![]() culture of mesenchymal stem cells
专利摘要:
MESENQUIMAL STEM CELL CULTURE, provides methods for culture of mesenchymal stem cells. The methods comprise culturing MSC'S in a medium comprising nicotinamide and fibroblast growth factor 4 (FGF4). Populations of mesenchymal stem cells generated using the methods described here and their uses are also provided. 公开号:BR112014020119A2 申请号:R112014020119-6 申请日:2013-02-13 公开日:2020-10-27 发明作者:Tony Peled;Yair Steinhardt 申请人:Gamida-Cell Ltd; IPC主号:
专利说明:
[01] [01] The present patent application, in some respective applications, relates to methods for expanding mesenchymal stem cells and cell populations generated in this way. [02] [02] Mesenchymal stem cells (MSC's | mesenchymal stem cells) are non-hematopoietic cells capable of differentiating into specific types of mesenchymal or connective tissues, including adipose, bone, cartilaginous, elastic, neuronal, hepatic, pancreatic, muscular and fibrous. The specific path of differentiation through which these cells enter depends on various influences, from mechanical influences and / or endogenous bioactive factors, such as growth factors and cytokines, and / or local microenvironmental conditions established by the host tissue. [03] [03] MSCs reside in a variety of tissue hosts throughout the adult organism and have the ability to 'regenerate' specific cell types for those tissues. Examples of such tissues include adipose tissue, umbilical cord blood, periosteum, synovial membranes, muscles, dermis, pericytes, blood, bone marrow and trabecular bone. [04] [04] The multipotent nature of mesenchymal stem cells makes these cells an attractive therapeutic tool and a candidate for transplants, capable of fulfilling the role in a wide range of clinical applications in the context of both cell and gene therapy strategies. Mesenchymal cells can be used to enhance post-transplant hematopoietic graft, correct acquired and inherited disorders of bones and cartilage, to implant prosthetic devices in connective and skeletal tissues, and as vehicles for gene therapy. [05] [05] In cultivation, expanded MSCs express a panel of key markers, including CD105 (endogline, SsH2), CD73 (ecto-5 'nucleotidase, SH3, SH4), CDI66 (ALCAM), CD29 (Bl-integrin), CD44 (H-CAM) and CD90 (Thy-l1). In contrast to hematopoietic stem cells, they lack expression of CD45, CD34 and CD133. [06] [06] MSCs can be identified by their ability to form fibroblast unit-forming colonies (CFU-F | colony forming units-fibroblast) in vitro. However, these cells are heterogeneous with regard to their ability to proliferate and differentiate. At least two morphologically distinct populations of MSCs have been identified because they differ not only in size, but also in their cell division rates and differentiation capacity. In addition, the analysis of colonies of MSC's derived from single cells of adult bone marrow revealed a differential capacity of the colonies to undergo osteogenic, adipogenic and chondrogenic differentiations. [07] [07] In most cases, unfractionated cells derived from bone marrow are used as an initial population for the cultivation of MSC's. This isolation method depends on the adhesion of fibroblast-like cells to a plastic surface and the removal of non-adherent hematopoietic cells. The resulting cells are poorly defined and give rise not only to heterogeneous MSC populations, but also to osteoblast and / or osteoprogenitor cells, fat cells, reticular cells, macrophages and endothelial cells. To define the initial population more precisely, surface markers, such as SH2 (CD1I05), SH3 / SH4 (CD73), SSEA-4 and the low-affinity nerve growth factor receptor (CD271), which enrich for MSC's, were employed [Simmons PJ et al. (1991) Blood 78:55 to 62; Conconi MT et al., (2006) Int J Mol Med 18: 1089 to 1096; Gang EJ et al., (2007) Blood 109: 1743 to 1751; Liu PG, (2005); Zhongguo Shi Yan Xue Ye Xue Za Zhi 13: 656 to 659; Quirici N, et al., (2002) Exp. Hematol 30: 783 to 791]. Another example of a cell surface antigen that was targeted for isolation of homogeneous populations of mesenchymal stem cells is the precursor-1 stromal antigen ( STRO-1). The STRO-1 antigen is expressed on the surface of approximately 10 to 20% of adult human BM that includes all CFU-F, Glycophorin-A nucleated red blood cells, and a small subset of CD19 B cells, but is not expressed in cells -hematopoietic stem (HSC | hematopoietic stem cells) and progenitors (Simmons and Torok- Storb, 1991). STRO-1 is widely regarded as a marker for mesenchymal / stromal precursor cells, as it has been strongly linked to clonogenicity, plasticity and other characteristics of mesenchymal progenitor cells [Psaltis et al., (2010), Journal of Cellular Physiology, 530 to 540]. The high co-expression of STRO-1 (STRO-lBright) with other surface markers, such as CD1I06, CD49a, CDI46 or STRO-3, have been shown to greatly increase the cloning efficiency of BM MNC (Gronthos et al., 2008 , Methods Molecular Biology, 449: 45 to 57). The newly isolated STRO-1 "**" 'BM MNC also have other trademark features that are characteristic of multipotent stem cells, including in vivo stillness, high telomerase activity, and an undifferentiated phenotype. In addition, this cell population lacks hematopoietic stem cells (CD34), leukocytes (CD45), and associated erythroid markers (Glycophorin-A). [08] [08] More recently, the platelet derived growth factor-receptor (PDGF-RB | platelet derived growth factor receptor-fb; CD1I40b) has been identified as a selective marker for the isolation of clonogenic MSCs [Buhring HJ, (2007 ) Ann NY [09] [09] Although MSCs multiply relatively easily in vitro, their proliferative potential and stem cell characteristics are continually diminished during prolonged cultivation. For example, it has been shown that expansion in culture leads to premature senescence (the aging process characterized by continuous morphological and functional changes). The cells become much larger, with an irregular and flat shape, and the cytoplasm becomes more granular. These effects associated with senescence are continuously acquired since the beginning of in vitro culture (PLoS ONE, May 2008 | Volume 3 | Edition 5 | and 2213). As a result, the successful manufacture of large batches from a donor of homogeneous MSCs that preserve their characteristics after an expansion in culture remains a challenge. [010] [010] Due to the low or absent expression of MHC molecules, especially class II mesenchymal stem cells, these cells can be considered immune, thus paving the way for the allogeneic transplantation of such cells for the treatment of a wide variety of disorders. Consequently, improved methods of expanding banks of mesenchymal stem cells have become an important factor in commercializing their use. [011] [011] The role of growth factors in increasing proliferation and survival in MSCs has been extensively studied in recent years, and many factors have been proposed to increase the expansion efficiency of these cells. [012] [012] For example, many protocols regarding the expansion of MSCs include cultivation in the presence of basic fibroblast growth factor (b-FGF | basic fibroblast growth factor) (Vet Res Commun. 2009 Dec; 33 (8): 811 a 821). It has been shown that b-FGF not only maintains the potential for proliferation of MSCs, but also retains the potentials for osteogenic, adipogenic and chondrogenic differentiation in all early mitogenic cycles. [013] [013] The vascular endothelial growth factor (VEGF | vascular endothelial growth factor) has also been shown to increase the proliferation of MSCs [Pons et al., Biochem Biophys Res Commun 2008, 376: 419 to 422]. [014] [014] The exogenous addition of Hepatocyte growth factor (HGF | hepatocyte growth factor) to MSC populations has been shown to affect proliferation, migration and differentiation (Furge et al., Oncogene 2000, 19: 5582 to 5589]. [015] [015] Another growth factor proposed to increase the expansion of MSC's is the platelet-derived growth factor (PDGF | Platelet derived growth factor), which has been shown to be a potent mitogen of MSC's [Kang et al., J Cell Biochem 2005, 95: 1135 to 1145). [016] [016] Both the epidermal growth factor (EGF | epidermal growth factor) and the EGF in connection with heparin have been shown to promote the expansion of ex-vivo MSC's without triggering differentiation in any specific strain [Tamama et al., Stem Cells 2006 , 24: 686 to 695; Krampera et al., Blood 2005, 106: 59 to 66]. In addition to its mitogenic effect on MSCs, EGF also increases the number of colony-forming units by 25% [Tamama et al., J Biomed Biotechnol 2010, 795385]. [017] [017] Other authors have suggested the use of Wnt signaling agonists to expand MSC '”s based on experiments that study the proliferation of Wnt signaling in MSC'” s. Canonical Wnt signaling has been shown to keep stem cells in an undifferentiated but self-renewing state. The addition of Wnt3a by activating the canonical pathway of Wnt increased both proliferation and survival, while avoiding differentiation in the osteoblastic lineage in MSC's [Boland et al., J Cell Biochem 2004, 93: 1210 to 1230]. [018] [018] The choice of using growth factors in MSCs was initially determined based on previously existing knowledge about the effect of a specific growth factor on cell morphogenesis. This was done with the dual intention of expanding MSCs and making them differentiate in the line in which they are favored. Transforming growth factor beta (TGFB | Transforming growth factor beta), for example, is known to influence chondrogenic lineage cells in vivo, promoting the early stages of mesenchymal condensation, precondrocyte proliferation, extracellular matrix production and deposition of specific cartilage molecules, while inhibiting terminal differentiation. When applied to MSC's, cells demonstrate increased proliferation and a tendency toward chondrogenic lineage [Bonewald et al., JyJ Cell Biochem 1994, 55: 350 to 357; Longobardi L, J Bone Miner Res 2006, 21: 626 to 636]. [019] [019] BMP-3, another member of the transforming growth factor beta family, known to improve bone differentiation, has been shown to increase the proliferation of MSCs three times [Stewart A et al., Cell Physiol 2010, 223: 658 a 666]. [020] [020] Nicotinamide (NA | nicotinamide), an amide form of niacin (vitamin B3), is an exchange-based substrate and a potent inhibitor of NAD (+) -dependent enzymes endowed with mono and poly-ADP-ribosyltransferase activities. ADP-ribosylation is involved in the modification of a diverse matrix of biological processes (Corda D, Di Girolamo M. 2003; 22 (9): 1953 to 1958; Rankin [021] [021] WO 07/063545 discloses the use of nicotinamide for the expansion of hematopoietic stem cells and / or populations of progenitor cells. [022] [022] US Patent Application No. 20050260748 instructs the isolation and expansion of mesenchymal stem cells with nicotinamide in the presence of a low concentration of calcium. [023] [023] Additional history of the technique includes Farre et al., Growth Factors, 2007 Apr; 25 (2): 71 to 76. SUMMARY [024] [024] According to one aspect of some applications of the present patent application, a method of culturing mesenchymal stem cells (MSC's) is provided comprising culturing a population of MSC's in a medium containing nicotinamide and factor 4 of fibroblast growth (FGF4 | fibroblast growth factor 4), thus cultivating MSC's. [025] [025] In accordance with an aspect of some applications of the present patent application, a method of expanding a population of stem cells is provided - "mesenchymal, the method comprising culturing a population sown" of mesenchymal stem cells for a period of time sufficient for cell expansion, characterized in that in at least a portion of the time period, the culture is carried out in a medium devoid of nicotinamide, and in at least a second portion of the time period, the culture be carried out in a medium comprising nicotinamide and FGF4, thus generating an expanded population of mesenchymal stem cells. [026] [026] In accordance with an aspect of some applications of the present application, a method is provided to generate cells useful for transplantation in an individual, the method comprising: (a) culturing mesenchymal stem cells, in accordance with with the methods described in this document, to generate a population of cultured mesenchymal stem cells; (b) contact with the population of mesenchymal stem cells cultured with a differentiating agent, thus generating useful cells for transplantation in an individual. [027] [027] In accordance with an aspect of some applications of the present application, a method is provided to generate cells useful for transplantation, the method comprising: (a) the expansion of mesenchymal stem cells according to the methods described in this document; and (b) the contact of mesenchymal stem cells with a differentiating agent, thus generating useful cells for transplantation. [028] [028] In accordance with an aspect of some applications of the present application, an isolated population of mesenchymal stem cells generated according to the methods described in this document is provided. [029] [029] An isolated population of differentiated cells generated according to the methods described in this document. [030] [030] In accordance with an aspect of some applications of the present application, a method of treating a disease or disorder is provided, the method comprising transplanting, in an individual in respective need, a therapeutically effective amount of the isolated population cells described in this document, thereby treating the disease or disorder. [031] [031] According to one aspect of some applications of the present application, a cell culture comprising stem cells — mesenchymal and a medium comprising nicotinamide and FGF4 is provided. [032] [032] According to some applications of the present patent application, the medium comprises DMEM (Dulbecco's modified Eagle medium | Eagle medium modified by Dulbecco). [033] [033] According to some applications of the present patent application, the medium comprises serum or platelet lysate. [034] [034] According to some applications of the present patent application, mesenchymal stem cells are derived from tissue selected from the group consisting of bone marrow, adipose tissue, and blood from the placenta and umbilical cord. [035] [035] According to some applications of the present application, nicotinamide is selected from a group consisting of nicotinamide, a nicotinamide analog, a nicotinamide metabolite, a nicotinamide analog metabolite and respective derivatives. [036] [036] According to some applications of the present patent application, the culture is carried out on a plastic surface. [037] [037] According to some applications of the present patent application, the population of MSC's is comprised of a heterogeneous population of cells. [038] [038] According to some applications of the present patent application, at least 70% of the heterogeneous cell population are MSC's. [039] [039] According to some applications of the present patent application, the calcium concentration of the medium is greater than 1.8 mM. [040] [040] According to some applications of the present patent application, the culture is carried out for at least 1 week. [041] [041] According to some applications of the present patent application, the culture is carried out by at least 3 passages. [042] [042] According to some applications of the present patent application, the concentration of nicotinamide is 1 to 20 mM. [043] [043] According to some applications of the present patent application, the medium is devoid of platelet-derived growth factor (PDGF). [044] [044] According to some applications of the present patent application, the expansion is carried out under conditions that do not induce the differentiation of mesenchymal stem cells. [045] [045] According to some applications of the present patent application, the population sown of mesenchymal stem cells was sown in the absence of nicotinamide. [046] [046] According to some applications of the present patent application, the population sown of mesenchymal stem cells was sown in the presence of nicotinamide. [047] [047] According to some applications of the present patent application, the medium is devoid of platelet-derived growth factor (PDGF). [048] [048] According to some applications of the present patent application, the medium devoid of nicotinamide is devoid of FGF4. [049] [049] According to some applications of the present patent application, the nicotinamide-free medium comprises FGF4. [050] [050] According to some applications of the present application, the culture in the medium comprising nicotinamide is carried out before the culture of the medium devoid of nicotinamide. [051] [051] According to some applications of the present application, the culture in the medium devoid of nicotinamide is carried out before the culture in the medium comprising nicotinamide. [052] [052] According to some applications of the present patent application, the culture in the medium comprising nicotinamide is carried out for at least one day. [053] [053] According to some applications of the present patent application, the culture in the medium comprising nicotinamide is carried out for at least one week. [054] [054] According to some applications of the present patent application, the culture in the medium devoid of nicotinamide is carried out for at least one day. [055] [055] According to some applications of the present patent application, the culture in the medium devoid of nicotinamide is carried out for at least one week. [056] [056] According to some applications of the present patent application, the culture in the medium comprising nicotinamide is carried out in a medium comprising calcium, characterized in that the calcium concentration is greater than 1.8 mM. [057] [057] According to some applications of the present patent application, the culture in the medium devoid of nicotinamide is carried out in a medium comprising calcium, characterized in that the calcium concentration is greater than 1.8 mM. [058] [058] According to some applications of the present patent application, the differentiating agent comprises a growth factor. [059] [059] According to some applications of the present patent application, the differentiating agent comprises a polynucleotide that encodes the differentiating agent. [060] [060] According to some applications of the present application, the polynucleotide encodes the 6 bone morphogenic protein 2 (BMP2 | bone morphogenic protein 2). [061] [061] According to some applications of the present patent application, the isolated population of mesenchymal stem cells is substantially homogeneous. [062] [062] According to some applications of the present patent application, at least 40% of the cells express VCAM1 / CD106. [063] [063] According to some applications of the present patent application, at least 90% of the cells have a diameter of less than 20 µm. [064] [064] According to some applications of the present patent application, the isolated population of mesenchymal stem cells is less granular than mesenchymal stem cells generated under identical conditions, but in the absence of nicotinamide. [065] [065] According to some applications of the present patent application, less than 30% of the cells express CD45, more than 95% of the cells express CD90 and more than 90% of the cells express CD105 and CD44, [066] [066] According to some applications of the present patent application, the disease or disorder is selected from the group consisting of a bone or cartilaginous disease, a neurodegenerative disease, a heart disease, a liver disease, cancer, nerve damage, autoimmune diseases, GvHD (Graft-versus-host disease | DECH [Graft Against Host Disease]), wound healing and tissue regeneration. [067] [067] According to some applications of the present patent application, mesenchymal stem cells cultured with nicotinamide and / or nicotinamide and FGF4 secrete high levels of growth factors and reduced levels of pro-inflammatory factors in the medium. [068] [068] Unless otherwise defined, all technical and scientific terms used herein have the same meaning as would normally be understood by someone of ordinary skill in the technique to which the present application for a patent belongs. Although methods and materials similar or equivalent to those described herein can be used in the practice or testing of the present patent application, suitable methods and materials are described below. In case of conflicts, the patent specification, including definitions, will prevail. In addition, the materials, methods and examples are illustrative only and should not be limiting. BRIEF DESCRIPTION OF THE FIGURES [069] [069] The patent or application file contains at least one figure represented in colors. Copies of that patent or patent application publication with colored figure (s) will be provided by the Office upon request and payment of the necessary fee. [070] [070] Some applications of the present invention patent application are described in this document, by way of example only, with reference to the figures and images accompanying them. With specific reference now to the figures in detail, it is noted that the specific ones are shown by way of example and for the purpose of illustrative discussion of the applications of the present application for an invention patent. In this regard, the description taken with the figures becomes apparent to those skilled in the art as the applications of the present invention patent application can be practiced. [071] [071] Figure 1 is a bar graph illustrating that the basic fibroblast growth factor (bFGF | basic fibroblast growth factor) has a negative effect on nicotinamide's ability to increase mesenchymal stem cell proliferation. [072] [072] Figures 2A - B illustrate that the growth factor similar to the binding of EGF to heparin (HB-EGF | heparin-binding EGF-like growth factor) has a negative effect on the ability of nicotinamide to increase the proliferation of two different batches of mesenchymal stem cells. [073] [073] Figures 3A - D are bar graphs illustrating the synergistic activity of nicotinamide (NAM) and FGF4 in the expansion of mesenchymal stem cells. Four different batches of MSC cultures were treated with FGF4 (50 ng / ml), NAM (5 mM) or a combination of FGF4 + NAM. Cumulative cell counts are displayed at the indicated passages. [074] [074] Figures 4A - B are graphs illustrating that nicotinamide (NAM) preserves the non-differentiated state of MSC's grown with FGF4. Two different batches of MSC 's cultures were treated with FGF4 (50 ng / ml), NAM (5 mM) or a combination of FGF4 + NAM. The cell size was analyzed by the Cedex cell counter. [075] [075] Figures 5A - D are graphs illustrating that cells expanded with a combination of NAM + FGF4 are non-differentiated MSC's (CDI05 + CD45-). Four different batches of MSC cultures were treated with FGF4 (50 ng / ml), NAM (5 mM) or a combination of FGF4 + NAM. The percentage of MSC (CD115 + CD45-) was analyzed by FACS. [076] [076] Figures 6A - D are bar graphs illustrating inconsistent results obtained after the expansion of MSC's with NAM + PDGF-BB. Four different batches of MSC cultures were treated with PDGF-BB (50 ng / ml), NAM (5 mM) or a combination of PDGF-BB + NAM. Cumulative cell counts are displayed at the indicated passages. [077] [077] Figures 7A - D are graphs illustrating that MSC's cultures treated with PDGF-BB or a combination of PDGF-BB + NAM comprise a greater fraction of cells, except for MSC's that contaminate cultures when compared to MSC's grown on absence of PDGF-BB. Four different batches of MSC cultures were treated with PDGF-BB (50 ng / ml), NAM (5 mM) or a combination of PDGF-BB + NAM. The percentage of MSC (CDI05 + CD45-) was analyzed by FACS. [078] [078] Figures 8A - B are bar graphs illustrating a consistent synergistic effect between NAM and FGF4, in contrast to the absence of a synergistic or additive effect between FGF4 and PDGF-BB. In addition, a combination of NAM, FGF4 and PDGF-BB had an adverse effect on the expansion of MSC's. MSC's cultures were treated with PDGF-BB (50 ng / ml), FGF4 (50 ng / ml) and NAM (5 mM), or with a combination of two or three factors, as indicated. Cumulative cell counts are displayed at the indicated passages. [079] [079] Figures 9A - B are graphs illustrating that the PDGF-BB supports cell expansion, except for MSCs in MSC cultures. This effect is not alleviated by NAM and / or FGF4. MSC's cultures were treated with PDGF-BB (50 ng / ml), FGF4 (50 ng / ml) and NAM (5 mM), or with a combination of two or three factors, as indicated. The percentage of MSC (CDIO5 + CD45-) was analyzed by FACS. [080] [080] Figures 10A - H are photographs from day 34 of the culture of MSC's, illustrating that PDGF-BB supports cell expansion, except for MSC's in cultures of MSC's. This effect is not alleviated by NAM and / or FGF4. MSC's cultures were treated with PDGF-BB (50 ng / ml), FGF4 (50 ng / ml) and NAM (5 mM), or with a combination of two or three factors, as indicated. [081] [081] Figure 11 is a bar graph illustrating the percentage of adherent cells derived from BM, expressing mesenchymal stem cell markers in the +/- NAM seeded culture, before the first pass. Mononuclear cells were isolated from the bone marrow using Ficoll and the method of "plastic adhesion" in the presence or absence of Nicotinamide. The non-adherent cells were washed between 3 and 4 days later, and the medium was changed every 3 or 4 days. FACS analyzes were performed in order to obtain expression levels of surface molecules before the first pass (8 days after sowing). [082] [082] Figures 12A - C are graphs illustrating the phenotypic nature of mesenchymal stem cells derived from adipose tissue after six passes in different concentrations of nicotinamide. [083] [083] Figure 13 is a bar graph illustrating the phenotypic nature of bone marrow-derived mesenchymal stem cells after the first passage of treated cultures +/- in a different concentration of nicotinamide. Mononuclear cells were isolated from the bone marrow using Ficoll and the method of "plastic adhesion". The non-adherent cells were washed between 3 and 4 days later, and the medium was changed every 3 or 4 days. FACS analyzes were performed in order to obtain expression levels of surface molecules after the first pass (8 days after sowing). [084] [084] Figure 14 is a bar graph illustrating the effect of different concentrations of nicotinamide (added in passage 3, and with each subsequent passage) on the number of MSC's in passage 6. Nicotinamide considerably increased the expansion of mesenchymal stem cells. derived from adipose tissue in culture. [085] [085] Figure 15 is a graph illustrating the effect of nicotinamide on the expansion of bone marrow-derived mesenchymal stem cells. Nicotinamide was added from the beginning of the cultivation and with each subsequent passage. [086] [086] Figure 16 is a graph illustrating the effect of different concentrations of nicotinamide on the expansion of adipose-derived mesenchymal stem cells. Nicotinamide was added from passage 3 and to each subsequent passage. [087] [087] Figures 17A - B illustrate that mesenchymal stem cells cultured in nicotinamide expand more rapidly than stem cells [088] [088] Figure 18 is a graph illustrating the effect of nicotinamide on the cumulative cell count of mesenchymal stem cells grown in a large batch. [089] [089] Figure 19 is a bar graph illustrating the results of one or two experiments - * performed, illustrating that the effect of nicotinamide on the proliferation of mesenchymal stem cells is not dependent on the specific lot of serum used. [090] [090] Figures 20A - C are graphs and drawings illustrating the beneficial effect of culture in the presence of nicotinamide on cell size and granularity. Figure 20C demonstrates that cells cultured in the presence of nicotinamide are smaller and less granular (most cells are in the red circle), as opposed to cells cultured without nicotinamide, which are larger and more granular (black circle). For Figure 20A, the concentration of nicotinamide used was 5 mM. [091] [091] Figures 21A - B are graphs and drawings illustrating that mesenchymal stem cells grown in the presence of nicotinamide are less granular than mesenchymal stem cells grown in the absence of nicotinamide, under identical conditions. [092] [092] Figure 22 is a graph illustrating that culturing MSC's in the presence of nicotinamide (5 mM) increases the cumulative CFU-F count. [093] [093] Figures 23A - D are photographs illustrating that nicotinamide reduces the amount of senescence of mesenchymal stem cells. Mesenchymal stem cells derived from bone marrow were cultured for 5 passages of +/- 5 mM NAM. The cells were fixed, and the X-Gal tag was performed to detect senescent cells (blue tag). [094] [094] Figures 24A - B are bar graphs illustrating that nicotinamide modulates the expression of surface markers in mesenchymal stem cells - VCAM1 / CD106 (Figure 24A) and CD54 (Figure 24B). Observe the increased expression of VCAM1 / CD1I06 and the reduced expression of CD54 in cells cultured in the presence of nicotinamide; [095] [095] Figures 25A to B are photographs illustrating the results of an in vitro wound healing assay, which was performed with MSC's grown with (Figure 25B) or without (Figure 25A) nicotinamide in passage 3. Wound healing was observed 4 days after the formation of the wound. [096] [096] Figure 26 is a graph illustrating the effect of nicotinamide on the duplication interval of bone marrow-derived mesenchymal stem cells. Nicotinamide was added from the beginning of the cultivation and with each subsequent passage. [097] [097] Figure 27 is a graph illustrating the effect of nicotinamide, with and without FGF4, on the proliferation of bone marrow-derived MSCs, through culture passages. The bone marrow-derived mesenchymal stem cells were isolated using Ficoll and the plastic adhesion method, and were cultured by several passages with fetal bovine serum. -NAM -FGF4 = controls (light blue circles), -NAM + FGF4 = culture with 50 ng / ml FGF4 (dark blue circles), + NAM -FGF4 = culture with 5 mM NAM (pink circles ), + NAM + FGF4 = culture with 50 ng / ml FGF4 and 5 mM NAM (circles in red). Note the synergistic effect of Nicotinamide and FGFA4, added together, through all the passages of MSC's proliferation. [098] [098] Figure 28 is a bar graph illustrating the improvement in the content of Hepatocyte Growth Factor (HGF | Hepatocyte Growth Factor) in conditioned medium from MSC's cultures treated with nicotinamide and FGF4. Bone marrow mesenchymal stem cells were isolated using Ficoll and the plastic adhesion method, and were cultured by several passages with fetal bovine serum, with aggregated nicotinamide and FGF4 (+ NAM + FGF4), with aggregated nicotinamide (+ NAM -FGF4) and without nicotinamide or FGF aggregates (-NAM-FGF4). Twenty-four hours after passage 4, the medium was changed and fresh medium without fetal bovine serum or FGF4 was added. The medium grown from the cultures of passage 4 was collected and tested for HGF content by ELISA. -NAM, -FGF4 = control; + NAM -FGF4 = 5 mM NAM, + NAM + FGF4 = 5 mM NAM + 50 ng / ml FGF4. Note the significant effect of nicotinamide and FGF4 combined on HGF secretion. [099] [099] Figure 29 is a bar graph illustrating the improvement in the content of Transforming Growth Factor-B (TGFB) of the conditioned medium from MSC's cultures treated with nicotinamide and FGF4. Bone marrow mesenchymal stem cells were isolated and cultured as in Figure 28, above. The culture medium was changed to a medium without fetal bovine serum or without FGF4, 24 hours before passage 4, collected from the cultures of passage 4 and tested for TGFRB content by ELISA. -NAM, -FGF4 = control; + NAM -FGF4 = 5 mM NAM, + NAM + FGF4 = 5 mM NAM + 50 ng / ml FGF4. Note the significant effect of nicotinamide and FGF4 combined on the secretion of TGFÊ. [0100] [0100] Figure 30 is a bar graph illustrating the improvement in the content of the Keratinocyte Growth Factor (KGF | Keratinocyte Growth Factor) of the conditioned medium from MSC's cultures treated with nicotinamide and FGF4. Bone marrow mesenchymal stem cells were isolated and cultured as in Figure 28, above. The culture medium was changed to a medium without fetal bovine serum or without FGF4, 24 hours before passage 4, collected from the cultures of passage 4 and tested for KGF content by ELISA. -NAM, [0101] [0101] Figure 31 is a bar graph illustrating the reduction in the cytokine content IL-6 (IL-6) of the conditioned medium from MSC's cultures treated with nicotinamide and FGF4. Bone marrow mesenchymal stem cells were isolated and cultured as in Figure 28, above. The culture medium was changed to a medium without fetal bovine serum or without FGF4, 24 hours before passage 4, collected from the cultures of passage 4 and tested for IL-6 content by ELISA. -NAM, -FGF4 = control; + NAM -FGF4 = 5 mM NAM, + NAM + FGF4 = 5 mM NAM + 50 ng / ml FGF4. Note the significant reduction through nicotinamide and FGF4 combined in IL-6 secretion. [0102] [0102] Figure 32 is a graph illustrating the effect of nicotinamide, with and without FGF4, on the proliferation of MSC's derived from adipose tissue, through 4 culture passages. The mesenchymal stem cells derived from adipose tissue were isolated using collagenase digestion and the plastic adhesion method, and were cultured by several passages with fetal bovine serum. -NAM -FGF4 = controls (blue diamonds), + NAM -FGF4 = culture with 5 mM NAM (red squares), + NAM + FGF4 = culture with 50 ng / ml FGF4 and 5 mM NAM (triangles in green). Note the synergistic effect of [0103] [0103] Figure 33 is a graph detailing the effect of nicotinamide, with and without FGF4, on the proliferation of nucleated cells during the proliferation of MSCs derived from adipose tissue in passage 4. Mesenchymal stem cells derived from adipose tissue were isolated and cultured as described in Figure 32. -NAM -FGF4 = controls, + NAM -FGF4 = culture with 5 mM / ml NAM, + NAM + FGF4 = culture with 50 ng / ml FGF4 and 5 mM / ml NAM. Observe the synergistic effect of Nicotinamide and FGF4 together on the proliferation of total nucleated cells in the culture. [0104] [0104] Figure 34 is a bar graph illustrating the beneficial effect of culturing MSC's derived from adipose tissue in the presence of nicotinamide and FGF4 on the size of the cultured mesenchymal stem cells. Mesenchymal stem cells derived from adipose tissue were isolated and cultured as described in Figure [0105] [0105] Figure 35 is a bar graph, showing the effect of nicotinamide with and without FGF4 on the differentiation of ex-vivo expanded hematopoietic cells. Umbilical cord-derived premature hematopoietic progenitor cells (CD 2133+) were isolated using microspheres of CDl33 and CliniMACS (Milentyi, Inc), and were cultured for 3 weeks in MEMa supplemented with 50 ng / ml of newly acting cytokines and fetal bovine serum, + 2.5 or 5 mM nicotinamide (NAM), + 10, 50 or 200 ng / ml FGF4. After cultivation for three weeks, CD38-CD133 + cells were labeled and counted by FACS. Column 1 = Control: -NAM, -FGF4, Column 2 = + 2.5 mM NAM, Column 3 = + 5 mM NAM, Column 4 = + 10 ng / ml FGF4, Column 5 = +50 ng / ml FGF4, Column 6 = +200 ng / ml FGF4, Column 7 = +2.5 mM NAM, +10 ng / ml FGF4, Column 8 = +2.5 mM NAM, +50 ng / ml FGF4, Column 9 = +2.5 mM NAM, +200 ng / ml FGF4, Column 10 = +5 mM NAM, +10 ng / ml FGF4, Column 11 = +5 mM NAM, +50 ng / ml FGF4, Column 12 = +5 mM NAM, +200 ng / ml FGF4. [0106] [0106] Note the significantly larger fraction of non-differentiated premature progenitors (CD38- CD133 +) in cultures treated with nicotinamide (columns 2 and 3), in the absence of any significant effect of FGF4 only (columns 4 to 6), and in the absence of no significant effect of FGF4 on nicotinamide-mediated inhibition of hematopoietic progenitor cell differentiation (columns 7 to 12). [0107] [0107] Figure 36 is a bar graph, showing the effect of nicotinamide with and without FGF4 on differentiation of ex-vivo expanded hematopoietic cells. The hematopoietic progenitor cells (CD 133+) derived from umbilical cord were isolated and cultured, with and without nicotinamide and FGF4, as in Figure 35. After cultivation for three weeks, CD38 + cells were marked and counted by FACS. Columns 1 through 12 as well as in Figure 35. Note the significant inhibition of differentiation (CD38 + cells) in cultures treated with nicotinamide (columns 2 and 3), in the absence of any significant effect of FGF4 only on differentiation (columns 4 to 6 ), and in the absence of any significant effect of FGF4 on nicotinamide-mediated inhibition of hematopoietic progenitor cell differentiation (columns 7 to 12). [0108] [0108] Figure 37 is a bar graph showing the effect of nicotinamide, with and without FGF4, on the differentiation of the myeloid lineage from ex-vivo hematopoietic cells. The hematopoietic progenitor cells (CD 133+) derived from umbilical cord were isolated and cultured, with and without nicotinamide and FGF4, as in Figure 35. After cultivation for three weeks, the differentiated cells of myeloid lineage (CD33 +) were marked and counted by FACS. Columns 1 to 12 as well as in Figure 35. Note the significant inhibition of myeloid lineage differentiation (CD33 + cells) in cultures treated with nicotinamide (columns 2 and 3), in the moderate improvement of myeloid lineage differentiation by FGF4 only (columns 4 to 6), and in the absence of any significant effect of FGF4 on nicotinamide-mediated inhibition of myeloid hematopoietic cell differentiation (columns 7 to 12). [0109] [0109] Figure 38 is a bar graph showing the effect of nicotinamide, with and without FGF4, on the differentiation of the lymphoid lineage from ex-vivo hematopoietic cells. The hematopoietic progenitor cells (CD 133+) derived from umbilical cord were isolated and cultured, with and without nicotinamide and FGF4, as in Figure 35. After cultivation for three weeks, differentiated cells of lymphoid lineage (CD1I9 +) were marked and counted by FACS. Columns 1 through 12 as well as in Figure 35. Note the impressive inhibition of lymphoid lineage differentiation (—CD19 + cells) in cultures treated with nicotinamide (columns 2 and 3), in the absence of any significant effect on lymphoid lineage differentiation by FGF4 only (columns 4 to 6), and in the absence of any significant effect of FGF4 on nicotinamide-mediated inhibition of the differentiation of lymphoid hematopoietic cells (columns 7 to 12). DESCRIPTION OF THE SPECIFIC APPLICATIONS [0110] [0110] The present patent application, in some respective applications, refers to methods of expanding mesenchymal stem cells and cell populations generated by them. [0111] [0111] Before explaining at least one application of the present invention patent application in detail, it should be understood that the present invention patent application is not necessarily limited in its application by the details set out in the description below or exemplified by the Examples. The present invention patent application is capable of executing other applications or of being practiced or executed in several ways. [0112] [0112] The multipotent nature of mesenchymal stem cells (MSCs) make these cells an attractive therapeutic tool and a candidate for transplants, capable of fulfilling the role in a wide range of clinical applications in the context of both genetic and cellular therapy strategies. For example, mesenchymal stem cells can be used to enhance post-transplant hematopoietic graft, to aid tissue regeneration, to promote wound healing and to correct a multitude of other inherited and acquired disorders. Expansion protocols for efficient mesenchymal stem cells, which do not have deleterious effects on the differentiation potential and the potential for target tissue graft of cells, are crucial for the success of any of these strategies. [0113] [0113] In addition, MSCs are attractive for clinical therapy in regenerative medicine and in inflammatory conditions, due to their ability to differentiate, provide trophic support, and modulate the innate immune response. The therapeutic potential of MSC is tested in multiple clinical trials for indications such as bone and cartilage reconstitution, cardiac regeneration, critical limb ischemia, acute ischemic conditions, diabetes, Crohn's disease and graft versus host disease. [0114] [0114] MSCs are immunoprivileged and can be transplanted without the need to perform the combination of HLA (human leukocyte antigen | human leukocyte antigen) between the donor and the recipient and can therefore be manufactured on a large scale and marketed as a product of commercially available cells. The success of large-scale batch production from a donor is highly dependent on donor and serum selection, the potential of seeded cells for prolonged cultivation expansion and the duration of manufacture. Although MSC multiplies relatively easily in vitro, its proliferative potential is continuously decreased and its doubling interval increases during cultivation. As a result, successful manufacturing for marketing large batches of homogenous MSCs from a donor remains a challenge. [0115] [0115] While studying the effect of growth factors on MSC expansion, the present inventors found that growth factors such as basic FGF (bFGF), HB-EGF or platelet-derived growth factor (PDGF) have a non-reproducible effect or even negative when cultured in the presence of nicotinamide in the proliferation of mesenchymal stem cells (Figures 1, 2, 6). [0116] [0116] In sharp contrast, FGF4 has shown, surprisingly, a reproducible and synergistic activity along with nicotinamide in the expansion / proliferation of mesenchymal stem cells. [0117] [0117] As illustrated in Figures 3A to D, the present inventors demonstrated that nicotinamide potentiates the effect of FGF4 on the proliferation of mesenchymal stem cells. [0118] [0118] In addition, the present inventors demonstrated an unexpected effect of nicotinamide, on the size of cells, of mesenchymal stem cells cultured with FGF4. [0119] [0119] As shown in Figures 4A to B, MSC '"s generated through nicotinamide and FGF4 cultivation are smaller than mesenchymal stem cells cultured according to the same methods, but in the presence of only and similar FGF4 to MSC cultured with nicotinamide only. For example, between days 10 and 32, mesenchymal stem cells cultured in nicotinamide and FGF4 have a diameter less than approximately 20 µm, whereas cells cultured in the presence of FGF4, but in the absence of nicotinamide, have a diameter greater than 1. Thus, nicotinamide imposed an undifferentiated state in MSC grown with FGF4. [0120] [0120] While further reducing the present patent application to practice, the present inventors demonstrated that the percentage of cells expressing the MSC marker, CDI1IO0O5 + CD45-, is preserved in cultures treated with nicotinamide and FGF4 (Figures 5A to D). [0121] [0121] Furthermore, the present inventors found that the use of nicotinamide, during specific stages of the selection and expansion protocol, was advantageous to populations of mesenchymal stem cells. Thus, for example, the sowing of mesenchymal stem cells, in the presence of nicotinamide and high calcium concentrations, increased their seeding efficiency, as well as noticed when analyzing the marker phenotype of the cells (Figures 11 to 13). Mesenchymal stem cells could be successfully expanded by at least six passages, in the presence of nicotinamide, without inducing differentiation, as illustrated by the composition of the cell surface marker (Figures 12A to C). Still, it was demonstrated that nicotinamide promoted the expansion of a more homogeneous and less granular population of MSCs (Figures 20A to Ce 21h to B). [0122] [0122] The present inventors have demonstrated, experimentally, that MSC's grown with nicotinamide proliferated more quickly, and as a result, their doubling interval (see Figure 26) is reduced and the cultures reach confluence in a period of considerably shorter time (Figures 14 to 17, 26 and 27). The proliferative effect of nicotinamide has also been demonstrated in large cultures of MSC's (Figure 18). In addition, the effect was not restricted to selected batches of serum (Figure 19), a considerable advantage for the manufacture of large batches of MSC's. Furthermore, the present inventors demonstrated that the proliferative effects of nicotinamide, in combination with fibroblast growth factor 4 (FGF4), were not observed for stem cells of non-mesenchymal origin, such as progenitor cells or hematopoietic stem cells ( for example, CD133 +) (see Example 10, and Figures 35 to 38 shown here). In fact, only FGF4, or the same in combination with nicotinamide, did not have any effect on the proliferation or differentiation of progenitor cells or hematopoietic stem cells cultured ex vivo (see Figures 35 to 38, ranges 1 and 4 to 12 ). [0123] [0123] Thus, in accordance with an aspect of the present patent application, a method of culturing mesenchymal stem cells (MSC's) is provided comprising culturing a population of MSC's in a medium containing nicotinamide and factor 4 of fibroblast growth (FGF4). [0124] [0124] In addition, the present inventors have now discovered that cultivating a mixed population of mesenchymal stem cells, in the presence of nicotinamide, increases the mesenchymal stem cell phenotype, so that a subsequent pre-selection or selection with a marker of mesenchymal stem cells provides a more homogeneous population of mesenchymal stem cells, thus providing a method for obtaining enriched populations of subsets of mesenchymal stem cells. This was substantiated by the present inventors, when they demonstrated that the cultivation of MSC's in the presence of nicotinamide increases the expression of a specific adhesion molecule - Vascular cell adhesion protein 1 | VCAM1 / CD106; see Figure 24A) . Conversely, the present inventors demonstrated that culturing MSCs, in the presence of nicotinamide, decreased the expression of a marker for cell senescence (CD54; see Figure 24B), thereby providing a method for obtaining enriched populations of mesenchymal stem cells through the depletion of the cell population for non-relevant cells. [0125] [0125] The use of a selection or organization in the next step increases the strictness of the organization and selection specificity for MSCs, and, in addition, potentially reduces possible contamination from the starting material. [0126] [0126] Thus, in accordance with one aspect of the present patent application, a method of isolating mesenchymal stem cells (MSCs) from a mixed population of cells is provided, comprising: [0127] [0127] The term “mesenchymal stem cell” or “MSC” is used interchangeably for adult cells that are not terminally differentiated, which can divide to yield cells that are either stem cells, or cells that irreversibly differentiate into give rise to others, from a lineage of mesenchymal cells, for example, adipose, bony, cartilaginous, elastic and fibrous, myoblast connective tissues) as well as tissues other than those originating in the embryonic mesoderm (for example, neural cells) various influences of bioactive factors, such as cytokines. [0128] [0128] The MSC cultures used by some applications of the present patent application preferably include three groups of cells that are defined by their morphological characteristics: small and agranular cells (referred to here as RS-1), small cells and granular (referred to below as RS-2) and large, moderately granular cells (referred to below as mature MSCs). The presence and concentration of such cells in the culture can be analyzed by identifying the presence or absence of several cell surface markers, using, for example, immunofluorescence, in situ hybridization, and activity assays. [0129] [0129] When MSCs are grown under the conditions of cultivation of some applications of the present patent application, they exhibit negative marking for the hematopoietic stem cell markers CD34, CDI1B, CD43 and CD45. A small fraction of cells (less than 10%) may be weakly positive for CD31 and / or CD38 markers. In addition, mature MSC's may be weakly positive for the hematopoietic stem cell marker, CD117 (c-Kit), moderately positive for the MSC's osteogenic marker, Stro-l [Simmons, PJ & Torok-Storb, B. ( 1991). Blood 78, 5562] and positive for the peripheral T thymocyte and lymphocyte marker, CD90 (Thy-l1). On the other hand, RS-1 cells are negative for CD117 and Strol markers and are weakly positive for CD90, and RS-2 cells are negative for all of these markers. [0130] [0130] Mesenchymal cells cultured under the conditions of cultivation of some applications of the present patent application can secrete biologically active factors in the medium. The present inventors have observed that the medium collected from the mesenchymal cells cultured with nicotinamide comprises high levels of growth factors and cytokines (for example, [0131] [0131] According to a preferred application of that aspect of the present patent application, mesenchymal stem cells are human. [0132] [0132] According to another application of that aspect of the present patent application, mesenchymal stem cells are isolated from newborn human beings. [0133] [0133] Mesenchymal stem cells can be isolated from a variety of tissues, including, but not limited to, bone marrow, peripheral blood, blood, placenta (eg, fetal side of the placenta), umbilical cord blood, umbilical cord, fluid amniotic, and adipose tissue of the placenta. [0134] [0134] A method of isolating “mesenchymal stem cells from peripheral blood is described by Kassis et al [Bone Marrow Transplant. May 2006; 37 (10): 967 to 976). A method of isolating mesenchymal stem cells from placental tissue is described by Zhang et al [Chinese Medical Journal, 2004, 117 (6): 882 to 887]. Methods for isolating and culturing adipose tissue, blood from the placenta and umbilical cord of mesenchymal stem cells is described by Kern et al [Stem Cells, 2006; 24: 1294 to 1301]. [0135] [0135] The bone marrow can be isolated from an individual's iliac crest by aspiration. BM low-density mononuclear cells (BMMNC | bone marrow mononuclear cells) can be separated by a FICOLL-PAQUE density gradient or by eliminating red blood cells using Hetastarch (hydroxyethyl starch). Preferably, mesenchymal stem cell cultures are generated by diluting BM aspirates (usually 20 ml) with equivalent volumes of Hank's balanced salt solution (HBSS | Hank's balanced salt solution; GIBCO Laboratories, Grand Island, NY, USA ), and stratification of cells diluted on approximately ml of a Ficoll column (Ficoll-Paque; Pharmacia, Piscataway, NJ, USA). After 30 minutes of centrifugation at a speed of 2,500 x g, the mononuclear cell layer is removed from the interface and suspended in HBSS. The cells are then centrifuged at a speed of 1,500 x g for 15 minutes, and suspended again in a complete medium (MEM, in a medium without deoxyribonucleotides or ribonucleotides; GIBCO); 20% fetal calf serum (FCS | fetal calf serum) derived from a batch selected for the rapid growth of MSC's (Atlanta Biologicals, Norcross, GA); 100 units / ml penicillin (GIBCO), 100 ug / ml streptomycin (GIBCO); and 2 mM L-glutamine (GIBCO). [0136] [0136] MSCs derived from adipose tissue can be obtained from any tissue containing fat, for example, from epididymal fat or through liposuction, and mononuclear cells can be manually isolated by removing fat and fat cells, or using the Celution System (Cytori Therapeutics), following the same procedure described above for the preparation of MSC's. [0137] [0137] As mentioned, the method is carried out by culturing (for example, ex-vivo or in vitro) the mesenchymal stem cells in a medium comprising nicotinamide and FGF4. [0138] [0138] In accordance with this aspect of the present patent application, cells are grown under conditions that do not induce differentiation (for example, in the absence of differentiating factors or in the presence of a non-differentiating quantity of differentiating factors) . [0139] [0139] The present invention patent application contemplates the cultivation of mesenchymal stem cells directly after isolation of their source, or the cultivation of populations of cells that have been pre-selected for mesenchymal stem cells. Thus, the present invention patent application contemplates the cultivation of both heterogeneous populations of cells that comprise the MSCs, and of the more homogeneous cell populations, which have been enriched for the MSC's, characterized by more than 70%, more than 80% , more than 90% or more than 95%, more than 98% of them are MSCs. In addition, enrichment for MSC's is contemplated concurrently with cultivation, as also described here below. [0140] [0140] It will be appreciated that the composition of the heterogeneous cell population is dependent on the cell source. So, for example, if the placenta is selected as the cell source, the heterogeneous population of cells will comprise the cells of the placenta, as well as the mesenchymal stem cells. If the bone marrow is selected as the cell source, the heterogeneous population of cells will comprise the blood cells. However, as shown in Example 10, according to some applications of the present patent application, the cultivation of mesenchymal stem cells under the conditions of cultivation of some applications of the present patent application (for example, nicotinamide and FGF4 in combination) results in the selective expansion of mesenchymal stem cell populations, although it does not have a proliferative effect concomitant with mesenchymal stem cell populations. [0141] [0141] According to one method, the cell population is grown (in vitro or ex-vivo) on polystyrene plastic surfaces (for example, in a flask), so that they enrich for mesenchymal stem cells by removing non-adherent cells (for example, non-mesenchymal stem cells). This enrichment method for MSC's can be carried out before cultivation in nicotinamide and FGF4, concomitant with cultivation in nicotinamide and FGF4 and / or after cultivation in nicotinamide and FGF4. [0142] [0142] Other selection methods for MSCs are known in the art, including, for example, positive selection against mesenchymal stem cell markers and / or negative selection against progenitor cell and hematopoietic stem cells, such as CD34, CD133 , CD8, etc. Methods for determining cell surface expression of proteins are well known in the art. examples include “immunological methods, such as FACS analyzes, as well as biochemical methods (cell surface labeling, for example, radioactive, by fluorescence, by avidin-biotin). [0143] [0143] It will be appreciated that a selection step can also be performed after cultivation in nicotinamide and FGF4. This can be done either as a pre-selection step, or instead of a pre-selection step [0144] [0144] As used herein, “nicotinamide” refers to nicotinamide, as well as products that are derived from nicotinamide, its nicotinamide analogues and metabolites or nicotinamide analogues, such as, for example, NAD, NADH and NADPH. [0145] [0145] As used in this document, the phrase “nicotinamide analog” refers to any molecule that is known to act in a similar way to nicotinamide. Representative examples of nicotinamide analogs include, without limitation, benzamide, nicotine thioamide (the nicotinamide thiol analogue), nicotinic acid, α-amino-3-indolepropionic acid, and inhibitors of the sirtuin family of histone / protein deacetylases. [0146] [0146] Examples of derivatives of nicotinamide analogues include, but are not limited to, substituted benzamides, substituted nicotinamides and nicotine thioamides and nicotinamides and nicotine thioamides substituted by N. [0147] [0147] In a specific application, nicotinamide is supplied at a concentration of at least approximately 1 mM to 20 mM. In another application, the concentration of nicotinamide is provided at a concentration of at least approximately 1 mM to 10 mM, for example, approximately 2.5 mM, approximately 5 mM, approximately 7.5 mM. [0148] [0148] Fibroblast growth factor 4, the product of the FGF4 gene (location on the map: l1g913.3), FGF-4 / HBGF-4 / KFGF, is a long 176 176 protein, derived through N cleavage -terminal 30 AAs of precursor protein. FGF-4 contains a single N-linked glycosylation site. Non-glycosylated FGF-4 is cleaved into two terminally truncated peptides in NH2 (13 and 15 kDa), which are more active with greater heparin affinity than wild type. [0149] [0149] According to a specific application, FGF4 is a human FGF4. [0150] [0150] FGF4 recombinant protein is commercially available (for example, with Sigma Aldrich, where it is produced in baculovirus and cleaved at the N-terminal to yield a 148 AA protein; or with Invitrogen, where it is produced in E.coli ). [0151] [0151] In a specific application, FGF4 is supplied to the culture at a concentration of at least approximately 1 to 1000 ng / ml. In another application, the concentration of FGF4 is provided at a concentration of at least approximately 10 to 200 ng / ml, at 100 ng / ml, for example, approximately 50 ng / ml. [0152] [0152] According to a specific application, the culture medium comprising both nicotinamide and FGF4 is devoid of additional growth factors, such as PDGF, HB-EGF or bFGF (FGF2). [0153] [0153] It will be appreciated that when referring to a medium without a specific component, the present application for a patent contemplates that the medium comprises that component, but in a concentration below its minimum activity. Thus, for example, some specific media may comprise small amounts of the growth factors described above, however, the methods of the present patent application refer to a medium being devoid of exogenously aggregated growth factors, in addition to what is included in a commercial medium formula, or resulting from general adjustments to the concentrations of medium components. Thus, according to a specific application, the medium comprising nicotinamide and FGF4 can comprise any of the additional growth factors mentioned above, but at a concentration of less than 1 ng / ml. [0154] [0154] A typical cell medium to which nicotinamide and FGF4 can be added is Dulbecco modified MEM (DMEM). Alternatively, the cell medium may be Ham's F12. Other contemplated media include HEM RPMI, F-12, and the like. [0155] [0155] It will be noted that many of the culture media contain nicotinamide as a vitamin supplement, for example, MEMa (8.19 µM nicotinamide), RPMI (8.19 µM nicotinamide), DMEM (32.78 µM nicotinamide) and Glascow's medium (16.39 µM nicotinamide), however, the methods of the present patent application refer to exogenously aggregated nicotinamide, supplementing any nicotinamide and / or nicotinamide portion included in the medium formula, or resulting "from the general adjustments of the concentrations of components of the medium. [0156] [0156] In an application of the present patent application, the cell culture medium has a high calcium concentration greater than approximately 2 mM, or greater than approximately 5 mM. It will be appreciated that the calcium concentration is calculated as the total calcium concentration, including the concentration already present in the culture medium. [0157] [0157] So, for example, if the medium is Dulbecco-modified MEM (DMEM) (which already has a calcium ion concentration of approximately 1.8 mM), no additional calcium needs to be added. If the cell medium is Fl12 of Ham, which has a calcium ion concentration of approximately 0.9 mM, more calcium must be added so that the total calcium concentration is above 1.8 mM. In one application, the source of the additional calcium may be serum. [0158] [0158] During cultivation, the medium may contain supplements required for cellular metabolism, such as glutamine and other useful amino acids, vitamins, minerals and proteins, such as transferrin and the like. The medium may also contain antibiotics to prevent contamination with yeasts, bacteria and fungi, [0159] [0159] Normoxia or hypoxia conditions are also contemplated. [0160] [0160] According to an application, the culture medium is devoid of serum (for example, serum-free medium) and comprises serum substitutes, including, but not limited to, platelet lysate (during sowing and / or expansion ). [0161] [0161] According to another application, the medium comprises approximately 10% fetal bovine serum. Human serum is also contemplated. [0162] [0162] Cultivation, in accordance with this aspect of the present patent application, can be carried out for a limited amount of time, so that no expansion takes place (for example, only during the sowing stage) or can be carried out for longer periods of time, in order to allow the expansion of mesenchymal stem cells (for example, cell propagation), thus obtaining respective increased amounts. [0163] [0163] For each round of propagation, adherent cells can be cultured using trypsin / EDTA or by scraping cells and dissociated by passing through a narrow, plastic Pasteur pipette, and preferable and laminated again at a density of approximately 100 to approximately 10,000 cells / cmº. [0164] [0164] In accordance with this aspect of the present patent application, a sufficient period of time for cell expansion can be taken to denote the length of time required for at least one cell to be divided. [0165] [0165] According to an application, cultivation is carried out for at least one day, at least two days, at least three days, at least four days, at least five days, at least six days, at least one week , at least two weeks, at least three weeks, at least four weeks, or at least five weeks. [0166] [0166] According to another application, cultivation should not be carried out for more than ten weeks. [0167] [0167] According to yet another application, cells are made possible to expand by at least two population duplications, at least four population duplications, at least six population duplications, at least eight population duplications, at least ten population doubling, at least 15 population doubling, at least 20 population doubling, at least 25 population doubling, at least 30 population doubling, at least 35 population doubling, at least 40 population doubling, or at least 45 population duplications. [0168] [0168] According to another application, cells are not allowed to expand by more than 50 population duplications. [0169] [0169] The present patent application contemplates the additional methods of expanding mesenchymal stem cells, as well as (or instead of) cultivation in nicotinamide and FGF4. [0170] [0170] Since the present inventors have found that when, at least, a portion of the expansion process time is carried out in the presence of nicotinamide, increasing numbers of mesenchymal stem cells are obtained, additional methods of expansion preferably include the cultivation in the presence of nicotinamide. [0171] [0171] Thus, in accordance with another aspect of the present patent application, a method of expanding a population of mesenchymal stem cells is provided, the method comprising culturing a population sown of mesenchymal stem cells for a period of sufficient time for cell expansion, characterized in that, for at least a portion of the time period, the culture is carried out in a medium devoid of nicotinamide; and for at least a second portion of the time period, the culture is carried out in a medium comprising - “nicotinamide and FGF4, thus generating an expanded population of mesenchymal stem cells. [0172] [0172] The term "expand", as used in this document, refers to the increase in the number of cells in the cell population due to replication. [0173] [0173] In accordance with this aspect of the present patent application, cells are expanded under conditions that do not induce differentiation (for example, in the absence of differentiating factors). [0174] [0174] The population sown of undifferentiated mesenchymal stem cells may be a heterogeneous population of cells or a purified population of mesenchymal stem cells, as described above. [0175] [0175] As mentioned, a medium being devoid of nicotinamide refers to a medium comprising less than the minimum effective amount of nicotinamide (e.g. less than 0.5 mM, or, more preferably, less than 0.05 mM) . Thus, means comprising small amounts of nicotinamide (as described hereinabove) can be used for that aspect of the present application. Thus, according to a specific application, the medium without exogenously added nicotinamide may comprise, before adding exogenous nicotinamide as a supplement, [0176] [0176] According to an application, MSC's are at least 50% purified, at least 75% purified or at least 90% purified. [0177] [0177] The population of mesenchymal stem cells can be seeded (and also cultivated) in any medium, including those described above or those disclosed in US Patent Application No. 20050260748, incorporated herein by reference. [0178] [0178] The cultivation time ratio in the presence of nicotinamide and FGF4: cultivation in the absence of nicotinamide can vary and include all ratios, from 1:99; 2:98; 3:97; 4:96, 5:95; 6:94; 7:93; 8:92; 9:91; 10:90; 11:89; 12:88; 13:87; 14:86; 15:85; 16:84; 17:83; 18:82; 19:81; 20:80; 21:79; 22:78; 23:77; 24:76; 25:75; 26:74 27:73; 26:72; 29:71; 30:70; 31:69; 32:68; 33:67; 34:66; 35:65; 36:64; 37:63; 38:62; 39:61; 40:60; 41:59; 42:58; 43:57; 44:56; 45:55; 46:54; 47:53; 48:52; 49:51; 50:50; 51:49; 52:48; 53:47; 54:46; 55:45; 56:44; 57:43; 58:42; 59:41; 60:40; 61:39; 62:38; 63:37; 64:36; 65:35; 66:34; 67:33; 68:32; 69:31; 70:30; 71:29; 72:28; 73:27; 74:26; 75:25; 76:24; 77:23; 78:22; 79:21; 80:29; 81:19; 82:18; 83:17; 84:16; 85:15; 86:14; 87:13; 88:12; 89:11; 90:10; 91: 9; 92: 8; 93: 7; 94: 6; 95: 5; 96: 4; 97: 3; 98: 2; 99: 1. [0179] [0179] According to an application, at least one complete round of propagation is carried out in the presence of nicotinamide. [0180] [0180] It will be appreciated that cultivation in the medium comprising nicotinamide is carried out before or after cultivation in the medium devoid of nicotinamide. [0181] [0181] According to applications of the present patent application, the nicotinamide-free medium comprises FGF4 (or in the same, or in a different concentration as the nicotinamide-containing medium). [0182] [0182] According to other applications of the present patent application, the medium devoid of nicotinamide is also devoid of FGF4. [0183] [0183] Still, the present inventors contemplate more than one cultivation stage in the presence of nicotinamide and FGF4, interspersed with cultivation stages in the absence of nicotinamide, and vice versa. [0184] [0184] According to one application, cultivation in the presence of nicotinamide and FGF4 is carried out for at least one day, at least two days, at least three days, at least four days, at least five days, at least six days, at least one week, at least two weeks, at least three weeks, at least four weeks or at least five weeks. [0185] [0185] According to another application, cultivation in the absence of nicotinamide is carried out for at least one day, at least two days, at least three days, at least four days, at least five days, at least six days, at least one week, at least two weeks, at least three weeks, at least four weeks or at least five weeks. [0186] [0186] As mentioned, the second stage of the purification process is the selection for MSC's based on the expression of a stem cell surface marker - mesenchymal. The selection or organization step can comprise the selection of mesenchymal stem cells (MSCs) from the mixed population of cells, through one or more of such surface markers. The use of a selection or organization in the next step increases the rigor of the organization and selection specificity for MSCs, and, in addition, potentially reduces the risk of possible contamination from the starting material. [0187] [0187] Prior to organization, merged cell populations are typically dispersed, using cell dispersing agents. Preferably, single cell populations are obtained. Examples of agents that can be used to disperse cells include, but are not limited to, collagenase, dispase, Accutase6, trypsin (e.g., trypsin-EDTA), and papain. Alternatively or additionally, crushing can also be performed to increase cell dispersion. [0188] [0188] According to a specific application, the selection is made by the selection cells, which express VCAM-1 / CD106 (NP 001069.1) above a predetermined level. [0189] [0189] According to another application, the selection is made by the selection cells, which express at least one of CD1I05 (SH2), CD73 (SH3 / 4), CD44, CD90 (Thy-1), CD71, STRO -1, CD29, CDl66, CD1146, CD106 and CD271 above a predetermined level. [0190] [0190] According to a specific application, the surface marker is the stromal precursor antigen -1 (STRO-1), CD1I05 or VCAM (CD106). [0191] [0191] According to another application, the selection is made by the selection cells that express, at least, one of CD34, CD11B, CD43 and CD45 below a predetermined level. [0192] [0192] A variety of methods are known for selection or organization based on the expression of antigens, and any of these can be used in the selection or organization step described here. In particularly preferred applications, the analysis is performed using a flow cytometer, and the cells are subsequently organized based on the specific characteristics of each cell's scattered and fluorescent light. Thus, the selection or organization can be carried out through the organization of fluorescence activated cells (FACS | fluorescence activated cell sorting). [0193] [0193] As well as known in the art, FACS involves the exposure of cells to a reporter, such as an antibody fluorescently labeled, which binds and labels the antigens expressed by the cell. Methods of producing antibodies and labeling them to form rapporteurs are known in the art, and described, for example, in Harlow and Lane. The antibodies that can be used for FACS analysis are instructed in Schlossman S, Boumell L. et al, [Leucocyte Typing V. New York: Oxford University Press; 1995] and are widely available on the market. The cells are then passed through a FACS machine, which organizes the cells among themselves based on the labeling. [0194] [0194] Alternatively or additionally, magnetic cell organization (MACS) or immunopositioning can be used to organize cells. [0195] [0195] As mentioned above, the merged cell populations are analyzed by a Flow Cytometer, such as a laser scanning Cytometer. A Flow Cytometer typically consists of a laser light source, a flow measurement chamber, and an optical system consisting of lenses, filters, and light detectors. Two photomultiplier tubes (light detectors), one at 180 degrees and another at 90 degrees from the laser, are used to measure the dispersion of the right (SSC) and frontal (FSC) angles, respectively. Three fluorescence detectors, each having a filter and a photomultiplier tube, are used to detect fluorescence. The three detectors identify green (FL1--530 nm), orange (FL2--585 nm), and red (FL3--650 nm) fluorescence. The cells are identified by the organizational logic applied to all five detector signals (FSC, SSC, FL1, FL2, FL3) using a computer. [0196] [0196] The exemplary Flow Cytometers that can be used in this aspect of the present patent application are manufactured by companies such as Becton Dickinson (USA), Backman Coulter (USA), Partec (Germany). [0197] [0197] The FACS machine can be configured so that the cells of a specific frontal and / or lateral dispersion are selected. The light scattered ahead (FSC) is proportional to the area or size of the cell surface. FSC is a measurement of mostly diffracted light, and is detected exactly off the axis of the incident laser beam in the frontal direction by a photodiode. FSC provides a suitable method of detecting particles larger than a certain size, regardless of their fluorescence. [0198] [0198] Light scattered to the side (SSC) is proportional to the granularity of the cell or its internal complexity. SSC is a measurement of mostly refracted and reflected light, which occurs at any interface within the cell where there is a change in the refractive index. The SSC is collected at approximately 90 degrees from the laser beam by a collecting lens, and is then redirected by a beam separator to the appropriate detector. [0199] [0199] Thus, for example, the present patent application contemplates the selection of cells that have a diameter approximately less than one, by switching in a specific frontal dispersion, and a specific granularity by switching in a specific lateral dispersion . [0200] [0200] the present application for a patent contemplates the selection of specific cell populations based on the level of expression on the cell surface. Thus, in the case of FACS, the machine must be configured so that cell populations switched for events are marked with a fluorescent intensity of approximately between 20 and 100 (dark), approximately between 100 and 500 (moderate) or approximately between 500 and 2000 or higher (glossy). The following cell populations are covered by the present application for a patent: VCAN1 shiny cells; Moderate VCAMI1 cells; Dark cells VCAM1; STRO-1 shiny cells; Moderate STRO-1 cells; Dark cells STRO-1; CD105 shiny cells; Moderate CD105 cells; [0201] [0201] It will be appreciated that cell populations can be selected based on the expression of more than one of the above mentioned markers - for example, at least two of the above mentioned markers or at least three of the above mentioned markers. [0202] [0202] The cell populations described above are normally enriched for cells that do not express CD45. Thus, according to another application, less than 10% of the cells, in the cell populations described above, express CD45, as measured by FACS. [0203] [0203] According to another application, more than 90% of the cells, in the cell populations described above, express CD90, as well as measured by FACS. [0204] [0204] According to another application, more than 95% of the cells, in the cell populations described above, express CD90, as well as measured by FACS. [0205] [0205] According to another application, more than 90% of the cells, in the cell populations described above, express CD44, as well as measured by FACS. [0206] [0206] According to another application, more than 95% of the cells, in the cell populations described above, express CD44, as well as measured by FACS. [0207] [0207] As mentioned, additional steps are contemplated by the present inventors before, during or after the two-step protocol described in this document. Such additional steps may involve cultivation on a plastic surface, as described here above, and / or additional expansion steps, for example, as well as the nicotinamide cultivation described here above. [0208] [0208] In some applications, cells are selected according to size, for example, through a cell counter based on the exclusion of Trypan Blue and on graphical analysis. Suitable cell counters include, but are not limited to, Cedex counters (Roche Innovatis). The number of cells that can be cultured, according to any of the methods of the present application, can be any number, including small batches - for example, 100 x 10 ° cells to larger batches - for example, 100 x 10 ”or 100 x 10”. [0209] [0209] When larger batches are required, cells are normally grown in a bioreactor (or in multilevel industrial flasks), the size of which is selected corresponds to the number of cells being grown. [0210] [0210] Examples of flasks and plates that can be used to grow MSC's in commercial quantities include, for example, the [0211] [0211] As used in this document, the term “bioreactor” refers to any device in which biological and / or biochemical processes develop, under monitored and controlled operating and environmental conditions, for example, PH, temperature, pressure, supply nutrients and waste removal. According to an application of the present patent application, the basic classes of bioreactors suitable for use with the present patent application include static bioreactors, agitated flask bioreactors, rotating wall bioreactors, hollow fiber bioreactors and bioreactors direct perfusion, as also described in WO 2005/007799, the content of which is incorporated by reference. [0212] [0212] The cultured population of cells generated using the methods described here can be further treated after cultivation or stored (for example, cryopreserved) in the presence of a cryopreservant. Such cryopreservatives include dimethyl sulfoxide (DMSO), glycerol, and the like. [0213] [0213] The cell populations generated after the cultivation and / or expansion method of the present patent application can be used for a variety of purposes, including in the search for detection agents that affect the respective differentiation, and for use therapeutic. Alternatively or additionally, cell populations can be stored (for example, frozen) until required. [0214] [0214] According to one application, populations of mesenchymal stem cells generated using the methods disclosed here can be used for other differentiation protocols. [0215] [0215] Methods for differentiating mesenchymal stem cells from different cell lines are known in the art. [0216] [0216] Differentiation cells can be obtained by culturing or differentiating the MSC, in a culture environment that enriches for cells with the desired phenotype, for example, osteoblasts, adipocytes, etc. Cultivation can comprise agents that enhance differentiation to a specific strain. [0217] [0217] Osteogenic differentiation can be performed by plating cells and culturing at confluence, followed by cultivation in a medium comprising glycerol-beta phosphate, ascorbic acid and retinoic acid (see Cowan et al. (2005) Tissue engineering 11 , 645 to 658). [0218] [0218] To induce adipogenic differentiation, separate cells must be seeded - again in 24-well plates (7 x 10'cells / ml) and treated in adipogenic medium for three weeks. Two exemplary adipogenic media are provided: DMEM supplemented with 0.05 mg / ml gentamicin, 2 mM L-glutamine, 10% FBS, 0.5 µM 3-isobutyl-1-methylxanthine (IBMX, Sigma), 0 , 5 µM hydrocortisone (Sigma) and 60 µM indomethacin (Sigma), or adipogenic MSC stimulating supplements purchased from StemCell Technologies, as instructed by the manufacturer. Adipogenic differentiation can be assessed by red 6-oil marking: the cells are fixed with methanol at -20 ºC for 10 minutes, and treated with 60% isopropanol for 3 minutes. The plates can be marked in red O oil (Sigma) for 10 minutes, and rinsed under running water. After the rinse, the plates can be contrasted with Mayer hematoxylin (Sigma) for 1 minute, and rinsed under running water. [0219] [0219] Myocyte differentiation can be performed by plating cells and culturing at confluence, followed by culture in a medium comprising equine serum, dexamethasone and hydrocortisone (see Eun et al. (2004) Stem Cells 22: 617 to 624 ); or 5-azacitidine (see Fukuda et al. (2001) Artificial Organs 25: 187). [0220] [0220] Chondrocyte differentiation can be performed by plating cells and culturing in confluence, followed by culture in a medium comprising dexamethasone, ascorbic acid 2- [0221] [0221] Neuronal differentiation is known in the art. For example, the generation of neurons and / or oligodendrocytes from mesenchymal stem cells should be performed as described in United States Patent Nos. 20100021434 and 20090257987. [0222] [0222] Alternatively or additionally, mesenchymal stem cells can be genetically modified to express an agent (for example, a polypeptide, a siRNA or a miRNA) that is useful in the treatment of a disease, or that alternatively conducts its differentiation towards a specific lineage. [0223] [0223] Thus, for example, mesenchymal stem cells can be genetically modified to express bone morphogenic factor 2 (BMP2) in order to promote differentiation in bone. [0224] [0224] Alternatively, mesenchymal stem cells can be genetically modified to express Pd-x, in order to promote differentiation in pancreatic cells. [0225] [0225] Since mesenchymal stem cells are known to lodge and migrate towards wounds, cells can be used as carriers, transporting useful molecules to the wound site. Useful molecules can be molecules that are found inherently within stem cells - "mesenchymal (for example, growth factors), or they can be artificially positioned within cells (for example, proteins or polynucleotides transfected within cells). [0226] [0226] Both the differential and undifferentiated mesenchymal stem cell populations described in this document can be used to treat a multitude of disorders, the specific disorders being selected according to the state of differentiation of the cells. [0227] [0227] Thus, according to another aspect of the present patent application, a method of treating a disease or disorder is provided, the method comprising transplanting into a subject in need thereof, a therapeutically effective amount of the isolated cell population of the present invention patent application. [0228] [0228] According to an application, the disease or disorder is selected from the group consisting of bone or cartilage disease, neurodegenerative disease, heart disease, liver disease, cancer, nerve damage, wound healing, autoimmune diseases, graft versus host disease, spinal cord injury and tissue regeneration. [0229] [0229] Bone defects suitable for treatment using the cells of the present application include, but are not limited to, osteogenesis imperfecta, fracture, congenital bone defects, and the like. [0230] [0230] Furthermore, the mesenchymal stem cells of the present patent application can be implanted in an individual to provide support for connective and bone tissues of orthopedic and other prosthetic devices (for example, dental), such as prostheses and / or dental implants. [0231] [0231] The mesenchymal stem cells of the present patent application can be used to treat diseases of the central nervous system [CNS | central nervous system]. [0232] [0232] Representative examples of CNS diseases and disorders that can be beneficially treated with the cells described here include, but are not limited to, a pain disorder, a motor disorder, a dissociative disorder, a mood disorder, an affective disorder , a neurodegenerative disease or disorder and a seizure disorder. [0233] [0233] More specific examples of such conditions include, but are not limited to, Parkinson's disease, amyotrophic lateral sclerosis (ALS | ALS [amyotrophic lateral sclerosis]), multiple sclerosis, Huntington's disease, autoimmune encephalomyelitis, diabetic neuropathy, neuropathy glaucomatous, macula degeneration, [0234] [0234] As mentioned, since MSCs can differentiate into cartilage, the mesenchymal stem cells of the present patent application may be suitable for the treatment of joint conditions, including, but not limited to, osteoarthritis, rheumatoid arthritis , inflammatory arthritis, chondromalacia, avascular necrosis, traumatic arthritis and the like. [0235] [0235] Bone marrow-derived mesenchymal stem cells (MSC's) are known to interact with hematopoietic stem cells (HSC's) and immune cells, and represent a potential cell therapy to enhance allogeneic hematopoietic graft and prevent graft disease versus host (GVHD). When hematopoietic stem cell numbers were limited, human graft in NOD-SCID mice was observed only after co-infusion of unrelated human MSC's, but not with co-infusion of the mouse mesenchymal cell line. Unrelated human MSCs did not induce T cell activation in vitro and suppressed T cell activation through tuberculin and unrelated allogeneic lymphocytes in a dose-dependent manner. Cell-free MSCs grow stromal cells and supernatants from mice, and human dermal fibroblasts did not induce this effect. [0236] [0236] It is known that, when MSC's are introduced into the infarcted heart, they can prevent deleterious remodeling and improve recovery. MSCs were injected directly into the infarction, or were administered intravenously and appear to lodge at the wound site. Examination of the interaction of allogeneic MSCs with immune cells indicates low rejection by TT cells. The persistence of halogenic MSCs in vivo suggests their potential “commercially available” therapeutic use for multiple recipients (Pittenger MF, et al Circ Res. 2004 Jul 9; 95 (1): 9-20). [0237] [0237] The use of ex-vivo expanded mesenchymal cells for transplantation has the following advantages: [0238] [0238] Reduces the volume of blood or other tissue required for the reconstitution of a recipient system of adult tissues. [0239] [0239] Enables the storage of a small number of unexpanded mesenchymal cells, for example, the formation of umbilical cord blood or peripheral blood, for potential future use. [0240] [0240] In the case of an autologous transplant of recipients with malignancies, the contaminating tumor cells in the autologous infusion generally contribute to the recurrence of the disease. The selection and expansion of mesenchymal cells will reduce the burden of tumor cells in the final transplant. [0241] [0241] Tissue regeneration: The mesenchymal cell populations of the present patent application can be used to promote tissue regeneration. The transplantation of mesenchymal stem cells holds great promise of benefits in regenerative medicine, autoimmune diseases, inflammatory conditions, reconstructive surgery for ischemic, acute and chronic conditions, tissue engineering, regeneration of new tissues and naturally healing sick or injured organs. [0242] [0242] Gene therapy: For successful gene therapy, in the long run, a high frequency of genetically modified cells with transgenes, integrated stably within their genome, is a mandatory requirement. Currently, gene transfer in pure and / or progenitor stem cells is highly inefficient. The ability to store and process a selected population of mesenchymal cells ex vivo and the enhancement of their graft and housing potential could provide an increased likelihood of successful use of genetically modified cell transplantation [Palmiter Proc Natl Acad Sci USA 91 ( 4): 1219 to 1223, (1994)]. [0243] [0243] In one of the methods described in this document, cells can be obtained from an autologous, semi-autologous or non-autologous donor, embryo or umbilical cord / human placenta (for example, allogeneic or xenogenic). For example, cells can be isolated from a human cadaver or a donor individual. [0244] [0244] The term semi-autologous refers to cell donors that are partially unmatched with recipient cells in a major histocompatibility complex (MHC | major histocompatibility complex) located in class I or II. [0245] [0245] The cells of the present application can be administered to the treated individual using a variety of transplantation approaches, the nature of which depends on the implant site. [0246] [0246] According to one application, cells are not transplanted into the body in a medium comprising nicotinamide. [0247] [0247] Cells can be transplanted into an injured or healthy region of tissue. In some cases, the exact location of the injured tissue area may be unknown, and the cells may be inadvertently transplanted into a healthy region. In other cases, it may be preferable to administer the cells in a healthy region, thereby avoiding any further injury to that region. Whatever the case, after the transplant, the cells preferably migrate to the injured area. [0248] [0248] The terms or phrases "transplant", "cell replacement" or "graft" are used interchangeably here and refer to the introduction of the cells of the present patent application in the target tissue. As mentioned, the cells can be derived from the recipient or from an allogeneic, semi-allogeneic or xenogenic donor. Other xeno-origins are also contemplated. [0249] [0249] The cells of the present patent application can be transplanted through direct injection into an organ, injection into the bloodstream, intraperitoneal injection, injection directly into lymphoid organs, etc. Proper transplantation methods can be determined by monitoring the housing and graft of cells implanted in the desired organ, the expression of genes or specific markers to Organs desired organs, and the function of the individual's derived organ. In the pancreas, for example, maintenance of euglycemia, insulin secretion and / or C-peptide, may be a measure of restoration of function in a diabetic host animal after cell replacement therapy, as disclosed below. In the liver, for example, the synthesis of albumin can be monitored. [0250] [0250] MSC's normally down-regulate MHC class 2 and are therefore less immunogenic. Embryonic or newborn cells obtained from umbilical cord blood, umbilical cord Wharton jelly, or placenta are even less likely to be strongly immunogenic and therefore less likely to be rejected, since such cells, especially, they are immunosuppressive and immunoregulatory to start. [0251] [0251] However, since non-autologous cells can induce an immune reaction when administered to the body, several approaches can be taken to reduce the likelihood of rejection of non-autologous cells. This includes the administration of cells in privileged locations, or alternatively, suppression of the recipient's immune system, providing - “an anti-inflammatory treatment that can be indicated to control autoimmune disorders to initiate and / or encapsulate non-autologous cells / semi-autologous immunoisolation, and semipermeable membranes before transplantation. Encapsulation techniques are generally classified as microencapsulation, involving small spherical vehicles, and macroencapsulation, involving larger membranes of hollow fiber and flat plate (Uludag, H. et al. Technology of mammalian cell encapsulation. Adv Drug Deliv Rev. 2000; 42: 29 to 64. [0252] [0252] Methods of preparing microcapsules are known in the art and include, for example, those disclosed by Lu M 2Z, et al., Cell encapsulation with alginate and alpha-phenoxycinnamylidene-acetylated poly (allylamine). Biotechnol Bioeng. 2000, 70: '479 to 483, Chang T M and Prakash S. Procedures for microencapsulation of enzymes, cells and genetically engineered microorganisms. Mol. Biotechnol. 2001, 17: 249 to 260, and Lu M Z, et al., A novel cell encapsulation method using photosensitive poly (allylamine alpha-cyanocinnamylideneacetate). J. Microencapsul. 2000, 17: 245 to 251. [0253] [0253] For example, microcapsules are prepared by complexing the modified collagen with a 2-hydroxyethyl methacrylate (HEMA) terpolymer shell, methacrylic acid (MAA) and methyl methacrylate (MMA), resulting in a capsule thickness of 2 to 5 .mu.m. Such microcapsules can be further encapsulated with additional 2 to 5 .mu.my terpolymers in order to transmit a negatively charged smooth surface and to minimize the absorption of plasma protein (Chia, SM et al. Multi-layered microcapsules for cell encapsulation Biomaterials. 2002 23: 849 to 856). [0254] [0254] Other microcapsules based on alginate, a marine polysaccharide (Sambanis, A. Encapsulated islets in diabetes treatment. Diabetes Technol. Ther. 2003, 5: 665 to 668), or their derivatives. For example, microcapsules can be prepared by complexing polyelectrolytes between polyanion sodium alginate and sodium cellulose sulfate with poly (methylene-co-guanidine) hydrochloride in the presence of calcium chloride. [0255] [0255] It will be appreciated that cell encapsulation is improved when smaller capsules are used. Thus, quality control, mechanical stability, diffusion properties, and in vitro activities of encapsulated cells improved when the capsule size was reduced from 1 mm to 400.mu.m (Canaple L. et al., Improving cell encapsulation through size control J Biomater Sci Polym Ed. 2002; 13: 783 to 796). Furthermore, nanoporous biocapsules with well-controlled pore sizes as small as 7 nm, with adapted surface chemicals and the precise microarchitectures were found to successfully immunoisolate the microenvironments for the cells (Willians D. Small is beautiful: microparticle and nanoparticle technology in medical devices.Med Device Technol. 1999, 10: 6 to 9; Desai, TA Microfabrication technology for pancreatic cell encapsulation. Expert Opin Biol Ther. 2002, 2: 633 to 646. [0256] [0256] Examples of immunosuppressive agents include, but are not limited to, [0257] [0257] The cell populations of the present patent application can be provided per se, together with the culture medium containing them, isolated from the culture medium, and combined with a pharmaceutically acceptable carrier, as well as with additional agents that can promote cell grafting and / or organ function (eg, immunosuppressive agents, antibiotics, growth factor). Therefore, the cell populations of the present application can be administered in a pharmaceutically acceptable carrier or diluent, such as sterile aqueous saline and buffer solutions. The use of such carriers and diluents is well known in the art. [0258] [0258] The compositions of the present patent application may, if desired, be presented in a packaging device or dispenser, such as in an FDA approved kit, which may contain one or more dosage forms of units containing the active ingredient (for example, cells) Such packaging may, for example, comprise plastic or metal sheets, such as a pack of pills. the packaging device or dispenser may be accompanied by instructions for administration. The packaging device or dispenser may also be accompanied by a notification on a form prescribed by a government agency, which regulates the manufacture, use, or sale of pharmaceutical products, the notification of which reflects the body's approval of the compositions for administration form human or veterinary. Such notification, for example, may include labeling approved by the United States Food and Drug Administration (FDA | U.S. Food and Drug Administration) for prescription drugs or an approved product insert. Compositions comprising a preparation of the present patent application formulated in a pharmaceutically acceptable carrier can also be prepared, positioned in an appropriate container, and labeled for treating an indicated condition, as well as more detailed above. [0259] [0259] Cells prepared according to the methods of the present patent application can be administered to the individual per se, sown in a frame and / or in a pharmaceutical composition where it is mixed with appropriate carriers or excipients. [0260] [0260] As used herein, a "pharmaceutical composition" refers to a preparation of one or more of the active ingredients described herein with other chemical components, such as physiologically suitable carriers and excipients. The purpose of a pharmaceutical composition is to facilitate the administration of a compound to an organism. [0261] [0261] Hereinafter, the phrases “physiologically acceptable carrier” and “pharmaceutically acceptable carrier,” which can be used interchangeably, refer to a carrier or diluent that does not cause significant irritation to an organism and does not cancel biological activity and the properties of the administered compound. An adjuvant is included under these phrases. [0262] [0262] In this document, the term "excipient" refers to an inert substance added to a pharmaceutical composition to further facilitate the administration of an active ingredient. Examples of excipients, without limitation, include calcium carbonate, calcium phosphate, various sugars and types of starch, cellulose derivatives, gelatin, vegetable oils, and polyethylene glycols. [0263] [0263] Techniques for formulating and administering drugs can be found in the latest issue of “Remington's Pharmaceutical Sciences,” Mack Publishing Co., Easton, PA, which is incorporated herein in its entirety by reference. [0264] [0264] Suitable routes of administration may, for example, include oral, rectal, transmucosal, especially transnasal, intestinal or parenteral administration, including intramuscular, subcutaneous and intramedullary injections, as well as intrathecal, direct intraventricular, intraperitoneal, intranasal injections. , or intraocular. [0265] [0265] Alternatively, the pharmaceutical composition can be administered in a location that is not in a systemic manner, for example, by injecting the pharmaceutical composition directly into a patient's tissue region. [0266] [0266] Pharmaceutical compositions for use in accordance with the present application can therefore be formulated in the conventional manner, using one or more physiologically acceptable carriers comprising excipients and auxiliaries, which facilitate the processing of active ingredients in preparations that can be used pharmaceutically. The appropriate formulation is dependent on the chosen route of administration. [0267] [0267] For injection, the active ingredients of the pharmaceutical composition can be formulated in aqueous solutions, preferably in physiologically compatible buffers, such as Hank's solution, Ringer's solution, or physiological salt buffer. For transmucosal administration, penetrating agents appropriate for the barrier to be permeated are used in the formulation. Such penetrants are generally known in the art. [0268] [0268] Pharmaceutical compositions suitable for use in the context of the present patent application include compositions, characterized by the active ingredients being contained in an effective amount, to achieve the intended purpose. More specifically, a "therapeutically effective amount" means an amount of active ingredients (eg, a nucleic acid construct) effective in preventing, alleviating, or ameliorating the symptoms of a disorder (eg, ischemia) or prolonging the survival of the individual being treated. [0269] [0269] The determination of a therapeutically effective amount is within the skill of those skilled in the art, especially in light of the detailed disclosure provided in this document. [0270] [0270] For any preparation used in the methods of the present patent application, the dosage or therapeutically effective amount can be estimated initially from in vitro and cell culture assays. For example, a dose can be formulated in animal models to achieve a desired concentration or titer. Such information can be used to more accurately determine useful doses in humans. [0271] [0271] The therapeutic efficacy and toxicity of the active ingredients described in this document “can be determined by standard pharmaceutical procedures in vitro, in cell cultures or experimental animals. The data obtained from these in vitro assays, cell cultures and animal studies can be used in formulating a range of dosages for use in humans. The dosage may vary, depending on the dosage form employed and the route of administration used. The exact formulation, route of administration, and dosage can be chosen by the individual's physician, taking into account the patient's condition. (See, for example, Fingl, E. et al. (1975), “The Pharmacological Basis of Therapeutics,” Chap. 1, p. 1.) [0272] [0272] Depending on the severity and responsiveness of the condition being treated, the dosage can be simple, or from a variety of administrations, with the course of treatment lasting from several days to several weeks, or until the cure is effected , or until the reduction of the disease state is reached. [0273] [0273] The amount of a composition to be administered will obviously be dependent on the individual being treated, the severity of the suffering, the manner of administration, the judgment of the doctor who prescribes it, etc. [0274] [0274] As used herein, the term "method" refers to the ways, means, techniques and procedures for completing a given task including, but not limited to, those ways, means, techniques and procedures, or known, or readily developed in ways, means, techniques and procedures known to practitioners of chemical, pharmacological, biological, biochemical and medical techniques. [0275] [0275] As used herein, the term "treatment" includes nullifying, considerably inhibiting, decreasing or reversing the progress of a condition, considerable clinical or aesthetic improvement of the symptoms of a condition, or considerably preventing the appearance of clinical symptoms or effects of a condition. [0276] [0276] Various applications and aspects of the present invention application outlined above and claimed below in the claims section find experimental support in the following examples. [0277] [0277] Additional objectives, advantages and innovative features of the present patent application will become apparent to someone of ordinary skill in the art during the examination of the following examples, which should not be limiting. In addition, each of the various applications and aspects of the present patent application outlined above and claimed below in the claims section find experimental support in the following examples. EXAMPLES [0278] [0278] References are now made to the following examples, which together with the above descriptions, illustrate the present invention patent application in a non-restrictive manner. [0279] [0279] Generally, the nomenclature used in this document and the laboratory procedures used in the present application for a patent include biochemical, microbiological and recombinant molecular DNA techniques. Such techniques are explained in detail in the literature. See, for example, “Molecular Cloning: A laboratory Manual” Sambrook et al., (1989); “Current Protocols in Molecular Biology” Volumes I to III Ausubel, R. M., ed. (1994); Ausubel et al., "Current Protocols in Molecular Biology", John Wiley and Sons, Baltimore, Maryland (1989); Perbal, “A Practical Guide to Molecular Cloning”, John Wiley & Sons, New York (1988); Watson et al., “Recombinant DNA”, Scientific American Books, New York; Birren et al. (eds) “Genome Analysis: A Laboratory Manual Series”, Volumes 1-4, Cold Spring Harbor Laboratory Press, New York (1998); methodologies established in US Patents No. 4,666,828; [0280] [0280] Isolation of mesenchymal stem cells: Mesenchymal cells derived from adipose tissue and derived from bone marrow were isolated, based on their potential for plastic adhesion in an expanding medium, containing: High-glucose DMEM and 10% Bovine Serum Fetal (FBS, Hyclone, Logan, UT, USA) supplemented with 0.05 mg / ml gentamicin (Sigma) and 2 mM L-glutamine (Biological Industries, Israel). The cells were allowed to adhere for 3 to 4 days, and the non-adherent cells were washed with changes in the medium. The medium was subsequently changed with fresh medium every 3 or 4 days. [0281] [0281] Hematopoietic stem cells and progenitor cells: Hematopoietic stem cells derived from umbilical cord were isolated using CDl33 microspheres and CliniMACSO separator (Miltenyi, Inc. Auburn, CA), and were grown for 3 weeks in MEMa, supplemented with 50 ng / ml TPO, IL6, SCF, FlIt3, fetal bovine serum, + 2.5 or 5 mM nicotinamide, and / or 10, 50 or 200 ng / ml FGF4. After 3 weeks in cultivation, [0282] [0282] Maintenance and expansion: As soon as the adherent cells reached approximately 80 to 90% confluence, they were separated with 0.25% trypsin-EDTA (Sigma), washed twice in DMEM and with 10% Bovine Fetal Serum , with centrifugation, 400 g, for 5 minutes, and laminated again in a dilution of 1: 2 to 1: 1000 under the same culture conditions. [0283] [0283] Cell size measurement: Cell size was measured using Cedex Automated Cell Counter (Innovatis). The cells were diluted 1: 2 in Trypan Blue (Sigma) and the cell size was measured automatically under a microscope. [0284] [0284] Granularity measurement: After trypsin treatment, cells were analyzed for granularity by lateral dispersion FACS. [0285] [0285] Measuring the number of cells in the culture: The number of cells was measured using Cedex Automated Cell Counter (Innovatis). The cells were diluted 1: 2 in Trypan Blue (Sigma) and the number of cells was automatically measured under a microscope. [0286] [0286] Analysis of the surface antigen: At different times, the cells were separated with 0.25% trypsin-EDTA. The cells were washed with a PBS solution containing 1% BSA, and labeled (at 4 “C for 30 min) or with fluorescein isothiocyanate (FITC | fluorescein isothiocyanate), or with conjugated phycoerythrin antibodies (PE | phycoerythrin): 105 PE, 105 FITC (Serotec, Raleigh, NC), 45 FITC, 14 FITC, HLA-DR FITC, 106 PE, 31 PE (BD, Franklin Lakes NJ), 34 PE (Dako, Glostrup , Denmark), 73 PE, HLA class 1 PE, 49b PE (Pharmingen, San Diego, CA), 29 PE, 44 PE, 54 FITC, 59 PE, 90 PE (BioLegend, San Diego , CA). PE CD133 - (ACl41) (Miltenyi, Auburn, CA), FITC CD38 (Dako, Glostrup, Denmark), FITC CD19 (BD Biosciences, Franklin Lakes NI), FITC CD33 (BD Biosciences, Franklin Lakes NJ). [0287] [0287] The cells were then washed in the above buffer and analyzed using a FACScalibur * flow cytometer (Becton Dickinson, Franklin Lakes NJ). The cells were passed at a rate of approximately 1000 cells / second, using a 488 nm argon laser beam as the excitation light source. The emission of 10,000 cells was measured using logarithmic amplification, and analyzed using the CellQuest software (Becton Dickinson). Cells labeled with FITC and PE conjugated isotype control antibodies were used to determine fluorescence history. [0288] [0288] CFU-F assay: The cultured MSC's were seeded in 6-well plates, at a density of 50 to 100 cells / cm º and maintained in DMEM and 10% FBS. After 14 days, the cells were fixed using 10% cold formalin (Sigma) and labeled with Harris' hematoxylin (Sigma). Clones (clusters of more than 50 cells with an evident epicenter) are marked in blue-purple and counted using a microscope. [0289] [0289] Senescence evaluation test: Cultured MSCs were marked using the Senescence B-galactosidase Staining Kit (Cell Signaling). The cells were fixed and labeled for detection of beta-galactosidase activity at pH 6 using X-Gal and were incubated overnight at 37ºC in a dry incubator. [0290] [0290] Wound healing test in vitro: A wound was performed on the cultures of MSC's at approximately 70% confluence, using a 200 1pl or 1000 1nl tip. Four days later, the cells were fixed using 10% cold formalin (Sigma) and labeled with Harris hematoxylin (Sigma). The in vitro wound healing process was evaluated using a microscope. [0291] [0291] Wound healing test in vitro: A wound was performed on the cultures of MSC's at approximately 70% confluence, using a 200 pl or 1000 ul tip. Four days later, the cells were fixed using 10% cold formalin [0292] [0292] Treatment of mesenchymal cultures with nicotinamide: Mesenchymal cultures were started as described above, and supplemented with 1 to 15 mM nicotinamide alone, or in combination with growth factors or only growth factors, incubated at 37'C in a humidified atmosphere of 5% CO, in the air. With each passage and change of medium, cultures were supplemented with mesenchymal medium, nicotinamide and growth factors. [0293] [0293] In some experiments, adherent cells were cultured with or without nicotinamide and factors indicated 24 hours before passage 4, and The medium was replaced with a medium without fetal bovine serum or FGF4. [0294] [0294] In Vitro Migration Assay: An RPMI plus 10% FCS (0.6 ml) containing 100 ng / ml CXCL12 (R&D Systems) was placed in the lower chamber of a 24-well culture plate from transwell Costar (Corning, Inc, Corning, NY). The cells (2 x 10º) in a 100-pl medium were introduced into the upper chamber, over a porous membrane (pore size, 5 µm). After 4 hours, cells were collected from both chambers and counted using flow cytometry (FACSsort, Becton Dickinson and Co, San Jose, CA, USA). Spontaneous migration was performed as a control without CXCL12 in the lower chamber. [0295] [0295] In vivo analysis of targeting: NOD / SCID mice (aged 8 to 10 weeks) (Harlan Ltd., Israel) were sub-lethally irradiated (at 375cGy to 67cGy / min) and inoculated 24 hours later through the vein of the tail, or with CFSE-labeled mesenchymal stem cells grown in the presence of nicotinamide or with CFSE-labeled mesenchymal stem cells grown in the absence of nicotinamide. The mice were sacrificed 24 hours after the injection and bone marrow or other tissue samples were obtained. The targeting of human cells was detected by flow cytometry, through the visualization of CFSE-labeled cells on a background or on unmarked murine cells. The clear fluorescence of CFSE is sufficient to separate labeled human cells from unlabeled murine cells by at least 1 record. To quantify the targeting of human progenitor cells, bone marrow cells were labeled with monoclonal antibodies to APC-conjugated anti-human cells and human / CFSE * marker cells were enumerated. For each sample, 100,000 events were recorded and analyzed. [0296] [0296] Mesenchymal cell transplantation in NOD / SCID mice: NOD / SCID mice were created and kept in sterilized and intra-ventilated cells (Techniplast, Bugugiatte, Italy). the eight-week-old mice were sub-lethally irradiated, as described above. The mice were inoculated through the tail vein with mesenchymal cells grown in the presence or absence of nicotinamide. To avoid donor variability, mesenchymal cells from different units were grouped and used for the expansion of cultures, such as group injection. The mice were sacrificed at week 4, and the marrow samples were obtained by rinsing their femurs and tibiae with IMDM at 4 ºC. Cytometric flow analysis of NOD / SCID marrow cells was performed as described above, using monoclonal antibodies against human cell surface differentiation antigens, to identify the human cell graft. [0297] [0297] Delayed Type Hypersensitivity Assay: BALB / C mice were sensitized with oxazolone (4-ethoxymethylene-2-phenyloxazol-5-one), and 6 days later, stimulated - with oxazolone, injected into the ear. Immune modulation through the candidates' compositions, as indicated, was determined 24 hours after topical administration, by measuring the thickness of the ear with a caliper. [0298] [0298] Growth Factor Secretion: The MSC medium was cultured as indicated, and was devoid of fetal bovine serum and FGF4 24 hours before passage 4 was collected and tested by ELISA for growth factors and factors related to immunity secreted in the medium (human growth factor HGF, transforming growth factor beta TGF-B, keratinocyte growth factor KGF and interleukin 6 IL-6). The ELISA was performed using sandwich ELISA kits in the solid stage, specific for humans KGF (R&D systems, catHDKG00), IL-6 (R&D systems, catHD6050), TGF-RB1 (R&D systems, catfDB100B), HGF (R&D systems , catfDHG00). [0299] [0299] Keratinocyte Proliferation Assay: Normal human epidermal keratinocytes (Promocell, GmbH, Heidelberg, Germany) were cultured by passing through a keratinocyte growth medium, separated and seeded again in keratinocyte growth medium (containing 50% of a mixture of supplements) diluted, as indicated, with the conditioned medium of MSC's. The medium was changed twice a week, and the keratinocytes were separated and counted after reaching 90% confluence. [0300] [0300] Assay Similar to the Merged Lymphocyte Reaction: Peripheral blood-derived mononuclear cells (MNCs), containing more than 50% T-cells, were isolated by floating density centrifugation and activated with 3 ug / ml phytohemagglutinin (PHA phytohemagglutinin). After activation, cells were cultured with or without conditioned medium from the culture of MSC's and other factors. The response of PBMN cells to PHA activation was measured by the extent of TNF alpha secretion in the middle cell (pg / ml), 72 hours after initial activation, measured by ELISA. [0301] [0301] Statistics- The Wilcoxon Non-parametric Classification Test (Wilcoxon Rank Test) was applied to test the differences between the study groups. All tests applied were two-tailed, and a p value of 5% was considered statistically significant. The data were analyzed using SAS software (SAS Institute, Cary, NC). EXAMPLE 1 Analysis of nicotinamide in mesenchymal stem cells grown in the presence of growth factors MATERIALS AND METHODS [0302] [0302] Mesenchymal stem cells were selected and cultured in the presence of specific growth factors (basic fibroblast growth factor - bFGF, epidermal growth factor in connection with heparin - HB-EGF, fibroblast growth factor 4 - FGF-4 and platelet-derived growth factor, homodimer, subunit B, PDGF-BB), in the presence and absence of nicotinamide for three or four passages, and the number and size of cells was calculated. [0303] [0303] Two concentrations (of and 50 ng / ml) of each of the following factors were examined. [0304] [0304] The experimental groups were as follows: [0305] [0305] In addition, the influence of the combination: 5 mM NAM + 50 ng / ml FGF4 + 50 ng / ml PDGF-BB was examined in comparison to an individual supplement. RESULTS [0306] [0306] Figure 1 illustrates that basic FGF has a negative effect on nicotinamide's ability to increase mesenchymal stem cell proliferation. [0307] [0307] Figures 2A - B illustrate that the growth factor similar to the binding of EGF to heparin (HB-EGF) has a negative effect on the ability of nicotinamide to increase the proliferation of mesenchymal stem cells. [0308] [0308] Figures 3 to 5. illustrate that nicotinamide has a potentiating effect on FGF4's ability to increase the proliferation of mesenchymal stem cells. [0309] [0309] Figures 6A - D illustrate that PDGF-BB has an inconsistent effect on nicotinamide's ability to increase mesenchymal stem cell proliferation. [0310] [0310] Figures 7A - D illustrate that MSC cultures treated with PDGF-BB or a combination of PDGF-BB + NAM are contaminated with a greater fraction of cells that are not MSC's, when compared to cultures treated without PDGF-BB. [0311] [0311] Figures 8A to B, 9A to B and 10º to H illustrate that the combination of the three factors - FGF4, nicotinamide and PDGF-BB - have a detrimental effect on the proliferation of mesenchymal stem cells, when compared with the effect of FGF4 and nicotinamide in the absence of PDGF-BB. [0312] [0312] Figure 27 illustrates the consistent synergistic effect of nicotinamide and FGF4 combined in expansion (cumulative cell count) across passages. [0313] [0313] The present inventors demonstrated that nicotinamide increased the seeding efficiency (selection) of MSC's derived from bone marrow. The phenotypic characterization of these cells, after a nicotinamide passage, is shown in Figures 11 and 13. Figures 15, 17A to B, 22 and 26 illustrate the effect of nicotinamide on the expansion rate of bone marrow-derived MSCs. Low concentrations of nicotinamide (for example, 0.1 mM) [0314] [0314] The phenotypic characterization of MSC's derived from adipose tissue is shown in Figure 12. As illustrated in Figures 14 and 16, nicotinamide considerably improved the expansion of mesenchymal stem cells derived from adipose tissue in the culture. EXAMPLE 4 Nicotinamide increases tissue targeting of cultured mesenchymal cells [0315] [0315] To assess the effect of nicotinamide on targeting cultured mesenchymal cells, NOD / SCID mice are transplanted either with uncultivated mesenchymal cells, or with their total progeny, after 3 weeks of cultivation with cytokines, with or without nicotinamide. Before transplantation, cells are labeled with CFSE. Twenty-four hours after transplantation, the total number of CFSE-labeled cells and CFSE-labeled mesenchymal cells that went to the bone marrow of the recipient mouse are quantified by FACS. [0316] [0316] The results indicate an effect of nicotinamide in targeting the tissues of mesenchymal cells, if the targeting of mesenchymal cells treated with nicotinamide is significantly greater than the targeting of non-cultured mesenchymal cells and not subject to nicotinamide. EXAMPLE 5 Nicotinamide increases the functionality of chemokine receptors and adhesion molecules [0317] [0317] In order to determine the role of adhesion molecules and related molecules in nicotinamide-mediated improvement in cell targeting and grafting, the effect of nicotinamide on in vitro migration and the functionality of adhesion molecules can be tested. [0318] [0318] Using a transwell migration assay, CXCL12-induced migration of cultured and non-cultured mesenchymal cells is tested, evaluating the effects of nicotinamide on the function of integrin adhesion molecules. The improved stimulation of migration in cells treated with nicotinamide, compared to cells cultured without nicotinamide or cells not cultured, suggests that treatment of mesenchymal cells with nicotinamide can potentially increase the responsiveness of adhesion molecules to their ligands, [0319] [0319] The functional quality of the cell binding to adhesion molecules can also be investigated using shear flow analysis. The strong effect of nicotinamide on binding and retention mediated by adhesion molecules on substrate adhesion molecules can be evidenced by the significantly improved percentage of cells initially established, resistant to shear stress removal and evident in nicotinamide-treated mesenchymal cells. [0320] [0320] Nicotinamide treatment is tested for the ability to improve the targeting and grafting of transplanted cells by restocking the NOD / SCID mouse. In order to evaluate the repopulation capacity, the NOD / SCID mouse is transplanted with uncultivated mesenchymal cells (n = 12) over a range of doses, in order to obtain a sub-ideal transplant, and the posterior non-graft in a fraction of the mouse. or your progeny after 3 weeks of expansion with cytokines. Human cell grafting is evaluated at 4 weeks after transplantation. Mice are marked as positively grafted if 0.5% of the recipient's bone marrow cells express human CD45 antigen (CD45 +). In the event that the presence of nicotinamide in the culture results in a superior and clear graft of mesenchymal cells in the mouse at a predetermined dose range, while untreated cells do not graft or graft poorly, it can be concluded that nicotinamide is efficient in graft improvement and targeting transplanted mesenchymal cells. EXAMPLE 7 Further analysis of the effect of nicotinamide on mesenchymal stem cells MATERIALS AND METHODS [0321] [0321] Mesenchymal stem cells were isolated using the plastic adhesion method, as described above, and cultured by several passages with fetal bovine serum, t + NAM. The cells were selected in the presence of NAM. [0322] [0322] At approximately 80% confluence, adherent cells were collected after treatment with trypsin, counted, characterized and seeded again at a concentration of 1 x 10º cells / cm . [0323] [0323] Measurement of VCAM1 / CD106: After treatment with trypsin, cells were analyzed for CDI06 expression in FACS using anti-human CD106 PE antibodies. [0324] [0324] CD54 measurement: After treatment with trypsin, cells were analyzed for CDS54 expression in FACS using anti-human CD54 antibodies. RESULTS [0325] [0325] Figure 18 illustrates that the effect of nicotinamide on cell count was evident in large batches of mesenchymal stem cells, indicating that large commercial batches of MSC's can be manufactured with fewer passages. This guarantees a better quality of therapeutic cells due to a shorter cultivation time and preservation of the characteristics of stem cells through nicotinamide. [0326] [0326] Figure 19 illustrates that the effect of nicotinamide is not dependent on a specific lot of serum, and presents the results of one of the two experiments performed. The cultures in these experiments were treated individually (each group was passed when reaching the confluence). [0327] [0327] The number of senescent cells was reduced after culturing in nicotinamide (Figures 23 A - D). [0328] [0328] Figure 24A illustrates that mesenchymal stem cells cultured in the presence of nicotinamide express more adhesion molecules of VCAM1 / CD106 than mesenchymal stem cells cultured in the absence of nicotinamide, under identical conditions. [0329] [0329] Figure 24B illustrates that mesenchymal stem cells cultured in the presence of nicotinamide express less CD54 than mesenchymal stem cells cultured in the absence of nicotinamide, under identical conditions. [0330] [0330] Figure 25 illustrates that mesenchymal stem cells cultured in the presence of nicotinamide have greater capacity to perform wound closure than mesenchymal stem cells cultured in the absence of nicotinamide, under identical conditions. [0331] [0331] The cultivation of mesenchymal stem cells in the presence of nicotinamide and FGF4 provides a synergistic increase in the expansion potential of the mesenchymal stem cells, while maintaining the cells in an undifferentiated state (see Figures 3A to 3D, 4A to 4B, 5A to 5D and 27). In order to further characterize the expanded MSCs in these cultures, the secretion of cytokines in the culture medium was measured by ELISA. [0332] [0332] Figures 28 to 31 illustrate the significant increase in hepatocyte growth factor (HGF, Figure 28), transforming growth factor beta (TGF-B, Figure 29) and keratinocyte growth factor (KGF, Figure 30) with nicotinamide and FGF4 combined, compared to nicotinamide alone. THE [0333] [0333] When the medium of the mesenchymal stem cell grown with nicotinamide, or with nicotinamide and FGF4, was tested for effect on inflammation in the delayed hypersensitivity test in vivo, the reduction in the inflammatory response to stimulate with the sensitizing allergen (Oxazolone) was clearly observed (data not shown). Additional analyzes in the ex vivo lymphocyte reaction type assay clearly demonstrated the anti-inflammatory potential of the culture medium of MSC, nicotinamide and nicotinamide with FGF4, in reducing TNFa secretion through peripheral blood mononuclear cells, in response to PHA activation (data not shown). [0334] [0334] When the medium of mesenchymal stem cells cultured with nicotinamide or nicotinamide and FGF4 was tested for effect on keratinocyte proliferation in vitro, a significant induction of keratinocyte proliferation was clearly observed (data not shown). [0335] [0335] Thus, adherent mesenchymal stem cells, cultured with nicotinamide and nicotinamide in combination with FGF4, release biologically active factors into the medium, including factors with proliferation-inducing and anti-inflammatory activity. [0336] [0336] Proliferation and cell size distribution in mesenchymal stem cells, derived from adipose tissue and cultured with nicotinamide, with or without additional FGF4, was tested in up to 4 culture passages. [0337] [0337] Figures 32 to 33 illustrate the impressive effect of nicotinamide and FGF4 combined in the proliferation of adherent cells derived from adipose tissue, expressed as the total number of nucleated cells in the cultures, when compared to controls as well as the cultures treated with nicotinamide. [0338] [0338] The size of mesenchymal stem cells in culture is commonly used as an indicator of the degree of differentiation of MSCs, with the most prevalent undifferentiated state in cells of smaller size. Figure 34 illustrates the increased prevalence of smaller cells in cultures of MSC's derived from adipose tissue exposed to nicotinamide, and the even greater prevalence of smaller cells among MSC's derived from adipose tissue exposed to nicotinamide and FGF4. [0339] [0339] Thus, these results indicate that a combination of nicotinamide and FGF4 synergistically increase the proliferation rate of mesenchymal cells derived from adipose tissue, while efficiently maintaining cells in an undifferentiated state. EXAMPLE 10 Effect of Nicotinamide and FGF4 on Hematopoietic Stem Cell Differentiation [0340] [0340] In order to determine whether the effects of nicotinamide and FGF4 combined on mesenchymal stem cells are a specific or generalized phenomenon, or if they are not, hematopoietic stem cells were grown with and without FGF4 and nicotinamide, and the degree of differentiation of the component subpopulations were tested according to the cell surface markers. MATERIALS AND METHODS [0341] [0341] Hematopoietic stem cells, derived from CD133 + umbilical cord, were isolated and cultured for 3 weeks in a medium supplemented with newly acting growth factors and fetal bovine serum, nicotinamide and / or FGF4. After 3 weeks in culture, cells were marked with surface markers (CD38, CD133, CD19) and cell populations were determined by FACS analysis. Cell proliferation was assessed by total cell count over three weeks. Results [0342] [0342] FGF4 does not affect the differentiation of hematopoietic stem cells in culture: [0343] [0343] When hematopoietic premature progenitor cells (CD133 +) are cultured for 3 weeks in the presence of 2.5 or 5 mM nicotinamide, the proportion of differentiated cells decreases significantly, and the increase in the fraction of stem cells and cells progenitors is clearly observed (see Figure 35, columns 1, 2 and 3). Exposure of cells from 10 to 200 ng / ml of FGF4, on the other hand, has no effect on the degree of differentiation in a three-week culture, as evidenced by the fraction of cells that express CD38 and CD133 (see Figures 35 and 36, columns 1 and 4 to 6). The addition of FGF4, with or without nicotinamide, also had no discernible effect on total cell proliferation in culture (results not shown). [0344] [0344] The addition of FGF4 to cells cultured with nicotinamide did not increase or reduce nicotinamide inhibition of hematopoietic stem cell differentiation at any concentration of FGF4 (see Figures 35 to 36, ranges 1 and 7 to 12). [0345] [0345] FGF4 does not affect the differentiation of HSC in the compromised line of lymphoids or myeloids: [0346] [0346] In order to determine whether compromised hematopoietic stem cells were affected by exposure to FGF4 during culture, the abundance of cells expressing CD33 and CD19, representing differentiated and compromised cells of the lymphoid and myeloid lineage, respectively, was measured in cultures three weeks. Although nicotinamide has consistently reduced the fraction of compromised cells (see Figures 37 and 38, ranges 1 to 3 and 7 to 12), the addition of FGF4 alone, or in combination with nicotinamide, had no effect on the abundance of cells compromised myeloids or lymphoids (see Figures 37 and 38, ranges 1, 4 to 6 and 7-12); [0347] [0347] Thus, these results clearly indicate that the FGF4 proliferation-enhancing effects observed in mesenchymal stem cell culture, and the synergistic effects of mesenchymal stem cell exposure in nicotinamide and FGF4 combined, are not a phenomenon in general, and are not seen in ex-vivo hematopoietic stem cell cultures. [0348] [0348] Isolation: Mesenchymal cells derived from adipose tissue and derived from bone marrow are isolated, based on their potential for plastic adhesion in an expanding medium, containing: DMEM of high glucose and 10% of Bovine Fetal Serum (FBS, Hyclone , Logan, UT, USA) 0.05 mg / ml gentamicin (Sigma) and 2 mM L-glutamine (Biological Industries, Israel). The cells were allowed to adhere in the presence of nicotinamide +/- FGF4 for 3 to 4 days, and the non-adherent cells were washed with changes in the medium. [0349] [0349] Mesenchymal stem cells are cultured by several passages (1 to 3) with fetal bovine serum, + NAM, t + FGF4. The cells can be grown on plastic adherent plates. [0350] [0350] At approximately 80% confluence, adherent cells are collected after treatment with trypsin, counted, characterized and selected through the expression of CD1I05, CD106 or STRO-1 using directly or indirectly conjugated mice anti-human antibodies (Miltenyi Biotec), and organization of magnetic cells (MACS) or organization of fluorescence activated cells (FACS), sown again for future expansion. [0351] [0351] Measurement of VCAM1 / CD106, CD105 or STRO-1: after treatment with trypsin, cells are analyzed for expression of CD106, expression of STRO-1 or CDIO05 in FACS using anti-human CD106 PE antibodies, antibodies anti-human CD105 or anti-STRO-1 antibodies. EXAMPLE 12 Combined use of nicotinamide +/- FGF4 preceded by a selection using VCAM1 / CD1I06. CD1I05 or STRO-l for selection of mesenchymal stem cells MATERIALS AND METHODS [0352] [0352] Selection of MSC derived from adipose tissues and derived from bone marrow: Mesenchymal cells derived from adipose tissues and derived from bone marrow are selected by the expression CD1I05, CD106 or STRO-1, using anti-human antibodies from conjugated mice directly or indirectly (Miltenyi Biotec) and organization of magnetic cells (MACS). The selected cells are seeded at a concentration of 6 x 10º cells / cm In an expansion medium containing: DMEM of high glucose and 10% Fetal Bovine Serum (FBS, Hyclone, Logan, UT, USA) 0.05 mg / ml of gentamicin (Sigma) and 2 mM of L-glutamine (Biological Industries, Israel). The cells are allowed to adhere for 3 to 4 days, and the non-adherent cells were washed with changes in the medium. [0353] [0353] Culture of MSC's populations selected in NAM + FGF4: Mesenchymal stem cells are cultured by several passages with fetal bovine serum, + NAM, + FGF4. [0354] [0354] It is appreciated that certain features of the present invention patent application, which are, for clarity, described in the context of separate applications, can also be provided in combination in a single application. Conversely, several features of the present application for a patent, which are briefly described in the context of a simple application, can also be provided separately or in any suitable subcombination. [0355] [0355] Although the present application for a patent is described in conjunction with respective specific applications, it is evident that many alternatives, modifications and variations may be apparent to those skilled in the art. Consequently, it is intended to cover all of the aforementioned alternatives, modifications and variations that fall within the spirit and broad scope of the attached claims. All GenBank Accession publications, patents, patent applications and numbers mentioned in that specification are incorporated in this document in their entirety by reference in that specification, to the same extent as if each individual publication, patent or patent application or patent number GenBank Accession were specifically and individually indicated to be incorporated herein by reference. In addition, the citation or identification of any reference in that application should not be construed as an admission that such reference is available as a prior art to the present invention.
权利要求:
Claims (11) [1] 1. “CULTURE OF MESENQUIMAL STEM CELLS”, covers the method of culture of non-differentiated mesenchymal stem cells (MSCs), characterized by understanding the culture of a population of non-differentiated MSCs in a medium comprising nicotinamide and fibroblast growth factor 4 (FGF4) under conditions that do not induce differentiation, thus cultivating MSCs. [2] 2. “CULTURE OF MESENQUIMAL STEM CELLS”, covers the method according to claim 1, characterized in that said medium is devoid of additional growth factors and / or added differentiation factors. [3] 3. “MESENQUIMAL STEM CELL CULTURE”, covers the method according to claim 1 or 2, characterized in that said culture is carried out for at least 1 week or 3 passages. [4] 4. "MESENQUIMAL STEM CELL CULTURE", encompasses the method according to any one of claims 1 to 3, characterized in that the concentration of said nicotinamide is from 1 to 20 mM. [5] 5. "MESENQUIMAL STEM CELL CULTURE", encompasses the method according to any one of claims 1 to 4, characterized in that said medium is devoid of platelet-derived growth factor (PDGF). [6] 6. “CULTURE OF MESENQUIMAL STEM CELLS”, covers the method according to any one of claims 1 to 5, characterized in that said non-differentiated MSC is a population sown of mesenchymal stem cells grown in a medium devoid of nicotinamide, before said culture in the medium comprises nicotinamide and FGF4. [7] 7. "CULTURE OF MESENQUIMAL STEM CELLS", covers the method according to claim 6, characterized in that said culture in said medium, comprising nicotinamide and FGF-4, is carried out for at least one week. [8] 8. "CULTURE OF MESENQUIMAL STEM CELLS", covers the method according to claim 6, characterized in that said culture in said medium devoid of nicotinamide is carried out for at least one day. [9] 9. "CULTURE OF MESENQUIMAL STEM CELLS", covers the method according to any one of claims 1 to 8, characterized in that the calcium concentration of said medium is greater than 1.8 mM. [10] 10. "CULTURE OF MESENQUIMAL STEM CELLS", covers the method of generating useful cells for transplantation in an individual, the method characterized by comprising: a) the culture of mesenchymal stem cells, according to the method of any of the claims from 1 to 9, to generate a population of cultured mesenchymal stem cells; b) the contact of said population of cultured mesenchymal stem cells with a differentiating agent, thus generating useful cells for transplantation in an individual. [11] 11. "MESENQUIMAL STEM CELL CULTURE", encompasses the method according to claim 10, characterized in that said differentiating agent comprises a growth factor or a polynucleotide that encodes said growth factor.
类似技术:
公开号 | 公开日 | 专利标题 JP2018064572A|2018-04-26|Culturing of mesenchymal stem cells Cheng et al.2012|The influence of spheroid formation of human adipose-derived stem cells on chitosan films on stemness and differentiation capabilities Chong et al.2012|Human peripheral blood derived mesenchymal stem cells demonstrate similar characteristics and chondrogenic differentiation potential to bone marrow derived mesenchymal stem cells Heidari et al.2013|Comparison of proliferative and multilineage differentiation potential of sheep mesenchymal stem cells derived from bone marrow, liver, and adipose tissue US20190367883A1|2019-12-05|Regulating stem cells Bakhtina et al.2014|Characterization and differentiation potential of rabbit mesenchymal stem cells for translational regenerative medicine Sreejit et al.2012|Generation of mesenchymal stem cell lines from murine bone marrow US20120276070A1|2012-11-01|Induced Pluripotent Stem Cells and Related Methods US11174461B2|2021-11-16|Colony forming medium and use thereof CN113957043A|2022-01-21|Cell culture method of mesenchymal stem cells TW201920658A|2019-06-01|Stem cells derived from young pig and preparation method therefor KANNAIYAN et al.2015|Clinical prospects of scale-up foetal Whartons jelly derived multipotent stromal cells to fulfil the therapeutic demands US9982232B2|2018-05-29|Methods of generating cells with multilineage potential WO2021227573A1|2021-11-18|Xeno-free culture medium and method for expansion of mesenchymal stem cells by means of using same Ferro2013|Isolation, characterization and Ex-Vivo expansion of human synovial tissue derived-mesenchymal stem/stromal cells Clough2013|IL7 as a marker of a subset of bone marrow mesenchymal stromal cells Hiwase2010|Characterisation of placental mesenchymal stromal cells and their role in cord blood transplantation. IL193947A|2013-08-29|Cultured cells that stain as cd31bright, method for stimulating the same to differentiate into a progenitor/precursor cell population |, use of said pcp in the preparation of medicaments and apparatus for implantation comprising the same
同族专利:
公开号 | 公开日 CN104204192A|2014-12-10| HK1202581A1|2015-10-02| CN104204193B|2016-10-26| IL234095A|2018-06-28| CA2863795A1|2013-08-22| US20150004146A1|2015-01-01| JP2018064572A|2018-04-26| JP2015507921A|2015-03-16| WO2013121427A1|2013-08-22| HK1199905A1|2015-07-24| IL234095D0|2014-09-30| US10047345B2|2018-08-14| EP2814950A1|2014-12-24| IN2014MN01680A|2015-07-03| JP6348848B2|2018-06-27| EP2814951B1|2019-04-03| EP2814951A1|2014-12-24| CN104204193A|2014-12-10| US20150064273A1|2015-03-05| AU2013219945B2|2017-12-07| WO2013121426A1|2013-08-22| AU2013219945A1|2014-08-28| SG11201404608WA|2014-09-26|
引用文献:
公开号 | 申请日 | 公开日 | 申请人 | 专利标题 US3687808A|1969-08-14|1972-08-29|Univ Leland Stanford Junior|Synthetic polynucleotides| US3715345A|1970-08-03|1973-02-06|Lilly Co Eli|Glucagon separation process| NL154598B|1970-11-10|1977-09-15|Organon Nv|PROCEDURE FOR DETERMINING AND DETERMINING LOW MOLECULAR COMPOUNDS AND PROTEINS THAT CAN SPECIFICALLY BIND THESE COMPOUNDS AND TEST PACKAGING.| NL154599B|1970-12-28|1977-09-15|Organon Nv|PROCEDURE FOR DETERMINING AND DETERMINING SPECIFIC BINDING PROTEINS AND THEIR CORRESPONDING BINDABLE SUBSTANCES, AND TEST PACKAGING.| NL154600B|1971-02-10|1977-09-15|Organon Nv|METHOD FOR THE DETERMINATION AND DETERMINATION OF SPECIFIC BINDING PROTEINS AND THEIR CORRESPONDING BINDABLE SUBSTANCES.| US3901654A|1971-06-21|1975-08-26|Biological Developments|Receptor assays of biologically active compounds employing biologically specific receptors| US3853987A|1971-09-01|1974-12-10|W Dreyer|Immunological reagent and radioimmuno assay| US3867517A|1971-12-21|1975-02-18|Abbott Lab|Direct radioimmunoassay for antigens and their antibodies| NL171930C|1972-05-11|1983-06-01|Akzo Nv|METHOD FOR DETERMINING AND DETERMINING BITES AND TEST PACKAGING.| US3850578A|1973-03-12|1974-11-26|H Mcconnell|Process for assaying for biologically active molecules| US3876623A|1973-05-09|1975-04-08|Lilly Co Eli|Process for purifying insulin| US3863008A|1973-06-11|1975-01-28|American Home Prod|Somatostatin as stimulant of luteinizing hormone secretion| US3935074A|1973-12-17|1976-01-27|Syva Company|Antibody steric hindrance immunoassay with two antibodies| US3996345A|1974-08-12|1976-12-07|Syva Company|Fluorescence quenching with immunological pairs in immunoassays| US4034074A|1974-09-19|1977-07-05|The Board Of Trustees Of Leland Stanford Junior University|Universal reagent 2-site immunoradiometric assay using labelled anti | US3984533A|1975-11-13|1976-10-05|General Electric Company|Electrophoretic method of detecting antigen-antibody reaction| US4036945A|1976-05-03|1977-07-19|The Massachusetts General Hospital|Composition and method for determining the size and location of myocardial infarcts| US4098876A|1976-10-26|1978-07-04|Corning Glass Works|Reverse sandwich immunoassay| US4331647A|1980-03-03|1982-05-25|Goldenberg Milton David|Tumor localization and therapy with labeled antibody fragments specific to tumor-associated markers| US4879219A|1980-09-19|1989-11-07|General Hospital Corporation|Immunoassay utilizing monoclonal high affinity IgM antibodies| US4469863A|1980-11-12|1984-09-04|Ts O Paul O P|Nonionic nucleic acid alkyl and aryl phosphonates and processes for manufacture and use thereof| US5023243A|1981-10-23|1991-06-11|Molecular Biosystems, Inc.|Oligonucleotide therapeutic agent and method of making same| US4476301A|1982-04-29|1984-10-09|Centre National De La Recherche Scientifique|Oligonucleotides, a process for preparing the same and their application as mediators of the action of interferon| US4687808A|1982-08-12|1987-08-18|Biospecific Technologies, Inc.|Activation of biocompatible polymers with biologicals whose binding complements are pathological effectors| IL68218A|1983-03-23|1985-12-31|Univ Ramot|Compositions for cartilage repair comprising embryonal chondrocytes| US4816567A|1983-04-08|1989-03-28|Genentech, Inc.|Recombinant immunoglobin preparations| US5011771A|1984-04-12|1991-04-30|The General Hospital Corporation|Multiepitopic immunometric assay| US5550111A|1984-07-11|1996-08-27|Temple University-Of The Commonwealth System Of Higher Education|Dual action 2',5'-oligoadenylate antiviral derivatives and uses thereof| US4666828A|1984-08-15|1987-05-19|The General Hospital Corporation|Test for Huntington's disease| US5034506A|1985-03-15|1991-07-23|Anti-Gene Development Group|Uncharged morpholino-based polymers having achiral intersubunit linkages| US5235033A|1985-03-15|1993-08-10|Anti-Gene Development Group|Alpha-morpholino ribonucleoside derivatives and polymers thereof| US5185444A|1985-03-15|1993-02-09|Anti-Gene Deveopment Group|Uncharged morpolino-based polymers having phosphorous containing chiral intersubunit linkages| US5405938A|1989-12-20|1995-04-11|Anti-Gene Development Group|Sequence-specific binding polymers for duplex nucleic acids| US5166315A|1989-12-20|1992-11-24|Anti-Gene Development Group|Sequence-specific binding polymers for duplex nucleic acids| US4683202B1|1985-03-28|1990-11-27|Cetus Corp| US4801531A|1985-04-17|1989-01-31|Biotechnology Research Partners, Ltd.|Apo AI/CIII genomic polymorphisms predictive of atherosclerosis| JP2703893B2|1985-07-05|1998-01-26|ホワイトヘッド・インスティテュ−ト・フォ−・バイオメディカル・リサ−チ|Epithelial cells expressing foreign genetic material| US4980286A|1985-07-05|1990-12-25|Whitehead Institute For Biomedical Research|In vivo introduction and expression of foreign genetic material in epithelial cells| GB8606913D0|1986-03-20|1986-04-23|Hider R C|Treatment of sickle cell disease| EP0260032B1|1986-09-08|1994-01-26|Ajinomoto Co., Inc.|Compounds for the cleavage at a specific position of RNA, oligomers employed for the formation of said compounds, and starting materials for the synthesis of said oligomers| US5276019A|1987-03-25|1994-01-04|The United States Of America As Represented By The Department Of Health And Human Services|Inhibitors for replication of retroviruses and for the expression of oncogene products| US5264423A|1987-03-25|1993-11-23|The United States Of America As Represented By The Department Of Health And Human Services|Inhibitors for replication of retroviruses and for the expression of oncogene products| US5166320A|1987-04-22|1992-11-24|University Of Connecticut|Carrier system and method for the introduction of genes into mammalian cells| US4806484A|1987-08-07|1989-02-21|Igb Products, Ltd.|Perfusion airlift bioreactor| US4946778A|1987-09-21|1990-08-07|Genex Corporation|Single polypeptide chain binding molecules| JP3015383B2|1987-09-11|2000-03-06|ホワイトヘツド・インスチチユート・フオー・バイオメデイカル・リサーチ|Transduced fibroblasts and their use| US4924624A|1987-10-22|1990-05-15|Temple University-Of The Commonwealth System Of Higher Education|2,',5'-phosphorothioate oligoadenylates and plant antiviral uses thereof| US5188897A|1987-10-22|1993-02-23|Temple University Of The Commonwealth System Of Higher Education|Encapsulated 2',5'-phosphorothioate oligoadenylates| US5403711A|1987-11-30|1995-04-04|University Of Iowa Research Foundation|Nucleic acid hybridization and amplification method for detection of specific sequences in which a complementary labeled nucleic acid probe is cleaved| EP0348458B1|1987-11-30|1997-04-09|University Of Iowa Research Foundation|Dna molecules stabilized by modifications of the 3'-terminal phosphodiester linkage and their use as nucleic acid probes and as therapeutic agents to block the expression of specifically targeted genes| DE3851153T2|1987-12-11|1995-01-05|Whitehead Biomedical Inst|GENETIC MODIFICATION OF ENDOTHELIAL CELLS.| EP0400047B1|1988-02-05|1997-04-23|Whitehead Institute For Biomedical Research|Modified hepatocytes and uses therefor| JPH01222780A|1988-03-02|1989-09-06|Tosoh Corp|Method for refolding urokinase precursor-like protein| EP0406309A4|1988-03-25|1992-08-19|The University Of Virginia Alumni Patents Foundation|Oligonucleotide n-alkylphosphoramidates| US5081035A|1988-04-18|1992-01-14|The University Of Michigan|Bioreactor system| US5278302A|1988-05-26|1994-01-11|University Patents, Inc.|Polynucleotide phosphorodithioates| US5216141A|1988-06-06|1993-06-01|Benner Steven A|Oligonucleotide analogs containing sulfur linkages| GB8823869D0|1988-10-12|1988-11-16|Medical Res Council|Production of antibodies| US5272057A|1988-10-14|1993-12-21|Georgetown University|Method of detecting a predisposition to cancer by the use of restriction fragment length polymorphism of the gene for human poly polymerase| US5256775A|1989-06-05|1993-10-26|Gilead Sciences, Inc.|Exonuclease-resistant oligonucleotides| US6326198B1|1990-06-14|2001-12-04|Regents Of The University Of Michigan|Methods and compositions for the ex vivo replication of stem cells, for the optimization of hematopoietic progenitor cell cultures, and for increasing the metabolism, GM-CSF secretion and/or IL-6 secretion of human stromal cells| US5399493A|1989-06-15|1995-03-21|The Regents Of The University Of Michigan|Methods and compositions for the optimization of human hematopoietic progenitor cell cultures| US5437994A|1989-06-15|1995-08-01|Regents Of The University Of Michigan|Method for the ex vivo replication of stem cells, for the optimization of hematopoietic progenitor cell cultures, and for increasing the metabolism, GM-CSF secretion and/or IL-6 secretion of human stromal cells| US5192659A|1989-08-25|1993-03-09|Genetype Ag|Intron sequence analysis method for detection of adjacent and remote locus alleles as haplotypes| US5399676A|1989-10-23|1995-03-21|Gilead Sciences|Oligonucleotides with inverted polarity| US5264564A|1989-10-24|1993-11-23|Gilead Sciences|Oligonucleotide analogs with novel linkages| US5264562A|1989-10-24|1993-11-23|Gilead Sciences, Inc.|Oligonucleotide analogs with novel linkages| US5177198A|1989-11-30|1993-01-05|University Of N.C. At Chapel Hill|Process for preparing oligoribonucleoside and oligodeoxyribonucleoside boranophosphates| US5587361A|1991-10-15|1996-12-24|Isis Pharmaceuticals, Inc.|Oligonucleotides having phosphorothioate linkages of high chiral purity| US5220007A|1990-02-15|1993-06-15|The Worcester Foundation For Experimental Biology|Method of site-specific alteration of RNA and production of encoded polypeptides| US5149797A|1990-02-15|1992-09-22|The Worcester Foundation For Experimental Biology|Method of site-specific alteration of rna and production of encoded polypeptides| US5321131A|1990-03-08|1994-06-14|Hybridon, Inc.|Site-specific functionalization of oligodeoxynucleotides for non-radioactive labelling| US5470967A|1990-04-10|1995-11-28|The Dupont Merck Pharmaceutical Company|Oligonucleotide analogs with sulfamate linkages| US5612211A|1990-06-08|1997-03-18|New York University|Stimulation, production and culturing of hematopoietic progenitor cells by fibroblast growth factors| US5610289A|1990-07-27|1997-03-11|Isis Pharmaceuticals, Inc.|Backbone modified oligonucleotide analogues| US5677437A|1990-07-27|1997-10-14|Isis Pharmaceuticals, Inc.|Heteroatomic oligonucleoside linkages| US5608046A|1990-07-27|1997-03-04|Isis Pharmaceuticals, Inc.|Conjugated 4'-desmethyl nucleoside analog compounds| US5489677A|1990-07-27|1996-02-06|Isis Pharmaceuticals, Inc.|Oligonucleoside linkages containing adjacent oxygen and nitrogen atoms| US5602240A|1990-07-27|1997-02-11|Ciba Geigy Ag.|Backbone modified oligonucleotide analogs| US5541307A|1990-07-27|1996-07-30|Isis Pharmaceuticals, Inc.|Backbone modified oligonucleotide analogs and solid phase synthesis thereof| US5623070A|1990-07-27|1997-04-22|Isis Pharmaceuticals, Inc.|Heteroatomic oligonucleoside linkages| US5618704A|1990-07-27|1997-04-08|Isis Pharmacueticals, Inc.|Backbone-modified oligonucleotide analogs and preparation thereof through radical coupling| EP0541722B1|1990-08-03|1995-12-20|Sterling Winthrop Inc.|Compounds and methods for inhibiting gene expression| US5623065A|1990-08-13|1997-04-22|Isis Pharmaceuticals, Inc.|Gapped 2' modified oligonucleotides| US5177196A|1990-08-16|1993-01-05|Microprobe Corporation|Oligo and α-arabinofuranosyl precursors thereof| CA2089661C|1990-08-29|2007-04-03|Nils Lonberg|Transgenic non-human animals capable of producing heterologous antibodies| US5661016A|1990-08-29|1997-08-26|Genpharm International Inc.|Transgenic non-human animals capable of producing heterologous antibodies of various isotypes| US5625126A|1990-08-29|1997-04-29|Genpharm International, Inc.|Transgenic non-human animals for producing heterologous antibodies| US5633425A|1990-08-29|1997-05-27|Genpharm International, Inc.|Transgenic non-human animals capable of producing heterologous antibodies| US5545806A|1990-08-29|1996-08-13|Genpharm International, Inc.|Ransgenic non-human animals for producing heterologous antibodies| US5214134A|1990-09-12|1993-05-25|Sterling Winthrop Inc.|Process of linking nucleosides with a siloxane bridge| US5561225A|1990-09-19|1996-10-01|Southern Research Institute|Polynucleotide analogs containing sulfonate and sulfonamide internucleoside linkages| US5596086A|1990-09-20|1997-01-21|Gilead Sciences, Inc.|Modified internucleoside linkages having one nitrogen and two carbon atoms| CA2095153C|1990-10-31|2007-01-09|Lori F. Rafield|Genetic modification of endothelial cells| US5486359A|1990-11-16|1996-01-23|Osiris Therapeutics, Inc.|Human mesenchymal stem cells| US5714331A|1991-05-24|1998-02-03|Buchardt, Deceased; Ole|Peptide nucleic acids having enhanced binding affinity, sequence specificity and solubility| US5719262A|1993-11-22|1998-02-17|Buchardt, Deceased; Ole|Peptide nucleic acids having amino acid side chains| US5851832A|1991-07-08|1998-12-22|Neurospheres, Ltd.|In vitro growth and proliferation of multipotent neural stem cells and their progeny| US5571799A|1991-08-12|1996-11-05|Basco, Ltd.| oligoadenylate analogues useful as inhibitors of host-v5.-graft response| WO1993009220A1|1991-11-06|1993-05-13|Correa Paulo N|Cell culture medium| US5700922A|1991-12-24|1997-12-23|Isis Pharmaceuticals, Inc.|PNA-DNA-PNA chimeric macromolecules| CA2131385C|1992-03-04|2008-07-15|Bernhard O. Palsson|Methods, compositions and devices for maintaining and growing human stem or hematopoietic cells| US5792751A|1992-04-13|1998-08-11|Baylor College Of Medicine|Tranformation of cells associated with fluid spaces| US5633360A|1992-04-14|1997-05-27|Gilead Sciences, Inc.|Oligonucleotide analogs capable of passive cell membrane permeation| US5434257A|1992-06-01|1995-07-18|Gilead Sciences, Inc.|Binding compentent oligomers containing unsaturated 3',5' and 2',5' linkages| US5281521A|1992-07-20|1994-01-25|The Trustees Of The University Of Pennsylvania|Modified avidin-biotin technique| US5652355A|1992-07-23|1997-07-29|Worcester Foundation For Experimental Biology|Hybrid oligonucleotide phosphorothioates| US5320963A|1992-11-25|1994-06-14|National Research Council Of Canada|Bioreactor for the perfusion culture of cells| US5476925A|1993-02-01|1995-12-19|Northwestern University|Oligodeoxyribonucleotides including 3'-aminonucleoside-phosphoramidate linkages and terminal 3'-amino groups| CA2155515A1|1993-02-19|1994-09-01|Wendy Waegell|Expansion of stem cells in long term bone marrow cultures by neutralization of tgf-.beta.| US5654186A|1993-02-26|1997-08-05|The Picower Institute For Medical Research|Blood-borne mesenchymal cells| GB9304618D0|1993-03-06|1993-04-21|Ciba Geigy Ag|Chemical compounds| DE69407032T2|1993-03-31|1998-07-02|Sanofi Sa|OLIGONUCLEOTIDES WITH AMIDE CHAINS USE THE PHOSPHOESTER CHAINS| US5539082A|1993-04-26|1996-07-23|Nielsen; Peter E.|Peptide nucleic acids| US5342781A|1993-07-15|1994-08-30|Su Wei Wen W|External-loop perfusion air-lift bioreactor| US5378725A|1993-07-19|1995-01-03|The Arizona Board Of Regents|Inhibition of phosphatidylinositol 3-kinase with wortmannin and analogs thereof| WO1995004143A1|1993-07-28|1995-02-09|Fred Hutchinson Cancer Research Center|Creating novel hematopoietic cell lines by expressing altered retinoic acid receptors| US5504103A|1993-08-25|1996-04-02|Eli Lilly And Company|Inhibition of phosphatidylinositol 3-kinase with 17 β-hydroxywortmannin and analogs thereof| US6015686A|1993-09-15|2000-01-18|Chiron Viagene, Inc.|Eukaryotic layered vector initiation systems| US5599705A|1993-11-16|1997-02-04|Cameron; Robert B.|In vitro method for producing differentiated universally compatible mature human blood cells| CN1048254C|1993-12-09|2000-01-12|托马斯杰弗逊大学|Compounds and methods for site-directed mutations in eukaryotic cells| US6271363B1|1993-12-30|2001-08-07|President & Fellows Of Harvard College|Nucleic acids encoding hedgehog proteins| US6165747A|1993-12-30|2000-12-26|President & Fellows Of Harvard College|Nucleic acids encoding hedgehog proteins| US6261786B1|1993-12-30|2001-07-17|Imperial Cancer Res. Technology|Screening assays for hedgehog agonists and antagonists| US5789543A|1993-12-30|1998-08-04|President And Fellows Of Harvard College|Vertebrate embryonic pattern-inducing proteins and uses related thereto| WO1995021911A1|1994-02-09|1995-08-17|Unisyn Technologies, Inc.|High performance cell culture bioreactor and method| US5631219A|1994-03-08|1997-05-20|Somatogen, Inc.|Method of stimulating hematopoiesis with hemoglobin| AU687531B2|1994-03-11|1998-02-26|Baxter International Inc.|Flow-through bioreactor with grooves for cell retention| US5625050A|1994-03-31|1997-04-29|Amgen Inc.|Modified oligonucleotides and intermediates useful in nucleic acid therapeutics| US5489304A|1994-04-19|1996-02-06|Brigham & Women's Hospital|Method of skin regeneration using a collagen-glycosaminoglycan matrix and cultured epithelial autograft| US5480906A|1994-07-01|1996-01-02|Eli Lilly And Company|Stereochemical Wortmannin derivatives| US5807718A|1994-12-02|1998-09-15|The Scripps Research Institute|Enzymatic DNA molecules| US5770378A|1994-12-30|1998-06-23|Ligand Pharmaceuticals, Inc.|Tricyclic retinoids, methods for their production and use| US5736396A|1995-01-24|1998-04-07|Case Western Reserve University|Lineage-directed induction of human mesenchymal stem cell differentiation| US5837544A|1995-02-02|1998-11-17|Cell Genesys, Inc.|Method of inducing a cell to proliferate using a chimeric receptor comprising janus kinase| US5716616A|1995-03-28|1998-02-10|Thomas Jefferson University|Isolated stromal cells for treating diseases, disorders or conditions characterized by bone defects| US5733541A|1995-04-21|1998-03-31|The Regent Of The University Of Michigan|Hematopoietic cells: compositions and methods| US5674750A|1995-05-19|1997-10-07|T. Breeders|Continuous selective clonogenic expansion of relatively undifferentiated cells| US5925567A|1995-05-19|1999-07-20|T. Breeders, Inc.|Selective expansion of target cell populations| US5712154A|1995-06-07|1998-01-27|W.R. Grace & Co.-Conn.|Dual fiber bioreactor| US6680166B1|1995-06-07|2004-01-20|Claudy Jean Paul Mullon|Dual fiber bioreactor| US5728581A|1995-06-07|1998-03-17|Systemix, Inc.|Method of expanding hematopoietic stem cells, reagents and bioreactors for use therein| US6306575B1|1995-06-16|2001-10-23|Stemcell Technologies, Inc.|Methods for preparing enriched human hematopoietic cell preparations| US5730753A|1995-07-28|1998-03-24|Apls Co., Ltd.|Assembly for adjusting pricking depth of lancet| US5652356A|1995-08-17|1997-07-29|Hybridon, Inc.|Inverted chimeric and hybrid oligonucleotides| US6117850A|1995-08-28|2000-09-12|The Collaborative Group, Ltd.|Mobilization of peripheral blood precursor cells by β-glucan| US5776699A|1995-09-01|1998-07-07|Allergan, Inc.|Method of identifying negative hormone and/or antagonist activities| US6008204A|1995-09-01|1999-12-28|Allergan Sales, Inc.|Synthesis and use of retinoid compounds having negative hormone and/or antagonist activities| US5952345A|1995-09-01|1999-09-14|Allergan Sales, Inc.|Synthesis and use of retinoid compounds having negative hormone and/or antagonist activities| US5958954A|1995-09-01|1999-09-28|Allergan Sales, Inc.|Synthesis and use of retinoid compounds having negative hormone and/or antagonist activities| US5877207A|1996-03-11|1999-03-02|Allergan Sales, Inc.|Synthesis and use of retinoid compounds having negative hormone and/or antagonist activities| US6329169B1|1995-09-29|2001-12-11|Human Genome Sciences, Inc.|Nucleic acid molecules encoding cytostatin II| FR2739777B1|1995-10-11|1997-11-14|Cird Galderma|LIGAND ANTAGONIST RAR-GAMMA OR AGONIST RAR-ALPHA AS AN APOPTOSIS INHIBITOR| US6613565B1|1996-03-01|2003-09-02|Imclone Systems Incorporated|Use of delta-like protein to inhibit the differentiation of stem cells| AU2260097A|1996-03-12|1997-10-01|Life Technologies, Inc.|Hematopoietic cell culture nutrient supplement| US6043092A|1996-03-18|2000-03-28|University Of Pittsburgh|Cell culture media for mammalian cells| US5945309A|1996-03-19|1999-08-31|Human Genome Sciences, Inc.|Cytostatin III nucleic acids encoding| TW442459B|1996-04-15|2001-06-23|Hoffmann La Roche|Retinoid compounds, pharmaceutical composition comprising same and process for the preparation thereof| US6455678B1|1996-04-26|2002-09-24|Amcell Corporation|Human hematopoietic stem and progenitor cell antigen| JP2000508919A|1996-04-29|2000-07-18|リューベン・リサーチ・アンド・デベロップメント・ベー・ゼット・ウェー|Pluripotent rabbit embryonic stem cell line and its use in developing chimeric rabbits| US5851984A|1996-08-16|1998-12-22|Genentech, Inc.|Method of enhancing proliferation or differentiation of hematopoietic stem cells using Wnt polypeptides| CA2263817A1|1996-08-28|1998-03-05|Institut National De La Sante Et De La Recherche Medicale |Therapeutic combinations of rar antagonists and rxr agonists and use thereof| US5945337A|1996-10-18|1999-08-31|Quality Biological, Inc.|Method for culturing CD34+ cells in a serum-free medium| EP0973530A4|1996-12-10|2002-01-16|Hadasit Med Res Service|Serum-derived factor inducing cell differentiation and medical uses thereof| US6335195B1|1997-01-28|2002-01-01|Maret Corporation|Method for promoting hematopoietic and mesenchymal cell proliferation and differentiation| US6248587B1|1997-11-26|2001-06-19|University Of Southern Cailfornia|Method for promoting mesenchymal stem and lineage-specific cell proliferation| US6294330B1|1997-01-31|2001-09-25|Odyssey Pharmaceuticals Inc.|Protein fragment complementation assays for the detection of biological or drug interactions| CA2196496A1|1997-01-31|1998-07-31|Stephen William Watson Michnick|Protein fragment complementation assay for the detection of protein-protein interactions| US20030215445A1|1997-05-23|2003-11-20|Ginette Serrero|Methods and compositions for inhibiting the growth of hematopoietic malignant cells| US6372473B1|1997-05-28|2002-04-16|Human Genome Sciences, Inc.|Tissue plasminogen activator-like protease| CA2295474A1|1997-07-08|1999-01-21|Human Genome Sciences, Inc.|123 human secreted proteins| IT1293827B1|1997-08-07|1999-03-10|Dompe Spa|METHOD FOR EX VIVO EXPANSION OF HEMATOPOIETIC STEM CELLS| US6218128B1|1997-09-12|2001-04-17|Allergan Sales, Inc.|Methods of identifying compounds having nuclear receptor negative hormone and/or antagonist activities| US6440734B1|1998-09-25|2002-08-27|Cytomatrix, Llc|Methods and devices for the long-term culture of hematopoietic progenitor cells| FR2769491B1|1997-10-15|2000-01-07|Patrick Sangouard|ADJUSTABLE ARTIFICIAL SPHINCTER WITH MAGNETIC CONTROL| KR20040031107A|1997-11-12|2004-04-09|에프. 호프만-라 로슈 아게|Treatment of t-helper cell type 2 mediated immune diseases with retinoid antagonists| WO2000018885A1|1998-09-29|2000-04-06|Gamida Cell Ltd.|Methods of controlling proliferation and differentiation of stem and progenitor cells| US6962698B1|1998-02-17|2005-11-08|Gamida Cell Ltd.|Methods of controlling proliferation and differentiation of stem and progenitor cells| WO1999040783A1|1998-02-17|1999-08-19|Gamida Cell Ltd.|Method of controlling proliferation and differentiation of stem and progenitor cells| US6886568B2|1998-04-08|2005-05-03|The Johns Hopkins University|Method for fabricating cell-containing implants| US6413773B1|1998-06-01|2002-07-02|The Regents Of The University Of California|Phosphatidylinositol 3-kinase inhibitors as stimulators of endocrine differentiation| US6030836A|1998-06-08|2000-02-29|Osiris Therapeutics, Inc.|Vitro maintenance of hematopoietic stem cells| DK1084230T3|1998-06-08|2008-03-03|Osiris Therapeutics Inc|Regulation of hematopoietic stem cell differentiation using human mesenchymal stem cells| US6284540B1|1998-09-29|2001-09-04|Washington University|Artemin, a novel neurotrophic factor| CA2254429A1|1998-11-19|2000-05-19|Tully Michael Underhill|Compositions for promoting chondrogenesis| AU776150B2|1999-01-28|2004-08-26|Medical College Of Georgia Research Institute, Inc.|Composition and method for and attenuation of gene expression using double stranded RNA| JP4523169B2|1999-02-04|2010-08-11|プルリステムリミテッド|Method and apparatus for maintaining and increasing hematopoietic stem cells and / or progenitor cells| CA2372053C|1999-04-28|2008-09-02|Board Of Regents, The University Of Texas System|Compositions and methods for cancer treatment by selectively inhibiting vegf| DE19919242A1|1999-04-28|2000-11-02|Creavis Tech & Innovation Gmbh|Modular cell support systems for three-dimensional cell growth| WO2000073421A2|1999-06-02|2000-12-07|Lifebank Services, L.L.C.|Methods of isolation, cryopreservation, and therapeutic use of human amniotic epithelial cells| US6652872B2|1999-07-06|2003-11-25|Ramat At Tel Aviv University Ltd.|Scaffold formed of tissue treated to eliminate cellular and cytosolic elements| US20020159984A1|1999-11-12|2002-10-31|Quality Biological, Inc.|Cultivation of cells for long term engraftment| US6541249B2|1999-12-22|2003-04-01|Human Genome Sciences, Inc.|Immortalized human stromal cell lines| US6667176B1|2000-01-11|2003-12-23|Geron Corporation|cDNA libraries reflecting gene expression during growth and differentiation of human pluripotent stem cells| US6303374B1|2000-01-18|2001-10-16|Isis Pharmaceuticals Inc.|Antisense modulation of caspase 3 expression| AU6825501A|2000-06-05|2001-12-17|Burnham Inst|Methods of differentiating and protecting cells by modulating the p38/mef2 pathway| CA2417356A1|2000-08-01|2002-02-07|Yissum Research Development Company|Directed differentiation of embryonic cells| US7198940B2|2000-10-25|2007-04-03|Shot Hardware Optimization Technology, Inc.|Bioreactor apparatus and cell culturing system| ES2171131B1|2000-11-10|2003-12-16|Idebio S L|QUITOSANE BASED BIOLOGICAL PESTICIDE| US6642019B1|2000-11-22|2003-11-04|Synthecan, Inc.|Vessel, preferably spherical or oblate spherical for growing or culturing cells, cellular aggregates, tissues and organoids and methods for using same| JP2004528021A|2001-02-14|2004-09-16|アンスロジェネシスコーポレーション|Postpartum mammalian placenta, its use and placental stem cells derived therefrom| US7202080B2|2001-03-29|2007-04-10|Ixion Biotechnology, Inc.|Method for transdifferentiation of non-pancreatic stem cells to the pancreatic differentiation pathway| CN101172158A|2001-05-11|2008-05-07|得克萨斯州立大学董事会|Anti-CD26 monoclonal antibodies as therapy for diseases associated with cells expressing CD26| US20030017134A1|2001-06-19|2003-01-23|Technion Research And Development Foundation Ltd.|Methods and pharmaceutical compositions for immune deception, particularly useful in the treatment of cancer| DE10131388B4|2001-06-28|2004-07-08|Infineon Technologies Ag|Integrated dynamic memory and method for operating the same| CA2351156A1|2001-07-04|2003-01-04|Peter W. Zandstra|A bioprocess for the generation of pluripotent cell derived cells and tissues| KR20040023724A|2001-08-07|2004-03-18|기린 비루 가부시키가이샤|Process for Preparing Hematopoietic Stem Cells| WO2003035618A2|2001-10-24|2003-05-01|Iconix Pharmaceuticals, Inc.|Modulators of phosphoinositide 3-kinase| US6887401B2|2001-11-05|2005-05-03|Essilor International Compagnie General D'optique|Method for making transparent polythiourethane substrates in particular optical substrates| AUPR892501A0|2001-11-16|2001-12-13|Peter Maccallum Cancer Institute, The|Method of enhancing self renewal of stem cells and uses thereof| US7514075B2|2001-12-07|2009-04-07|Cytori Therapeutics, Inc.|Systems and methods for separating and concentrating adipose derived stem cells from tissue| US20050095228A1|2001-12-07|2005-05-05|Fraser John K.|Methods of using regenerative cells in the treatment of peripheral vascular disease and related disorders| DE10162814B4|2001-12-19|2005-06-23|Henkel Kgaa|Reconstructed hair follicle model| AU2003214614B2|2002-03-18|2008-11-20|Gamida-Cell Ltd.|Methods of inducing differentiation in ex vivo expanded stem cells| MXPA05001992A|2002-08-19|2005-08-03|Gamida Cell Ltd|Ex-vivo expansion of hematopoietic stem cell populations in mononuclear cell cultures.| IL152904D0|2002-01-24|2003-06-24|Gamida Cell Ltd|Utilization of retinoid and vitamin d receptor antagonists for expansion of renewable stem cell populations| US20050054097A1|2002-11-17|2005-03-10|Tony Peled|EX-VIVO expansion of hematopoietic system cell populations in mononuclear cell cultures| WO2003062404A1|2002-01-25|2003-07-31|Gamida-Cell Ltd.|Methods of expanding stem and progenitor cells and expanded cell populations obtained thereby| AR038703A1|2002-02-28|2005-01-26|Novartis Ag|DERIVATIVES OF 5-PHENYLTIAZOL AND USE AS AN INHIBITOR OF QUINASA P I 3| EP1601759A4|2003-03-07|2007-02-14|Gamida Cell Ltd|Expansion of renewable stem cell populations using modulators of pi 3-kinase| US7498171B2|2002-04-12|2009-03-03|Anthrogenesis Corporation|Modulation of stem and progenitor cell differentiation, assays, and uses thereof| US7247477B2|2002-04-16|2007-07-24|Technion Research & Development Foundation Ltd.|Methods for the in-vitro identification, isolation and differentiation of vasculogenic progenitor cells| US7470538B2|2002-12-05|2008-12-30|Case Western Reserve University|Cell-based therapies for ischemia| US20040247574A1|2003-05-27|2004-12-09|Christopherson Kent W.|Methods for enhancing stem cell engraftment during transplantation| IL161903D0|2003-07-17|2005-11-20|Gamida Cell Ltd|Ex vivo progenitor and stem cell expansion for usein the treatment of disease of endodermally- deri ved organs| WO2005007799A2|2003-07-17|2005-01-27|Gamida-Cell Ltd.|Methods for ex-vivo expanding stem/progenitor cells| US7435596B2|2004-11-04|2008-10-14|St. Jude Children's Research Hospital, Inc.|Modified cell line and method for expansion of NK cell| WO2005085422A1|2004-02-27|2005-09-15|Michigan State University|Adult stem cells and uses thereof| CN1950498A|2004-03-10|2007-04-18|加利福尼亚大学董事会|Compositions and methods for growth of embryonic stem cells| WO2006030442A2|2004-09-16|2006-03-23|Gamida-Cell Ltd.|Methods of ex vivo progenitor and stem cell expansion by co-culture with mesenchymal cells| CA2589034C|2004-11-02|2016-05-31|The Government Of The United States Of America, As Represented By The Secretary, Department Of Health & Human Services|Compositions and methods for treating hyperproliferative disorders| EP1859025A4|2005-01-31|2009-07-22|Cognate Therapeutics Inc|Adipose derived adult stromal cells exhibiting characteristics of endothelial cells| KR20090043559A|2006-08-15|2009-05-06|에이전시 포 사이언스, 테크놀로지 앤드 리서치|Mesenchymal stem cell conditioned medium| US8846393B2|2005-11-29|2014-09-30|Gamida-Cell Ltd.|Methods of improving stem cell homing and engraftment| CA2632288A1|2005-11-29|2007-06-07|Gamida-Cell Ltd.|Methods of improving stem cell homing and engraftment| WO2007066338A1|2005-12-08|2007-06-14|Ramot At Tel Aviv University Ltd.|Isolated oligodendrocyte-like cells and populations comprising same for the treatment of cns diseases| JP2010509316A|2006-11-09|2010-03-25|ガミダセルリミテッド|Use of ex vivo cultured hematopoietic cells to treat peripheral vascular disease| KR100995835B1|2007-12-18|2010-11-23|동국대학교 산학협력단|Method for preparation of hepatocytes with liver-specific functions using mesenchymal stem cells and umbilical cord-derived hepatocytes prepared thereby| US20090257987A1|2008-04-10|2009-10-15|Ramot At Tel Aviv University Ltd.|Method of generating dopamine-secreting cells| US8062632B2|2008-06-23|2011-11-22|National Yang-Ming University|Systems and methods for making hepatocytes from extrahepatic somatic stem cells and use thereof| TWI397585B|2008-07-16|2013-06-01|Univ Nat Taiwan|Cell culture method and application thereof| GB0818725D0|2008-10-13|2008-11-19|Habib Nagy A|Pharmaceutical composition| PE20100362A1|2008-10-30|2010-05-27|Irm Llc|PURINE DERIVATIVES THAT EXPAND HEMATOPOYETIC STEM CELLS| JP5606008B2|2009-04-13|2014-10-15|三菱重工業株式会社|Method for culturing bone marrow stromal cells and mesenchymal stem cells, method for producing transplanted cells for treatment of central nervous system diseases| EP2333047A1|2009-12-09|2011-06-15|Fresenius Medical Care Deutschland GmbH|Adult stem cell derived conditioned medium and/or adult stem cells for use in the therapeutic treatment of a tumor disease| WO2011080740A1|2009-12-29|2011-07-07|Gamida-Cell Ltd.|Methods for enhancing natural killer cell proliferation and activity| US8785395B2|2010-05-03|2014-07-22|The Texas A & M University System|Adult stem cells/progenitor cells and stem cell proteins for treatment of eye injuries and diseases| EP2814951B1|2012-02-13|2019-04-03|Gamida-Cell Ltd.|Culturing of mesenchymal stem cells| US9567569B2|2012-07-23|2017-02-14|Gamida Cell Ltd.|Methods of culturing and expanding mesenchymal stem cells| US9175266B2|2012-07-23|2015-11-03|Gamida Cell Ltd.|Enhancement of natural killer cell proliferation and activity|US8846393B2|2005-11-29|2014-09-30|Gamida-Cell Ltd.|Methods of improving stem cell homing and engraftment| AU2012262675B2|2011-06-03|2016-05-19|Mesoblast, Inc|Methods of treating or preventing neurological diseases| EA029577B1|2011-07-06|2018-04-30|Селл Терапи Лимитед|Progenitor cells of mesodermal lineage| EP2814951B1|2012-02-13|2019-04-03|Gamida-Cell Ltd.|Culturing of mesenchymal stem cells| US9175266B2|2012-07-23|2015-11-03|Gamida Cell Ltd.|Enhancement of natural killercell proliferation and activity| US9567569B2|2012-07-23|2017-02-14|Gamida Cell Ltd.|Methods of culturing and expanding mesenchymal stem cells| KR101583569B1|2013-05-15|2016-01-08|라정찬|Composition for Intravenous Administration Comprising Stem Cells| KR101548956B1|2013-09-05|2015-09-01|메디포스트|Method for culturing mesenchymal stem cells according to cell size| CN113959928A|2013-11-04|2022-01-21|贝克顿·迪金森公司|Immunomodulatory potential of pluripotent stromal cellpopulations| US10458985B2|2014-04-11|2019-10-29|The Administrators Of The Tulane Educational Fund|Cell-surface marker of early MSC aging| US20170112879A1|2014-06-09|2017-04-27|University Of Washington|Methods of protection against ischemia reperfusion injury| US10493105B2|2014-09-11|2019-12-03|Taiwan Mitochondrion Applied Technology Co., Ltd|Isolated adipose-derived mesenchymal stem cells treated with angelica extract or butylidenephthalide, and wherein the cells have an increased mitochondrial membrane potential and a decreased level of IL-8, and methods for treating parkinson's disease| JP6898706B2|2015-01-05|2021-07-07|ナショナル ヤン−ミン ユニバーシティ|Mesenchymal stem cells to treat liver disease| CN108138137A|2015-08-12|2018-06-08|车比奥泰有限公司|The adhesion stem cell in the umbilical cord source of enhancing, Its Preparation Method And Use| US10959425B2|2016-04-29|2021-03-30|Hope Biosciences, Llc|Method of banking stem cells| US10513689B2|2016-04-29|2019-12-24|Hope Biosciences, Llc|Culture media for multipotent stem cells| US10894947B1|2016-04-29|2021-01-19|Hope Biosciences, Llc|Method for generating protein rich conditioned medium| US11111480B2|2016-04-29|2021-09-07|Hope Biosctences, Llc|Culture media for multipotent stem cells| CN106701671A|2016-11-14|2017-05-24|天津市康婷生物工程有限公司|Culture system beneficial for mesenchymal stem cells to be applied in bone regeneration| US20200085881A1|2016-12-16|2020-03-19|Osaka Air Machine Service, Ltd.|Tissue healing agent| CN106520691A|2016-12-30|2017-03-22|潍坊医学院|Separation and culture method of skin mesenchymal stem cells| EP3568143A1|2017-01-11|2019-11-20|Paracelsus Medizinische Privatuniversität Salzburg - Privatstiftung|Mesenchymal stem cell-derived extracellular vesicles and their medical use| JP6974360B2|2017-01-31|2021-12-01|オリエンタル酵母工業株式会社|Pluripotent stem cell proliferation promoter| CN107083357B|2017-06-16|2021-04-02|四川农业大学|Method for inducing and culturing mesenchymal stem cells| KR102185697B1|2017-10-20|2020-12-03|본 테라퓨틱스 소시에테아노님|Mesenchymal stem cell differentiation method| CN108220231A|2018-01-23|2018-06-29|广州资生生物科技有限公司|A kind of stem cell media and its preparation method and application| CN108220161A|2018-01-24|2018-06-29|北京臻溪谷医学研究中心(有限合伙)|A kind of many cells prepare laboratory placement scheme| EP3770250A4|2018-03-22|2021-11-24|Oriental Yeast Co., Ltd.|Differentiation promoter for pluripotent stem cells| CN109810943A|2019-03-21|2019-05-28|河南科技大学|One boar muscle-derived mescenchymal stem cell isolation medium and isolated culture method| US10881693B2|2019-04-09|2021-01-05|Combangio, Inc.|Processes for making and using a mesenchymal stem cell derived secretome|
法律状态:
2020-11-03| B15I| Others concerning applications: loss of priority|Free format text: PERDA DAS PRIORIDADES US 61/597,899 E US 61/597,909, CONFORME AS DISPOSICOES PREVISTAS NA LEI 9.279 DE 14/05/1996 (LPI) ART. 167O E NO ART. 29 DA RESOLUCAO INPI-PR 77/2013, POR NAO ATENDEREM AO DISPOSTO NO ART. 2 DA RESOLUCAO INPI-PR 179/2017, POIS NAO FORAM APRESENTADAS TEMPESTIVAMENTE A CESSAO DAS REFERIDAS PRIORIDADES, QUE POSSUEM DEPOSITANTE DIFERENTE DO DEPOSITANTE DA FASE NACIONAL. | 2020-11-10| B06F| Objections, documents and/or translations needed after an examination request according [chapter 6.6 patent gazette]| 2020-11-17| B07D| Technical examination (opinion) related to article 229 of industrial property law [chapter 7.4 patent gazette]|Free format text: DE ACORDO COM O ARTIGO 229-C DA LEI NO 10196/2001, QUE MODIFICOU A LEI NO 9279/96, A CONCESSAO DA PATENTE ESTA CONDICIONADA A ANUENCIA PREVIA DA ANVISA. CONSIDERANDO A APROVACAO DOS TERMOS DO PARECER NO 337/PGF/EA/2010, BEM COMO A PORTARIA INTERMINISTERIAL NO 1065 DE 24/05/2012, ENCAMINHA-SE O PRESENTE PEDIDO PARA AS PROVIDENCIAS CABIVEIS. | 2020-12-29| B07E| Notification of approval relating to section 229 industrial property law [chapter 7.5 patent gazette]| 2021-01-26| B06U| Preliminary requirement: requests with searches performed by other patent offices: procedure suspended [chapter 6.21 patent gazette]| 2021-03-02| B08F| Application dismissed because of non-payment of annual fees [chapter 8.6 patent gazette]|Free format text: REFERENTE A 8A ANUIDADE. | 2021-05-11| B11B| Dismissal acc. art. 36, par 1 of ipl - no reply within 90 days to fullfil the necessary requirements| 2021-12-07| B350| Update of information on the portal [chapter 15.35 patent gazette]|
优先权:
[返回顶部]
申请号 | 申请日 | 专利标题 US201261597899P| true| 2012-02-13|2012-02-13| US201261597909P| true| 2012-02-13|2012-02-13| US61/597,909|2012-02-13| US61/597,899|2012-02-13| PCT/IL2013/050136|WO2013121426A1|2012-02-13|2013-02-13|Culturing of mesenchymal stem cells| 相关专利
Sulfonates, polymers, resist compositions and patterning process
Washing machine
Washing machine
Device for fixture finishing and tension adjusting of membrane
Structure for Equipping Band in a Plane Cathode Ray Tube
Process for preparation of 7 alpha-carboxyl 9, 11-epoxy steroids and intermediates useful therein an
国家/地区
|