptin in hematopoiesis and reproduction", NAT. MED., vol. 2, no. 5, May 1996 (1996-05-01), pages 585 - 589, XP002019361, DOI: doi:10.1038/nm0596-585 *
GAINSFORD T. ET AL.: "Leptin can induce proliferation, differentiation, and functional activation of hemopoietic cells", PROC. NATL. ACAD. SCI. U.S.A., vol. 93, no. 25, 10 December 1996 (1996-12-10), pages 14564 - 14568, XP002954111, DOI: doi:10.1073/pnas.93.25.14564 *
MIKHAIL A.A. ET AL.: "Leptin stimulates fetal and adult erythroid and myeloid development", BLOOD, vol. 89, no. 5, 1 March 1997 (1997-03-01), pages 1507 - 1512, XP002921616 *
OEZ S. ET AL.: "LW/SO cell line: a tool for studying the phenotypical characterization and commitment of hematopoietic stem cells", ANN. HEMATOL., vol. 72, no. 5, May 1996 (1996-05-01), pages 307 - 316 *
SIVAN E. ET AL.: "Leptin is present in human cord blood", DIABETES, vol. 46, no. 5, May 1997 (1997-05-01), pages 917 - 919 *
UMEMOTO Y. ET AL.: "Leptin stimulates the proliferation of murine myelocytic and primitive hematopoietic progenitor cells", BLOOD, vol. 90, no. 9, 1 November 1997 (1997-11-01), pages 3438 - 3443 *

Cited By (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10272110B2 (en) 2006-03-24 2019-04-30 Children's Medical Center Corporation Methods for promoting HSC engraftment
US8551782B2 (en) 2006-03-24 2013-10-08 Children's Medical Center Corporation Methods for promoting HSC engraftment
US8563310B2 (en) 2006-03-24 2013-10-22 Children's Medical Center Corporation Methods for promoting hematopoietic reconstitution
US9028811B2 (en) 2006-03-24 2015-05-12 Children's Medical Center Corporation Methods for promoting HSC self-renewal
US10278990B2 (en) 2006-03-24 2019-05-07 Children's Medical Center Corporation Methods for promoting hematopoietic reconstitution
US10736906B2 (en) 2006-10-20 2020-08-11 Children's Medical Center Corporation Method to enhance tissue regeneration
US9402852B2 (en) 2006-10-20 2016-08-02 Children's Medical Center Corporation Method to enhance tissue regeneration
WO2009155041A3 (fr) * 2008-05-28 2010-03-11 Children's Medical Center Corporation Méthode de modulation de la croissance de cellule souche hématopoïétiquie
WO2009155041A2 (fr) * 2008-05-28 2009-12-23 Children's Medical Center Corporation Méthode de modulation de la croissance de cellule souche hématopoïétiquie
US9051548B2 (en) 2009-02-03 2015-06-09 Children's Medical Center Corporation Methods for enhancing hematopoietic stem/progenitor cell engraftment
US10092599B2 (en) 2009-02-03 2018-10-09 Children's Medical Center Corporation Methods for enhancing hematopoietic stem/progenitor cell engraftment
US10159697B2 (en) 2009-02-03 2018-12-25 Children's Medical Center Corporation Methods for enhancing hematopoietic stem/progenitor cell engraftment
US9737567B2 (en) 2009-02-03 2017-08-22 Children's Medical Center Corporation Methods for enhancing hematopoietic stem/progenitor cell engraftment
US9056085B2 (en) 2009-02-03 2015-06-16 Children's Medical Center Corporation Methods for enhancing hematopoietic stem/progenitor cell engraftment
US10111907B2 (en) 2011-12-02 2018-10-30 Fate Therapeutics, Inc. Methods of treating ischemia
US10172888B2 (en) 2011-12-02 2019-01-08 Fate Therapeutics, Inc. Enhanced stem cell composition
US9452186B2 (en) 2011-12-02 2016-09-27 Fate Therapeutics, Inc. Enhanced stem cell composition
US10980838B2 (en) 2011-12-02 2021-04-20 Fate Therapeutics, Inc. Methods of treating ischemia
US11052118B2 (en) 2011-12-02 2021-07-06 Fate Therapeutics, Inc. Enhanced stem cell composition
US10851412B2 (en) 2013-03-15 2020-12-01 Fate Therapeutics, Inc. Cell potency assay for therapeutic potential

Also Published As

Publication number Publication date
KR20080042761A (ko) 2008-05-15

Similar Documents

Publication Publication Date Title
WO2008056963A1 (fr) Procédé permettant la prolifération de cellules souches au moyen de leptine
Sutherland et al. Quantitative assays for human hemopoietic progenitor cells
JP6053853B2 (ja) 間葉系幹細胞を調製する方法、その組成物及びキット
Dexter et al. Stromal cells in haemopoiesis
US20080118477A1 (en) Umbilical cord mesenchymal stem cells support cord blood hematopoiesis
AU2018232905A1 (en) Method of producing erythrocytes without feeder cells
EP2669368B1 (fr) Isolation et purification de cellules souches hématopoïétiques à partir d'aspirâts de liposuccion
US20140295555A1 (en) Method of culturing cells
US20050186182A1 (en) Methods of using G-CSF mobilized C-Kit+ cells in the production of embryoid body-like cell clusters for tissue repair and in the treatment of cardiac myopathy
CA2682243C (fr) Procedes et moyen destine a la proliferation des cellules souches et a la generation et a l'expansion subsequente de cellules progenitrices et production de cellules effectrices utilisees commes agents therapeutiques cliniques
BR112014020119A2 (pt) cultura de células-tronco mesenquimais
WO2011069121A1 (fr) Cellules souches mésenchymateuses (csm) isolées dans le sang périphérique mobilisé
JP2003513664A (ja) ヒト胚性幹細胞の造血分化
KR20120008223A (ko) 양막유래 중간엽 줄기세포 배양을 위한 배지조성물 및 이를 이용한 양막유래 중간엽 줄기세포의 배양방법
SG177143A1 (en) Lithium stimulation of cord blood stem cell proliferation and growth factor production
JP2002502617A (ja) 幹細胞および前駆細胞の増殖と分化を制御する方法
Palpant et al. Non-canonical Wnt signaling enhances differentiation of Sca1+/c-kit+ adipose-derived murine stromal vascular cells into spontaneously beating cardiac myocytes
US20210032596A1 (en) Method of producing erythrocytes
JP2011501960A (ja) 月経血幹細胞と共に臍帯血由来細胞を共培養する方法
WO2022120153A1 (fr) Procédé d'entretien ou d'expansion ex vivo à long terme de cellule d'érythroblaste humain, de cellule progénitrice de mégacaryocytes-érythrocytes humains ou de cellule progénitrice myéloïde commune
CN106190979B (zh) 体外培养造血干/前驱细胞的方法及其组成物
US8802434B2 (en) Biological cell culture, cell culture media and therapeutic use of biological cells
Ookura et al. Adipocyte differentiation of human marrow mesenchymal stem cells reduces the supporting capacity for hematopoietic progenitors but not for severe combined immunodeficiency repopulating cells
CN116396930B (zh) 间充质干细胞无血清培养基及其应用
RU2722669C1 (ru) Способ получения ассоциатов гемопоэтических и стромальных клеток-предшественников, способных подавлять активацию и пролиферацию аллогенных лимфоцитов

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 07833977

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 07833977

Country of ref document: EP

Kind code of ref document: A1