Methods for reprogramming cells and uses thereof

Information

  • Patent Grant
  • 11795439
  • Patent Number
    11,795,439
  • Date Filed
    Friday, January 24, 2020
    4 years ago
  • Date Issued
    Tuesday, October 24, 2023
    7 months ago
Abstract
A method of obtaining a pancreatic multipotent or unipotent cell including providing a cell of a first type which is not a pancreatic multipotent or unipotent cell; contacting the cell of a first type with an agent capable of remodeling the chromatin and/or DNA of the cell; transiently increasing expression of at least one pancreatic multipotent or unipotent gene regulator in the cell of a first type, to a level at which the at least one pancreatic multipotent or unipotent gene regulator is capable of driving transformation of the cell of a first type into the pancreatic multipotent or unipotent cell; and placing or maintaining the cell in a pancreatic cell culture medium and maintaining intracellular levels of the at least one pancreatic multipotent or unipotent gene regulator for a sufficient period of time to allow a pancreatic multipotent or unipotent cell to be obtained.
Description
FIELD OF THE INVENTION

The present invention relates to the field of eukaryotic cell reprogramming, and particularly to cell dedifferentiation. The invention is also concerned with methods of generating stable Neural Stem-Like Cells (NSLCs) from human somatic cells (and other cells) and the use of the cells so generated in human therapy.


REFERENCE TO SEQUENCE LISTING

A Sequence Listing submitted as an ASCII text file via EFS-Web is hereby incorporated by reference in accordance with 35 U.S.C. § 1.52(e). The name of the ASCII text file for the Sequence Listing is 32792254_1.txt, the date of creation of the ASCII text file is May 11, 2020, and the size of the ASCII text file is 1.54 KB.


BACKGROUND OF THE INVENTION

Cell Reprogramming


There is a desire in the medical, scientific, and diagnostic fields to reprogram an easily obtainable cell into a cell that is generally harder to obtain, or to reprogram a cell to have new or different functionalities, without fusing or exchanging material with an oocyte or another stem cell.


According to a first mechanism, a stem cell can naturally divide or differentiate into another stem cell, progenitor, precursor, or somatic cell. According to a second mechanism, somatic cell can sometimes transiently change its phenotype or express certain markers when placed in certain conditions, and then revert back when placed back into the original conditions. According to a second mechanism, the phenotype of many cells can be changed through forced expression of certain genes (for example, stably transfecting the c-myc gene into fibroblasts turns them into immortal cells having neuroprogenitor characteristics), however once this forced gene expression is removed, the cells slowly revert back to their original state. Therefore, none of the three above mechanisms should be considered true reprogramming: the first is considered natural differentiation which is part of a cell program that is already in place (going from a more undifferentiated to a more differentiated state), the second is a transient phenotypical change, and the third is a constantly forced cell type. A true stem cell: (i) self-renews almost ‘indefinitely’ (for significantly longer than a somatic cell), (ii) is not a cancerous cell, (iii) is not artificially maintained by forced gene expression or similar means (must also be able to be maintained in standard stem cell media), (iv) can differentiate to progenitor, precursor, somatic or other more differentiated cell type (of the same lineage), and (v) has all the characteristics of a stem cell and not just certain markers or gene expression or morphological appearance.


Despite the numerous scientific and patent publications claiming successful reprogramming or dedifferentiation, generally into a stem cell, almost all of these publications do not disclose true reprogramming because they fall under one of the mechanisms mentioned above. For instance, Bhasin (WO2010/088735), Cifarelli et al. (US2010/0003223), Kremer et al. (US2004/0009595), and Winnier et al. (US2010/0047908) all refer to reprogramming, dedifferentiation, and/or obtained stem cells (or progenitors) as phenotypical cell changes based only on a change in cell surface markers after culture in different media with supplements, with no evidence of true reprogramming or an actual stem cell (non-cancerous self-renewal with stem cells markers and no differentiation markers). The same is true for Benneti (WO2009/079007) who used increased expression of Oct4 and Sox2. Others, such as Akamatsu et al. (WO2010/052904) and You et al. (WO2007/097494, US2009/0246870), refer to having made stem cells, but these came about through constant artificial gene induction delivered by retrovirus (similar to cMyc) with no evidence of true stem cells that are not immortal/tumorigenic, and stable instead of transient. Others, such as Chen et al. (US2005/0176707) and You et al. (US2009/0227023), have made “multipotent cells”, but not stem cells. In addition these alledged multipotent cells were not stable (in the case of You et al. the cells could not even proliferate) and both used constant media supplements and conditions to force the phenotypical change. Yet others, such as Oliveri et al. (WO2009/018832) and Zahner et al. (US2002/0136709), have claimed the making of pluripotent, totipotent, multipotent, and/or unipotent cells automatically through genome-wide DNA demethylation and histone acetylation, but with no evidence of a stable, non-cancerous, true cell line.


True reprogramming appears to have been achieved with induced pluripotent stem cells (iPS cells) created independently by Yamanaka's group (Takahashi et al., 2007) and Thomson's group (Yu et al., 2007), and potentially by others before them, and although many of these cells were later found to be cancerous, some of them were not. These cells can be induced by true reprogramming since it was later shown that they can also be induced by non-gene integrating transient transfection (Soldner et al., 2009; Woltjen et al., 2009; Yu et al., 2009) as well as by RNA (Warren et al., 2010) or protein (Kim et al., 2009; Zhou et al., 2009) alone or by small molecules (Lyssiotis et al., 2009), and by similar methods. However, these cells are essentially identical to embryonic stem cells and have the same problems of uncontrolled growth, teratoma formation, and potential tumor formation.


A more desirable option is to have multipotent stem cells or pluripotent-like cells whose lineage and differentiation potential is more restricted so that they do not readily form teratomas and uncontrolled growth. There is thus a need for methods of creating multipotent stem cells, multipotent stem-like cells, and stem-like cells and method of reprogramming or transforming easily obtainable cells to highly desirable multipotent stem cells, multipotent stem-like cells, and stem-like cells.


Neural Stem-Like Cells (NSLC)


Repairing the central nervous system (CNS) is one of the frontiers that medical science has yet to conquer. Conditions such as Alzheimer's disease, Parkinson's disease, and stroke can have devastating consequences for those who are afflicted. A central hope for these conditions is to develop cell populations that can reconstitute the neural network, and bring the functions of the nervous system back in line. For this reason, there is a great deal of evolving interest in neural stem and progenitor cells. Up until the present time, it was generally thought that multipotent neural progenitor cells commit early in the differentiation pathway to either neural restricted cells or glia restricted cells.


Neural stem cells have promise for tissue regeneration from disease or injury; however, such therapies will require precise control over cell function to create the necessary cell types. There is not yet a complete understanding of the mechanisms that regulate cell proliferation and differentiation, and it is thus difficult to fully explore the plasticity of neural stem cell population derived from any given region of the brain or developing fetus.


The CNS, traditionally believed to have limited regenerative capabilities, retains a limited number of neural stem cells in adulthood, particularly in the dentate gyrus of the hippocampus and the subventricular zone that replenishes olfactory bulb neurons (Singec I et al., 2007; Zielton R, 2008). The availability of precursor cells is a key prerequisite for a transplant-based repair of defects in the mature nervous system. Thus, donor cells for neural transplants are largely derived from the fetal brain. This creates enormous ethical problems, in addition to immuno-rejection, and it is questionable whether such an approach can be used for the treatment of a large number of patients since neural stem cells can lose some of their potency with each cell division.


Neural stem cells provide promising therapeutic potential for cell-replacement therapies in neurodegenerative disease (Mimeault et al., 2007). To date, numerous therapeutic transplantations have been performed exploiting various types of human fetal tissue as the source of donor material. However, ethical and practical considerations and their inaccessibility limit the availability as a cell source for transplantation therapies (Ninomiy M et al., 2006).


To overcome barriers and limitations to the derivation of patient specific cells, one approach has been to use skin cells and inducing the trans-differentiation to neural stem cells and/or to neurons (Levesque et al., 2000). Transdifferentiation has been receiving increasing attention during the past years, and trans-differentiation of mammalian cells has been achieved in co-culture or by manipulation of cell culture conditions. Alteration of cell fate can be induced artificially in vitro by treatment of cell cultures with microfilament inhibitors (Shea et al., 1990), hormones (Yeomans et al., 1976), and Calcium-ionophores (Shea, 1990; Sato et al., 1991). Mammalian epithelial cells can be induced to acquire muscle-like shape and function (Paterson and Rudland, 1985), pancreatic exocrine duct cells can acquire an insulin-secreting endocrine phenotype (Bouwens, 1998a, b), and bone marrow stem cells can be differentiated into liver cells (Theise et al., 2000) and into neuronal cells (Woodbury et al., 2000). Other such as Page et al. (US 2003/0059939) have transdifferentiated somatic cells to neuronal cells by culturing somatic cells in the presence of cytoskeletal, acetylation, and methylation inhibitors, but after withdrawal of the priming agent, neuron morphology and established synapses last for not much than a few weeks in vitro, and complete conversion to a fully functional and stable type of neuron has never been demonstrated. These are thus transient cell phenotypes. Complete conversion to a fully functional and stable type of neuroprogenitor or neural stem cell has also never been demonstrated. Acquisition of a stable phenotype following transdifferentiation has been one of the major challenges facing the field.


Thus, there is a need in the biomedical field for stable, potent, and preferably autologouos neural stem cells, neural progenitor cells, as well as neurons and glial cells for use in the treatment of various neurological disorders and diseases. The same is true for many other types of cells. Recently, evidence have been obtained that genes of the basic Helix-Loop-Helix (bHLH) class are important regulators of several steps in neural lineage development, and over-expression of several neurogenic bHLH factors results in conversion of non-determined ectoderm into neuronal tissue. Proneural bHLH proteins control the differentiation into progenitor cells and their progression through the neurogenic program throughout the nervous system (Bertrand et al., 2002). MASH1, NeuroD, NeuroD2, MATH1-3, and Neurogenin 1-3 are bHLH transcription factors expressed during mammalian neuronal determination and differentiation (Johnson et al., 1990; Takebyashi et al., 1997; McCormick et al., 1996; Akazawa et al., 1995). Targeted disruptions of MASH1, Ngn1, Ngn2 or NeuroD in mice lead to the loss of specific subsets of neurons (Guillemot et al., 1993; Fode et al., 1998; Miyata et al., 1999).


U.S. Pat. No. 6,087,168 (Levesque et al.) describes a method for converting or transdifferentiating epidermal basal cells into viable neurons. In one example, this method comprises the transfection of the epidermal cells with one or more expression vector(s) containing at least one cDNA encoding for a neurogenic transcription factor responsible for neural differentiation. Suitable cDNAs include: basic-helix-loop-helix activators, such as NeuroD1, NeuroD2, ASH1, and zinc-finger type activators, such as Zic3, and MyT1. The transfection step was followed by adding at least one antisense oligonucleotide known to suppress neuronal differentiation to the growth medium, such as the human MSX1 gene and/or the human HES1 gene (or non-human, homologous counterparts). Finally, the transfected cells were grown in the presence of a retinoid and a least one neurotrophin or cytokine, such as brain derived neurotrophic factor (BDNF), nerve growth factor (NGF), neurotrophin 3 (NT-3), or neurotrophin 4 (NT-4). This technology yields 26% of neuronal cells; however, neither functionality nor stability of these cells was established. In addition, neural stem cells or neuroprogenitor cells are not produced according to this method.


A later process (Levesque et al., 2005; U.S. Pat. No. 6,949,380) mentions the conversion of the epidermal basal cell into a neural progenitor, neuronal, or glial cell by exposing the epidermal basal cell to an antagonist of bone morphogenetic protein (BMP) and growing the cell in the presence of at least one antisense oligonucleotide comprising a segment of a MSX 1 gene and/or HES1 gene. However, there is no evidence or examples that any neural progenitors or glial cells were produced according to this method, let alone any details or evidence that morphological, physiological or immunological features of neuronal cells was achieved. In addition, since there is also no information on functionality, stability, expansion, and yield about the cells which may or may not have been produced, it is possible that these cells actually are skin-derived precursor cells (Fernandes et al., 2004) that have been differentiated into neuronal cells.


In view of the above, there is thus a need for stable, potent, and preferably autologouos neural stem cells, neural progenitor cells, neurons and glial cells, as well as other types of cells, stem cells and progenitor cells. There is also a need for methods that could result in true cell dedifferentiation and cell reprogramming.


The present invention addresses these needs and provides various types of stem-like and progenitor-like cells and cells derived or differentiated from these stem-like or progenitor-like cells, as well as methods that can result in true cell dedifferentiation and cell reprogramming.


Additional features of the invention will be apparent from a review of the disclosure, figures and description of the invention herein.


SUMMARY OF THE INVENTION

The present invention relates to stem-like and progenitor-like cells and cells derived or differentiated from these stem-like or progenitor-like cells. The invention further relates to methods for cell dedifferentiation and cell reprogramming. The invention further features compositions and methods that are useful for reprogramming cells and related therapeutic compositions and methods.


One particular aspect relates to the development of a technology to reprogram a somatic cell or non-neuronal cell to a cell having one or more morphological physiological, and/or immunological features of a neural stem cell and which possess the capacity to differentiate along neuronal and glial lineages. According to some embodiments, the invention is more particularly concerned with methods of generating stable Neural Stem-Like Cells (NSLCs) from human somatic cells, human progenitor cells and/or of human stem cells, as well as cells, cell lines and tissues obtained by using such methods.


The invention further relates to compositions and methods to induce de-differentiation of human somatic cells into Neural Stem-Like Cells that express neural stem cell specific markers. According to the present invention it is possible to effect the conversion of cells to various types of differentiated neuronal cells that can be created from a single cell type taken from an individual donor and then reprogrammed and transplanted into the same individual. Upon induction cells according to the invention express neural stem-cell specific markers and become Neural Stem-Like cells.


According to one particular aspect, the invention relates to a method of transforming a cell of a first type to a desired cell of a different type. The comprises i) obtaining a cell of a first type; ii) transiently increasing in the cell of a first type intracellular levels of at least one reprogramming agent, whereby the transient increase induces direct or indirect endogenous expression of at least one gene regulator; iii) placing the cell in conditions for supporting the growth and/or the transformation of the desired cell and maintaining intracellular levels of the at least one reprogramming agent for a sufficient period of time to allow stable expression of the at least one gene regulator in absence of the reprogramming agent; and iv) maintaining the cell in culture conditions supporting the growth and/or the transformation of the desired cell. Such conditions are maintained for a sufficient period of time to allow a stable expression of a plurality of secondary genes. According to the invention the expression of one or more of the secondary genes is characteristic of phenotypical and functional properties of the desired cell while being not characteristic of phenotypical and functional properties of an embryonic stem cell. Therefore, at the end of the period of time, the desired cell of a different type is obtained.


According to another particular aspect, the invention relates to a method of transforming a cell of a first type to a cell of a second different type. The method comprises contacting the cell of a first type with one or more agents capable of increasing within said cell levels of at least one reprogramming agent and directly or indirectly remodeling the chromatin and/or DNA of the cell. The at least one reprogramming agent is selected for inducing directly or indirectly the expression of morphological and functional characteristics of a desired cell of a different type or different cell lineage.


According to another aspect, the invention relates to a method of transforming a cell of a first type to a cell of a second different type. The method comprises contacting the chromatin and/or DNA of a cell of a first type with an agent capable of remodeling chromatin and/or DNA of said cell; and increasing intracellular levels of at least one reprogramming agent. The at least one reprogramming agent is selected for inducing directly or indirectly the expression of morphological and functional characteristics of a desired cell of a different type or cell lineage.


A further aspect of the invention relates to a method of transforming a cell of a first type to a cell of a desired cell of a different type, comprising increasing intracellular levels of at least one reprogramming agent, wherein the at least one reprogramming agent is selected for inducing directly or indirectly the expression of morphological and functional characteristics of a desired second cell type; and maintaining the cell of a first type in culture conditions for supporting the transformation of the desired cell for a sufficient period of time to allow stable expression of a plurality of secondary genes whose expression is characteristic of phenotypical and functional properties of the desired cell, wherein at least one of the secondary genes is not characteristic of phenotypical and functional properties of an embryonic stem cell. At the end of the period of time the desired cell of a different type is obtained and the obtained cell is further characterized by a stable repression of a plurality of genes expressed in the first cell type.


A further aspect of the invention concerns a process wherein a cell of a first type is reprogrammed to a desired cell of a different type, the process comprising:

    • a transient increase of intracellular levels of at least one reprogramming agent, wherein the at least one reprogramming agent induces a direct or indirect endogenous expression of at least one gene regulator, and wherein the endogenous expression of the said at least one gene regulator is necessary for the existence of the desired cell of a different type;
    • a stable expression of said at least one gene regulator;
    • stable expression of a plurality of secondary genes, wherein the stable expression of the secondary genes is the result of the stable expression of the at least one gene regulator, and wherein: (i) stable expression of the plurality of secondary genes is characteristic of phenotypical and/or functional properties of the desired cell, (ii) stable expression of at least one of said secondary genes is not characteristic of phenotypical and functional properties of an embryonic stem cell, and wherein (i) and (ii) are indicative of successful reprogramming of the cell of the first type to the desired cell of the different type.


In particular embodiments, the at least one reprogramming agent in the process is a Msi1 polypeptide, or a Ngn2 polypeptide together with a MDB2 polypeptide. In particular embodiments, the at least one gene regulator is Sox2 Msi1, or both. In additional embodiments the at least one gene regulator may is one or more of the genes listed in Table A for Neural Stem-Like Cells.


According to another aspect, the invention relates to a method of obtaining a Stem-Like Cell (SLC), comprising:

    • i) providing a cell of a first type;
    • ii) transiently increasing in the cell intracellular levels of at least one reprogramming agent, whereby the transient increase induces direct or indirect endogenous expression of at least one gene regulator;
    • iii) placing the cell in conditions for supporting the transformation into the stem-like cell and maintaining intracellular levels of the at least one reprogramming agent for a sufficient period of time to allow stable expression of the at least one gene regulator in absence of the reprogramming agent;
    • iv) maintaining the cell in culture conditions for supporting the transformation into the stem-like cell for a sufficient period of time to allow stable expression of a plurality of secondary genes whose expression is characteristic of phenotypical and/or functional properties of the stem-like cell, wherein at least one of the secondary genes is not characteristic of phenotypical and functional properties of an embryonic stem cell. At the end of said period of time a stem-like cell is obtained.


According to another aspect, the invention relates to a method of obtaining a Stem-Like Cell. The method comprises increasing intracellular levels of at least one polypeptide specific to the desired stem cell type that is able to drive directly or indirectly transformation of the cell of the first type into the Stem-Like Cell. For increasing the yield or type of Stem-Like Cell, the method may further comprises contacting chromatin and/or DNA of a cell of a first type with a histone acetylator, an inhibitor of histone deacetylation, a DNA demethylator, and/or an inhibitor of DNA methylation; and/or increasing intracellular levels of at least one other polypeptide specific to the desired stem cell type that is able to drive directly or indirectly transformation of the cell of the first type into a Stem-Like Cell.


According to another aspect, the invention relates to a method of obtaining a Neural Stem-Like Cell (NSLC). The method comprises increasing intracellular levels of at least one neural stem cell specific polypeptide that is able to drive directly or indirectly transformation of the cell of the first type into a NSLC. For increasing the yield or type of NSLC, the method further comprises. contacting chromatin and/or DNA of a cell of a first type with a histone acetylator, an inhibitor of histone deacetylation, a DNA demethylator, and/or an inhibitor of DNA methylation; and/or increasing intracellular levels of at least one other neural stem cell specific polypeptide that is able to drive directly or indirectly transformation of the cell of the first type into a NSLC.


Another aspect of the invention concerns a method of obtaining a Neural Stem-Like Cell (NSLC). In one embodiment the method comprises transfecting a skin cell with a polynucleotide encoding Musashi1, Musashi1 and Neurogenin 2, Musashi1 and Methyl-CpG Binding Domain Protein 2 (MBD2), or Neurogenin 2 and Methyl-CpG Binding Domain Protein 2, thereby reprogramming the skin cell into a NSLC. In another embodiment the method comprises exposing a skin cell to: (i) an inhibitor of histone deacetylation, (ii) an inhibitor of DNA methylation, (iii) a histone acetylator, and/or (iv) a DNA demethylator such as a MBD2 polypeptide and/or transfecting with a polynucleotide encoding a MBD2 polypeptide; and further transfecting the cell (either simultaneously, before, or afterwards) with a polynucleotide encoding MUSASHI1 and/or with a polynucleotide encoding NGN2, thereby reprogramming the skin cell into a NSLC. Some other cells, such as keratinocytes and CD34+ cells, can also be used and reprogrammed.


In one particular embodiment, the method of obtaining a Neural Stem-Like Cell (NSLC), comprises:

    • providing a cell of a first type;
    • introducing into the cell one or more polynucleotide capable of transient expression of one or more the following polypeptides: Musashi1 (Msi1); a Musashi1 (Msi1) and a Neurogenin 2 (Ngn2); a Musashi1 (Msi1) and methyl-CpG binding domain protein 2 (MBD2); and Neurogenin 2 (Ngn2) and methyl-CpG binding domain protein 2 (MBD2); and
    • placing the cell in culture conditions supporting the transformation into a NSLC for a sufficient period of time to allow a stable expression of a plurality of genes whose expression is characteristic of phenotypical and functional properties of a NSLC.


At the end of the period of time a NSLC is obtained and the obtained NSLC is further characterized by a stable repression of a plurality of genes expressed in the first cell type.


According to another embodiment, the method of obtaining a Neural Stem-Like Cell (NSLC), comprises:

    • providing a cell of a first type which is not a NSLC;
    • increasing intracellular levels of at least one neural stem cell specific polypeptide, wherein the polypeptide is capable of driving directly or indirectly transformation of the cell of the first type into a NSLC; and
    • contacting the chromatin and/or DNA of the cell of a first type with a histone acetylator, an inhibitor of histone deacetylation, a DNA demethylator, and/or a chemical inhibitor of DNA methylation.


According to another embodiment, the method of obtaining a Neural Stem-Like Cell (NSLC), comprises:

    • obtaining a non-NSLC;
    • co-transfecting the non-NSLC with a first polynucleotide encoding a MBD2 polypeptide and with at least one second polynucleotide encoding a MUSASHI1 polypeptide and/or encoding a NGN2 polypeptide;
    • placing the co-transfected cell in culture conditions for supporting the transformation of NSLC until said NSLC is obtained.


Certain aspects of the invention concerns isolated cells, cell lines, compositions, 3D assembly of cells, and tissues comprising cells obtained using the methods described herein. Additional aspects concerns the use of such isolated cells, cell lines, compositions, 3D assembly of cells, and tissues of medical treatment and methods of regenerating a mammalian tissue or organ.


Yet, a further aspect concerns a method for repairing or regenerating a tissue in a subject. In one embodiment the method comprises the administration of a reprogrammed cell as defined herein to a subject in need thereof, wherein the administration provides a dose of reprogrammed cells sufficient to increase or support a biological function of a given tissue or organ, thereby ameliorating the subject's condition.


The benefits of the present invention are significant and include lower cost of cell therapy by eliminating the need of immuno-suppressive agents, no need for embryos or fetal tissue, thus eliminating ethical and time constraints, lower cost of production, and no health risks due to possible transmission of viruses or other disease. In addition, since the cells are created fresh, they tend to be more potent than cells that have been passaged multiple times.


Additional aspects, advantages and features of the present invention will become more apparent upon reading of the following non-restrictive description of preferred embodiments which are exemplary and should not be interpreted as limiting the scope of the invention.





BRIEF DESCRIPTION OF THE FIGURES


FIG. 1 is a panel of light micrograph (10×) presenting cell morphology changes of untransfected and transfected cells with Msi1 and MBD2 at various time points.



FIG. 2 is a panel of photomicrographs obtained using Cellomics™ (10×) and revealing NCAM positive cells in transfected cells with Msi1 or Ngn2 in the presence of MBD2. HFFs were pre-treated with cytochalasin B (10 g/ml) and transfected with pCMV6-XL5-Msi1 and pCMV6-XL5-MBD2 or pCMV6-XL4-Ngn2 and pCMV6-XL5-MBD2. After 24 h following transfection, the medium was changed and cells were cultured in proliferation medium (NPBM, Lonza) supplemented with EGF (20 ng/ml) and bFGF (20 ng/ml) for one week. Differentiation was induced by changing the medium to NbActive (BrainBits™) supplemented with NGF (20 ng/ml), bFGF (20 ng/ml), ATRA (5 μM) and Forskolin (10 μM). Cells were incubated at 37° C., 5% CO2, 5% O2 for 20 days.



FIG. 3 is a panel of photomicrographs obtained using Cellomics™ (10×) and revealing MAP2b positive cells in transfected cells with Msi1 or Ngn2 in the presence of MBD2. MAP2b positive cells were undetectable in untransfected cells and cells transfected with Pax6/MBD2. HFFs were pre-treated with cytochalasin B (10 kg/ml) and transfected with pCMV6-XL5-Msi1, pCMV6-XL4-Ngn2 or pCMV6-XL5-Pax6, and pCMV6-XL5-MBD2. After 24 h following transfection, the medium was changed and cells were cultured in proliferation medium (NPBM, Lonza) supplemented with EGF (20 ng/ml. Peprotech) and bFGF (20 ng/ml, Peprotech) for one week. Differentiation was induced by changing the medium to NbActive (BrainBits™) supplemented with NT-3 (20 ng/ml), bFGF (20 ng/ml), ATRA (5 μM) and Forskolin (10 μM). Cells were incubated at 37° C., 5% CO2, 5% O2 for 2 weeks.



FIG. 4. (A) is a panel of photographs showing that neurospheres formed by NSLCs from Example V were completely dissociated into single cell suspensions using Accutase and one single cell was monitored over time to reveal neurosphere formation capacity (Light microscope observation). Neurospheres stained positive for Sox2. (B) is a panel of photographs from immunohistochemistry results obtained using Cellomics™. Immunohistochemistry was performed, on day 20, to detect makers for neurospheres and compared to expression levels in neurospheres formed by normal human neuroprogenitor cells (hNPC, Lonza). In addition to Sox2, cells stained positive for the neural stem cells markers Musashi, CD133, Nestin, and GFAP. Cells also stained positive for BIII-tubulin (a marker for neurons), O4 (a marker for oligodendrocytes), and GFAP (a marker for astrocytes), indicating the tri-potent differentiation potential of both sets of cells (NSLC and hNPC), and negative for NGFrec and NeuN (markers for differentiated neurons) indicating that the cells were not terminally differentiated.



FIG. 5 is a panel photomicrographs from immunohistochemistry results obtained using Cellomics™. Immunohistochemistry was performed on HFFs, NSLCs, and hNPCs to detect expression of markers for fibroblasts as well as neural stem cells (Sox2, Nestin, GFAP) in adherent cultures (that prevented cells from floating and forming neurospheres). Nuclei were stained with Hoechst (upper level pictures). HFFs expressed fibroblasts markers while NSLCs created from these HFFs did not. In comparison, the NSLCs expressed neural stem cell markers similarly to hNPCs while the HFFs did not express any of these markers.



FIG. 6 is a panel photomicrographs showing Human NSLCs. Human NSLCs were induced to differentiate into neuronal lineages in the presence of NS-A differentiation medium (StemCell Technologies) in the presence of BDNF (20 ng/ml, Peprotech) and bFGF (40 ng/ml, Peprotech) for three weeks. At different time point of differentiation, immunostaining using Cellomics™ (10×) revealed differentiation of the cells as shown by the decrease of Sox2 positive cells and increase in the number and intensity of staining of p75, βIII-tubulin and GABA positive cells, as well as differentiated morphology, while the total number of cells increased as shown by Hoechst staining.



FIG. 7 is another panel of photomicrographs. HFF, Keratinocytes, and CD34+ were transfected with pCMV6-Msi1-Ngn2 and pCMV6-XL5-MBD2. After 24 h following transfection, the medium was changed to proliferation medium (StemCell Technologies) supplemented with EGF (20 ng/ml. Peprotech) and bFGF (20 ng/ml, Peprotech) for two week and then analyzed. Photomicrographs using Cellomics™ (10×) show that NSLCs created from all three types of cells are positive for Nestin, Sox2 and GFAP (markers for neural stem cells), while the original HFFs are not.



FIG. 8 is panel of photomicrographs showing the effect of CDM medium on the trans-differentiation of HFF towards neurons. HFF were pre-treated with cytochalasin B (10 g/ml) and histone deacetylation inhibitor (VPA, 4 mM) and DNA methylation inhibitor (5-Aza, 5 μM and cultured in CDM medium containing 3:1 ratio of Dulbecco's modified Eagle medium (DMEM, high glucose (4.5 g/L) with L-glutamine and sodium pyruvate) and Ham's F-12 medium supplemented with the following components: EGF (4.2×10−10M), bFGF (2.8×10−10M), ITS (8.6×10−5M), dexamethasone (1.0×10−7M), L-ascorbic acid phosphate magnesium salt n-hydrate (3.2×10−4M), L-3,3′,5-triiodothyronine (2.0×10−10M), ethanolamine (10−4M), GlutaMAX™ (4×10−3M), glutathione (3.3×10−6M). After 24 h the culture medium was replaced with 75% of CDM medium and 25% of Neuronal Proliferation medium (Lonza, Cat #CC-3210); during the following 3 days, the ratio of the medias were changed to 50%:50%, 25%:75%, and then 100% Neuronal Proliferation medium by the third day. Photomicrographs were taken by Cellomics™ (10×) after immunostaining the cells with βIII-tubulin (neuronal marker) and Hoechst (to stain nuclei) at different time-points. Cells started trans-differentiating within a few days and the trans-differentiatied cells were βIII-tubulin positive; however after one week a spontaneous reversion to fibroblastic shape and loss of βIII-tubulin expression was observed.



FIG. 9 is panel of photomicrographs showing characterization of reprogrammed cells within CDM at different time points following the transfection with Msi1 and Ngn2. The transfected cells were treated with Cytochalasin B (10 μg/ml), VPA (4 mM) and 5-AZA (5 μM) resulting in a disruption of the microfilaments and rounding up of the cells and loosening of the chromatin. Immunohistochemistry on the 3-Dimensional CDM was performed after one and two weeks using Cellomics™ (10×). The cells were positive for neuronal mature marker, such as MAP2b, but were absent in the untransfected control CDM.



FIG. 10 is another panel of photomicrographs. Cells within Day 4 CDM were lipotransfected with the two vectors pCMV6-XL5-Msi1 and pCMV6-XL4-Ngn2 individually or together in combination with pCMV-XL5-MBD2 for a period of 6 hours. In parallel, transfection was performed on fresh HFFs after the 6 hours using Nucleofection, and these fresh HFFs were placed on top of the CDM at the same time as the lipofectamine media was changed to fresh CDM medium after 6 hours. After 24 hours the medium was changed to Neural proliferation medium (NPBM, Lonza) with the presence of Noggin (50 ng/ml, Peprotech), recombinant hFGF (20 ng/ml, Peprotech), and recombinant hEGF (20 ng/ml, Peprotech) for one week. Differentiation was induced at day 7, by adding NS-A differentiation medium (StemCell Technologies) for 24 days. Immunohistochemistry was performed at various time points using Cellomics™ (10×). The CDM was stained with a specific antibody against Nestin (a marker for neural stem cells), and cells within the CDM expressed Nestin at all timepoints tested (Day 8, 15, and 21) following transfection. Cells within the untransfected control CDM did not express any Nestin.



FIG. 11 is a panel showing a picture of a polyacrylamide gel electrophoresis. NSLCs grown as adherent cultures or suspension cultures (as neurospheres) both express telomerase (which is expresses in all stem cells, but not in normal differentiated somatic cells). Both early (p5) and late (p27) passage NSLCs express telomerase. (The original HFFs from which the NSLCs were created did not express telomerase.) The samples (NSLCs) were spun down and protein concentration of the supernatant was determined using the BCA Assay. 900 ng of protein from each cell extract was added directly to the TRAP reaction mixture containing TRAP reaction buffer, dNTPs, template substrate (TS) primer, TRAP primer mix and Taq polymerase. The reaction mixtures were incubated at 30° C. for 30 minutes for template synthesis, followed by a PCR procedure (95° C./15 min for initial denaturation, 94° C./30 sec, 59° C./30 sec, 72° C./1 min for 32 cycles) for amplification of the extended telomerase products. To detect telomerase activity, polyacrylamide gel electrophoresis (PAGE) was performed for the reaction products on a 10% non-denaturing TBE gel. After electrophoresis, the gel was stained with SYBR® Green I Nucleic Acid Gel Stain for 30 minutes, followed by image capture using the Gel-Documentation System (Alpha Innotech). All 4 samples were telomerase positive (as indicated by the TRAP product ladder).



FIG. 12 is a panel showing a picture showing Southern blot analysis of two different NSLC samples analyzed for Msi1 and Ngn2 gene integration two weeks after transient transfection. The Dig-labeled PCR probe revealed distinct signals in the positive control samples where the Msi1/Ngn2 plasmid DNA was spiked into HFF genomic DNA for the equivalence of 1, 10 or 100 integrations per genome. There were a few weak and identical bands that appeared in the restriction enzyme digested genomic DNA from untransfected HFF and NSLC samples #1 and #2, suggesting that there was no plasmid DNA integration into the genomic DNA of NSLCs. These faint bands may represent the endogenous Ngn2 gene since the 1.2 kb Dig-labeled PCR probe contains a small part of the Ngn2 gene. There were positive signals in the lane of the DNA kb ladder as the bands belong to a number of plasmids digested to completion with appropriate restriction enzymes (NEB). This data shows that no, or only a tiny number of, NSLCs had plasmid integration into the host genome after transient transfection, and that the transiently transfected genes were only present in the cells for a short period of time (less than two weeks).



FIG. 13 is a panel which a line graph and a bar graph showing improvement and significantly better clinical scores in EAE mice treated with NSLCs. Female 8 weeks old C57BL/6 mice were immunized with MOG35-55 (Sheldon Biotechnology Centre McGill University) in CFA containing 5 mg/ml of desiccated (killed and dried) Mycobacterium tuberculosis H37Ra (Difco, inc) at two sites on the back, and injected with 200 ng of pertussis toxin (List Biological Laboratories, Inc) in PBS intraperitoneally on days 0 and 2. Once the mice started showing symptoms of EAE (on Day 13 post-immunization), they were intravenously injected with 200 μl of NSLC (1 million cells), hNPC (1 million cells), saline, or saline with cyclosporine. All mice except the saline control group received daily injections of cyclosporine. Mice were scored daily for clinical disease; data represent average daily scores. Mice that received a single injection of NSLCs had a significantly lower disease severity than mice that received hNPCs or cyclosporine alone.



FIG. 14 is a line graph showing the results of rotarod assessments according to Example XVII part 2. Rats were trained on the rotarod prior to the start of the experiment. Rats were placed on a stationary and rotating rotarod (rotating at 20 rpm) and the amount of time spent by the rats walking on the rotarod before falling off was monitored. Measurements were taken before (pre-surgery) and after (post-surgery) surgical left brain hemisphere ablation and treatment. The data points represent the mean number of falls by each animal during each 60 second testing session carried out at a constant speed of 20 rpm. Each group consisted of eight rats.



FIG. 15 is a line graph showing the results of the walking beam assessments according to Example XVII part 2. Rats were measured on their ability to cross a 100 cm long beam after surgical left brain hemisphere ablation and treatment. Two days after surgery, all groups fail to pass the test, and the animals are not able to stay in balance on the beam. One week after the surgery, all the animals show an improvement on their walking capacity, but no significant difference was noticeable between the different treated groups. From week 4 until week 26, the animals treated with NSLCs show significant improvement in their walking capacity compared to the other groups.



FIG. 16 is a panel showing photographs of ADSCs transiently transfected with various pluripotent vectors using nucleofector as described in Example XIX. Following the transfection cells were cultured in 6-well plates in suspension with a 50:50 mixture of ADSC complete medium (StemPro™-43) and embryonic stem cells medium (mTeSR1™, StemCell Technologies). After two days in culture, cells were re-transfected with the same plasmids and plated in 96 well-plates coated with Matrigel™ (BD Biosciences) in the presence of mTeSR1™ complete medium supplemented with thiazovivin (0.5 μM), an ALK-5 inhibitor (SB341542, Stemgent, 2 μM), and an inhibitor of MEK (PD0325901, Stemgent, 0.5 μM). Medium was changed every day and cells were cultured for 22 days at 37° C., 5% CO2, 5% O2, followed by AP staining and immunohistochemistry to analyse the expression of pluripotency markers. Cells formed colonies and were found to express both pluripotency markers Oct4 and AP after transfecting cells with pEF-Rex1-EF-Oct4-2A-Klf4-2A-RFP.



FIG. 17 is a panel showing photographs of ADSCs transiently transfected with pCMV6-XL5-Rex1/pCMV6-XL5-Klf4 and pCMV6-XL5-Rex1/pCMV6-XL4-Oct4. After the second transfection, ADSCs were cultured in 96-well plates coated with Matrigel™ for 24 days in the presence of mTeSR1™ medium supplemented with SB341542 and PD0325901 at 37° C., 5% CO2, 5% O2. In order to characterize subpopulations of cells after transfection, live staining, immunohistochemistry and AP staining were used. 1-5% of total cells transfected with Rex1/Oct4 or Rex1/Klf4 showed a SSEA-4+ and TRA-1-81+ phenotype (early pluripotency markers). The observation over time showed that the phenotype of these colonies moves from an early SSEA-4+ phenotype to a late Oct4+/Sox2/Nanog+ phenotype starting at Day 22, which was closer to the final reprogrammed state and a pluripotent-like cell.



FIG. 18 is a panel showing photographs of ADSCs transiently transfected with various pluripotent vectors. Following transfection the cells were plated in StemPro™ MSC SFM medium on Matrigel™ (BD Biosciences) coated 24 well plates and incubated at 37° C., 5% CO2, 5% O2. On day 1, media was changed to a mix of 75% StemPro™ MSC and 25% hES cell medium; the percentage of StemPro™ MSC SFM medium was decreased every day over four days to have 100% hES cell medium by day 4. Then medium was changed every two days. The hES cell medium consisted in Dulbecco's Modified Eagle's Medium (DMEM, Invitrogen) supplemented with 20% Knockout™ Serum Replacement (KSR, Invitrogen), 1 mM GlutaMAX™, 100 μM Non-essential Amino acids, 100 μM 3-mercaptoethanol and 10 ng/ml Fgf-2. In order to characterize subpopulations of cells after transfection, live staining, immunohistochemistry and AP staining were used. Transfected cells transfected with Oct4/UTF1/MBD2, Oct4/Dppa4/MBD2, FoxD3/Dppa4/MBD2, Oct4/FoxD3/Dppa4, and Sox2/FoxD3/UTF1 were positive for SSEA-4+, TRA1-60, and TRA-1-81+ phenotype (early pluripotency markers) at day 14.



FIG. 19 is a panel showing photographs of transiently transfected HFFs. HFFs were transiently transfected using the Nucleofector® II Device (Lonza) following the procedure described in Example II with the exception that 1 μg of each of the following 3 DNA plasmids was used: pCMV-Oct4nuc-IRES2-Sox2nuc, pCMV-Klf4nuc-IRES2-Cmycnuc and pCMV-Nanognuc-IRES2-Lin28. The cells were pre-treated with or without VPA and 5-Aza. Following transfection the cells were plated in the fibroblast medium, supplemented with or without VPA (2 mM) and 5-AZA (2.5 μM) on Matrigel™ (BD Biosciences) coated 6-well plates and incubated at 37° C., 5% CO2. On Day 1 and 2, media was changed to 100% mTeSR1™ medium (StemCell Technologies) supplemented with or without VPA and 5-AZA. On Day 3 and 6, cells were re-transfected as above and plated on Matrigel™ coated plates in mTeSR1™ medium supplemented with or without VPA and 5-AZA. Media was changed daily as above. Medium was supplemented with Y27632 (Stemgent, 10 μM) from Day 7 to Day 14 to promote viability and clonal expansion of potential reprogrammed cells. Cells were analysed at day 20 using the Alkaline Phosphatase Detection Kit (Millipore) and by immunohistochemistry analysis. Some cells stained positive for the pluripotency markers AP, SSEA-4 and TRA-1-81 (similar to Mel2 human embryonic stem cell line (positive control)). These clones were obtained only in the condition that did not contain inhibitors (i.e.: VPA and 5-AZA). No clones were observed for the condition treated with these inhibitors.



FIG. 20 is a panel showing photographs of transfected NSLCs and BG-01. NSLCs and BG-01 NS were transfected as previously described in Example II by two episomal vectors, pEF-Oct4nuc-IRES2-MBD2 (NC1) or pCMV-FoxD3-2A-Oct4-2A-Klf4 (F72). Following transfection cells were collected and plated onto uncoated petri-dishes in the presence of Proliferation medium and mTeSR1™ medium (50:50) into proliferation conditions at 37° C., 5% CO2. After 48 hours, cells were re-transfected by the same plasmid and plated in 96-well plates coated with Matrigel™ and cultured in the presence of mTeSR1™ medium supplemented by the small molecules BIX01294 (Stemgent, 2 μM) and BayK8644 (Stemgent, 2 μM) at 37° C., 5% O2 for 22 days, after which live staining and immunohistochemistry were performed to characterize subpopulations of cells for pluripotency markers. Cells formed colonies positive for both TRA-1-81 and SSEA-4 indicative of pluripotent-like cells.



FIG. 21 is a panel showing bright field pictures at day 17 of fibroblasts transfected with Msi1/Ngn2 and pCMV6-XL5-MBD2 placed in different media conditions and showing different morphologies and degree of differentiation. (a) Cells in neural proliferation medium from day 1 to day 12, and then in neural differentiation medium with cytokines from day 12 to 17. (b) Cells in neural proliferation medium from day 1 to day 12, and then in NbActive4 medium with cytokines from day 12 to 17. (c) Cells in neural differentiation medium with cytokines plus Fgf-2 from day 1 to day 12, and then in the same medium but without Fgf-2 from day 12 to 17. (d) Cells in NbActive4 medium with cytokines plus Fgf-2 from day 1 to day 12, and in then the same medium but without Fgf-2 from day 12 to 17. (e) Cells in CDM II medium with cytokines plus Fgf-2 from day 1 to day 12, and in then the same medium but without Fgf-2 from day 12 to 17.



FIG. 22 is a panel showing pictures of immunochemistry results at day 17 of fibroblasts transfected with Msi1/Ngn2 and pCMV6-XL5-MBD2 in FIG. 21. FIGS. 22A and 22B: Cells were in NS-A Proliferation Medium from day 1 to day 12, and then in NS-A Differentiation Medium (A) or NBActive4 medium (B) with cytokines from day 12 to 17. There were more cells in B, but Differentiation from day 12-17 was too short to induce expression of βIII-tubulin in both cases. FIGS. 22C-E: Cells were in NS-A Differentiation Medium (C) or NbActive4 medium (D) from day 1-17 (with FGF-2 supplementation from day 1-12), or CDM II medium from day 1-12 and then NS-A Differentiation Medium from day 12-17 (E). There were a large number of cells in C and a much smaller number of cells in D and E. Cells were immunopositive for both GFAP and βIII-tubulin in all cases and placing the cells in differentiation or non-proliferation media from day 1 onwards appears to have induced a more direct transformation into neurons and glia, with more intense βIII-tubulin than GFAP positive cells in E.



FIG. 23 is a panel showing two heat maps providing a global overview of the gene expression comparison between either NSLC vs. HFF (Set 1), or NSLC vs. hNPC (Set 2). NSLC has a distinct gene expression profile when compared to either HFF or hNPC. Based on the intensity (the higher the intensity, the higher the relative change in expression), NSLC is much more similar to hNPC than to HFF.



FIG. 24 is a panel showing pictures of NSLCs. NSLCs were tested to determine if they are a population of Skin-Derived Precursors Cells (SKPs). SKPs capable of proliferating in response to EGF and bFGF, express nestin and fibronectin, and can differentiate into both neuronal and mesodermal progeny including into adipocytes. For this purpose a standard protocol for turning SKPs into adipocytes was performed, in which adipocyte-derived stem cells (ADSCs) and NSLCs were cultured in StemPro™ proliferation medium and differentiation towards adipocytes were induced by culturing these cells in differentiation medium consisting in DMEM/F12 (50:50), ITS (1:100), HEPES (1:100), GlutaMAX™ (1:100), T3 (0.2 nM), Rosiglitasone (0.5 μg/ml), IBMX (100 μM) and Dexamethasone (1 μM). Three days later, IBMX and Dexamethasone were withdrawn from the medium. At day 10, cells were fixed with a 4% formaldehyde solution for 10 min and stained with Oil Red O (Invitrogen) staining solution. Adipose cells appeared red with lipid droplets (bright white spots in left picture) specifically stained with Oil Red O; however NSLCs stained negative and had no presence of lipid droplet in the cells, but instead adopted neuronal cell morphology. These results conform that NSLCs are not a population of Skin-Derived Precursors Cells (SKPs).



FIG. 25 is a panel showing pictures of ADSCs transiently transfected with two pluripotent vectors: pEF-Rex1-EF-Oct4-2A-Klf4-2A-RFP (NF10) or pCMV-Sall4-2A-Oct4-2A-Klf4-2A-Nanog (S71) using nucleofector as described in Example XIX. Following the transfection cells were cultured in suspension with 50:50 ratio of adipocyte complete medium (StemPro-43) and embryonic stem cells medium (mTesR1, StemCell Technologies). After two days in culture, cells were re-transfected with the same plasmids listed above and cells were plated in 96 well-plates coated with Matrigel (BD Biosciences) in the presence of mTesR complete medium supplemented with thiazovivin (0.5 .mu.M), an ALK-5 inhibitor (SB 341542, Stemgent, 2 .mu.M), and inhibitor of MEK (PD0325901, Stemgent, 0.5 .mu.M). Medium was changed every day and cells were cultured for 22 days at 37° C., 5% CO2, 5% O2. Immunohistochemistry was performed to identify cells stained with different pluripotency markers. Cells formed colonies starting day 15 and were found to express pluripotency markers (Sox2, Oct4, Nanog, SSEA, TRA-1-60, TRA-1-81, E-cadherin).



FIG. 26 is a panel of immunofluorescence images illustrating the differentiation of PLSCs into ectoderm, mesoderm and endoderm lineages. Representative immunofluorescence images of embryonic bodies derived at day 12, 18, and 28 stained for stage neural markers (13111 tubulin and GFAP), for early mesoderm marker (Brachyury and GATA4); and for early endoderm marker (FoxA2).



FIG. 27 is a panel of images comparing the level of expression of selected genes between NSLC (human neural stem-like cells created from HFF), NP (human primary neural progenitor cells (Lonza)), FF (the human foreskin fibroblasts used to create the NSLC (ATCC)), NT2 (human NT2/D1 teratocarcinoma cell line (ATCC)), NT2-N (human NT2 neurons differentiated from the NT2/D1 cells), NT2-A (human NT2 astrocytes differentiated from the NT2/D1 cells), and NTC no template control for RT-PCR (only primers, no cDNA). (a): Stem Cell Markers: RT-PCR analysis shows that NSLCs express SOX2 but not OCT4a or NANOG. NT2 cells were used as positive control for SOX2, OCT4 and NANOG. NP cells were used as positive control for SOX2. (Note: OCT4a is a stem cell marker. OCT4b is a splicing variant of OCT4, which its expression is not limited to stem cells.) (b): Notch Signalling Pathway: NOTCH regulates cell fate determination, neuronal differentiation and glial determination. RT-PCR data show that various components of Notch signalling pathway are expressed in NSLCs. HES: hairy enhancer of split (a Notch target gene). (c): Early Neuronal Markers. NES: nestin; CD133: also known as Prominin 1 (PROW); PAX6: paired box 6; ASCL1: achaete-scute complex homolog 1 (also knows as MASH1). (d): Neuron Markers. NFL: neurofilament light chain; NFM: neurofilament medium chain; NFH: neurofilament heavy chain; SYN: synaptophysin; NSE: neuron-specific enolase; MAP2: microtubule associated protein 2a+2b. (e): Neurotrophic factors. BDNF: brain-derived neurotrophic factor; GDNF: glial cell-derived neurotrophic factor. (f): Astroglial markers. GFAP: glial fibrillary acidic protein. (g): Control. ACTB: beta actin.



FIG. 28 is a panel of immunofluorescence images illustrating functional cell-cell communication in NSLC cells through gap junctions. A single NSLC cell was pre-loaded with two dyes [Dil (red) and calcein AM (green)] and plated on an unlabeled layer of NSLC cells. Calcein readily transferred from the donor cells to a large population of receiving cells after 3 hours in vitro, confirming gap junctional intercellular communication among NSLC cells. Calcein AM (MW=994.7 kDa) coverts to calcein (MW=623 kDa) through estrase hydrolysis after it enters the cell. Calcein passes through gap junctions from donor cell to receiving cells. In contrast, Dil (a cationic membrane tracer used to label the donor cell) does not pass through gap junctions. (A) red filter; (B) green filter; (C) phase filter; (D) red+green filters; (E) red+phase filters; and (F) red+green+phase filters.



FIG. 29. Panel (A) shows a light micrograph of NSLCs. Panels (B), (C) and (D) are a panel of immunofluorescence images illustrating that NSLCs readily express synaptotagmin (a synaptic vesicle protein; panel C) and Map2 (a neural differentiation associated marker; panel B). Panel D is a composite image of panel B and C.



FIGS. 30 (A) and (B) are a panel of immunofluorescence imagesillustrating that NSLCs readily attach to PGA scaffolds designed for implantation into the brain. Carboxyfluourodiacetate (CFDA; green stain) confirms optimal cell survival (>93%) 24 hours after cells were plated on the scaffold. NSLC cells showed robust attachment and survival to the surface for at least one week in vitro. Furthermore, Neurites grow along the length of each PGA fiber, following the pattern of the scaffold.





DETAILED DESCRIPTION OF THE INVENTION

The present invention relates to methods for cell dedifferentiation and cell reprogramming. A significant aspect of the present invention is that it permits the use of a patient's own cells to develop different types of cells that can be transplanted after steps of in vitro dedifferentiation and in vitro reprogramming. Thus, this technology eliminates the problems associated with transplantation of non-host cells, such as, immunological rejection and the risk of transmitting disease. In addition, since the cells are “newly created”, they have the potential to be more potent than alternative sources of natural cells that have already divided multiple times.


Definitions


As used herein and in the appended claims, the singular forms “a,” “an”, and “the”, include plural referents unless the context clearly indicates otherwise. Thus, for example, reference to “a cell” includes one or more of such cells or a cell line derived from such a cell, reference to “an agent” includes one or more of such agent, and reference to “the method” includes reference to equivalent steps and methods known to those of ordinary skill in the art that could be modified or substituted for the methods described herein.


As used herein, the term “polynucleotide” refers to any DNA or RNA sequence or molecule, comprising encoding nucleotide sequences. The term is intended to encompass all polynucleotides whether occurring naturally or non-naturally in a particular cell, tissue or organism. This includes DNA and fragments thereof, RNA and fragments thereof, cDNAs and fragments thereof, expressed sequence tags, artificial sequences including randomized artificial sequences.


As used herein, the term “polypeptide” refers to any amino acid sequence having a desired functional biological activity (e.g. DNA demethylation). The term is intended to encompass complete proteins, fragments thereof, fusion proteins and the like, including carbohydrate or lipid chains or compositions.


“Trans-differentiation” refers to a direct switch of an already differentiated cell to another type of differentiated cell.


“De-differentiation” refers to the loss of phenotypic characteristics of a differentiated cell by activating or deactivating genes or metabolic pathways.


“Marker” refers to a gene, polypeptide, or biological function that is characteristic of a particular cell type or cellular phenotype.


“Genetically-engineered DNA sequence” is meant a DNA sequence wherein the component sequence elements of DNA sequence are organized within the DNA sequence in a manner not found in nature.


“Signal sequence” refers to a nucleic acid sequence which, when incorporated into a nucleic acid sequence encoding a polypeptide, directs secretion of the translated polypeptide from cells which express said polypeptide, or allows the polypeptide to readily cross the cell membrane into a cell. The signal sequence is preferably located at the 5′ end of the nucleic acid sequence encoding the polypeptide, such that the polypeptide sequence encoded by the signal sequence is located at the N-terminus of the translated polypeptide. By “signal peptide” is meant the peptide sequence resulting from translation of a signal sequence.


“Ubiquitous promoter” refers to a promoter that drives expression of a polypeptide or peptides encoded by nucleic acid sequences to which promoter is operably linked. Preferred ubiquitous promoters include human cytomegalovirus immediate early (CMV); simian virus 40 early promter (SV40); Rous sarcoma virus (RSV); or adenovirus major late promoter.


“Gene expression profiling” means an assay that measures the activity of multiple genes at once, creating a global picture of cellular function. For example, these profiles can distinguish between human neural stem cells and somatic cells that are actively dividing or differentiating.


“Transfection” refers to a method of gene delivery that introduces a foreign nucleotide sequences (e.g. DNA molecules) into a cell preferably by a non-viral method. In preferred embodiments according to the present invention foreign DNA is introduced to a cell by transient transfection of an expression vector encoding a polypeptide of interest, whereby the foreign DNA is introduced but eliminated over time by the cell and during mitosis. By “transient transfection” is meant a method where the introduced expression vectors and the polypeptide encoded by the vector, are not permanently integrated into the genome of the host cell, or anywhere in the cell, and therefore may be eliminated from the host cell or its progeny over time. Proteins, polypeptides, or other compounds can also be delivered into a cell using transfection methods.


“Neuroprogenitor Cell” refers to an immature cell of the nervous system, which can differentiate into neurons and glia (oligodendrocytes and astrocytes). “Neural Stem Cell” is an ectoderm germ layer derived multipotent stem cell having, as a physiological feature, a capacity to form neuroprogenitor cells and under physiological conditions that favor differentiation to form neurons and glia. “Neural Stem-Like Cell” or “NSLC” refers to any cell-derived multipotent stem cell having, as a physiological feature, a capacity to form other neural stem-like cells and neuroprogenitor-like cells and under physiological conditions that favor differentiation to form neuron-like cells and glial-like cells.


“Neurosphere” refers to a cellular aggregate of neural stem cells and neuroprogenitor cells that form a floating sphere formed as a result of proliferation of the neural stem cells and neuroprogenitor cells in appropriate proliferation conditions. NSLCs also form neurospheres consisting of aggregates of NSLCs and neuroprogenitor-like cells.


“Reprogrammed cell” refers to a cell that has undergone stable trans-differentiation, de-differentiation, or transformation. Some reprogrammed cells can be subsequently induced to re-differentiate. The reprogrammed cell stably expresses a cell-specific marker or set of markers, morphology, and/or biological function that was not characteristic of the original cell.


“Reprogrammed somatic cell” refers to a process that alters or reverses the differentiation status of a somatic cell, which can be either complete or partial conversion of the differentiated state to an either less differentiated state or a new differentiated state.


“Regeneration” refers to the capability of contributing to the repair or de novo construction of a cell, tissue or organ.


“Differentiation” refers to the developmental process of lineage commitment of a cell.


Differentiation can be assayed by measuring an increase in one or more cell-differentiation specific markers relative to the expression of the undifferentiated cell markers.


“Lineage” refers to a pathway of cellular development, in which a more undifferentiated cell undergoes progressive physiological changes to become a more differentiated cell type having a characteristic function (e.g., neurons and glia are of a neuroprogenitor linage, which is of an ectoderm lineage which formed from blastocysts and embryonic stem (ES) cells).


“Tissue” refers to an ensemble of cells (identical or not) and an extracellular matrix (ECM) that together carry out a specific function or set of functions.


“CDM” is meant a living tissue equivalent or matrix, a living scaffold, or cell-derived matrix.


Cell Transformation


Some aspects of the invention concerns methods and cells to transform or reprogram a given somatic cell into a pluripotent, multipotent and/or unipotent cell. Some aspects of the invention relates to methods for conditioning a somatic cell to reprogramming into a pluripotent, multipotent or unipotent cell.


The terms “transform” or “reprogram” are used interchangeably to refer to the phenomenon in which a cell is dedifferentiated or transdifferentiated to become pluripotent, multipotent and/or unipotent. The dedifferentiated cell could subsequently be redifferentiated into a different type of cell. Cells can be reprogrammed or converted to varying degrees. For example, it is possible that only a small portion of cells are converted or that an individual cell is reprogrammed to be multipotent but not necessarily pluripotent. Thus, the terms “transforming” or “reprogramming” methods can refer to methods wherein it is possible to reprogram a cell such that the “new” cell shows morphological and functional characteristics of a new or different specific cell lineage (e.g. the transformation of fibroblast cells into neuronal cells).


As used herein, the term “somatic cell” refers to any differentiated cell forming the body of an organism, apart from stem cells, progenitor cells, and germline cells (i.e. ovogonies and spermatogonies) and the cells derived therefrom (e.g. oocyte, spermatozoa). For instance, internal organs, skin, bones, blood, and connective tissue are all made up of somatic cells. Somatic cells according to the invention can be differentiated cells isolated from adult or can be fetal somatic cells. Somatic cells are obtained from animals, preferably human subjects, and cultured according to standard cell culture protocols available to those of ordinary skill in the art.


As used herein, “Stem cell” refers to those cells which retain the ability to renew themselves through mitotic cell division and which can differentiate into a diverse range of specialized cell types. It includes both embryonic stem cells that are found in blastocysts, and adult stem cells that are found in adult tissues. “Totipotent cells” refers to cells that have the ability to develop into cells derived from all three embryonic germ layers (mesoderm, endoderm and ectoderm) and an entire organism (e.g., human being if placed in a woman's uterus in the case of humans). Totipotent cells may give rise to an embryo, the extra embryonic membranes and all post-embryonic tissues and organs. The term “pluripotent” as used herein is intended to mean the ability of a cell to give rise to differentiated cells of all three embryonic germ layers. “Multipotent cells” refers to cells that can produce only cells of a closely related family of cells (e.g. hematopoietic stem cells differentiate into red blood cells, white blood cells, platelets, etc.). “Unipotent cells” refers to cells that have the capacity to develop/differentiate into only one type of tissue/cell type (e.g. skin cells).


The present invention allows the reprogramming of any cell to a different type of cell. Although the present application focuses primarily on the preparation of Stem-Like cells, especially, Neural Stem-Like Cells (NSLCs), the invention is not so restricted because many different types of cells can be generated according to the principles described herein. Similarly, while the Examples section describes embodiments where fibroblasts, keratinocytes, CD34+ cells, adipose-derived stem cells (ADSCs), neural stem cells (including NSLCs), and cells within a Cell-Derived Matrix (CDM) are reprogrammed, the invention is not limited such cells. The invention may be employed for the reprogramming of virtually any cell of interest.


Accordingly, a general aspect of the invention relates to a method of transforming a cell of a first type to a cell of a second different type. As used herein, examples of cells of a first type include, but are not limited to germ cells, embryonic stem cells and derivations thereof, adult stem cells and derivations thereof, progenitor cells and derivations thereof, cells derived from mesoderm, endoderm or ectoderm, and a cell of mesoderm, endoderm or ectoderm lineage such as an adipose-derived stem cell (ADSC), mesenchymal stem cell, hematopoietic stem cell (CD34+ cell), skin derived precursor cell, hair follicle cell, fibroblast, keratinocyte, epidermal cell, endothelial cell, epithelial cell, granulosa epithelial cell, melanocyte, adipocyte, chondrocyte, hepatocyte, lymphocyte (B and T lymphocyte), granulocyte, macrophage, monocyte, mononuclear cell, pancreatic islet cell, sertoli cell, neuron, glial cell, cardiac muscle cell, and other muscle cell.


As used herein, examples of cells of a second type include, but are not limited to germ cells, embryonic stem cells and derivations thereof, adult stem cells and derivations thereof, progenitor cells and derivations thereof, cells derived from mesoderm, endoderm or ectoderm, and a cell of mesoderm, endoderm or ectoderm lineage such as an adipose-derived stem cell, mesenchymal stem cell, hematopoietic stem cell, skin derived precursor cell, hair follicle cell, fibroblast, keratinocyte, epidermal cell, endothelial cell, epithelial cell, granulosa epithelial cell, melanocyte, adipocyte, chondrocyte, hepatocyte, lymphocyte (B and T lymphocyte), granulocyte, macrophage, monocyte, mononuclear cell, pancreatic islet cell, sertoli cell, neuron, glial cell, cardiac muscle cell, and other muscle cell. In addition, each of the above “-like” cell (a cell that has similar but not completely identical characteristics of the known natural type of the cell) is also included in the examples of cells of a second type.


According to one particular aspect, the method of transforming a cell of a first type into a cell of a second different type comprises the steps of:

    • i) providing a cell of a first type;
    • ii) transiently increasing in the cell of a first type intracellular levels of at least one reprogramming agent, whereby the transient increase induces direct or indirect endogenous expression of at least one gene regulator;
    • iii) placing the cell in conditions for supporting the transformation of the desired cell and maintaining intracellular levels of the at least one reprogramming agent for a sufficient period of time to allow stable expression of the at least one gene regulator in absence of the reprogramming agent; and
    • iv) maintaining the cell in culture conditions supporting the transformation of the desired cell for a sufficient period of time to allow a stable expression of a plurality of secondary genes whose expression is characteristic of phenotypical and functional properties of the desired cell. At least one of the stably expressed secondary genes is not characteristic of phenotypical and functional properties of an embryonic stem cell. At the end of said period of time the cell of the first type has been transformed into the desired cell of a different type. Preferably, the cell of a different type obtained after the transformation is further characterized by a stable repression of a plurality of genes expressed in the first cell type.


According to various embodiments, step iii) may be carried out consecutively to step ii), simultaneously with step ii), or before step ii).


According to a related aspect, the invention relates to a process wherein a cell of a first type is reprogrammed to a desired cell of a different type, the process comprising:

    • a transient increase of intracellular levels of at least one reprogramming agent, wherein the at least one reprogramming agent induces a direct or indirect endogenous expression of at least one gene regulator, wherein the endogenous expression of the at least one gene regulator is necessary for the existence of the desired cell of a different type;
    • a stable expression of said at least one gene regulator;
    • stable expression of a plurality of secondary genes, wherein the stable expression of the plurality of secondary genes is the result of the stable expression of the at least one gene regulator, and wherein: (i) stable expression of the plurality of secondary genes is characteristic of phenotypical and/or functional properties of the desired cell, (ii) stable expression of at least one of the secondary genes is not characteristic of phenotypical and functional properties of an embryonic stem cell, and wherein (i) and (ii) are indicative of successful reprogramming of the cell of the first type to the desired cell of the different type.


As used herein, “transiently increasing” refers to an increase that is not necessarily permanent and therefore, which may decrease or disappear over time. For instance, when referring to transiently increasing intracellular levels of at least one reprogramming agent in a cell, it means that the increase in present for a sufficient period of time for causing particular cellular events to occur (e.g. inducing stable endogenous expression of a gene regulator).


Typically a transient increase is not permanent and is not associated for instance to genome integration of an expression vector.


As used herein the term “reprogramming agent” refers to a compound that is capable of inducing directly or indirectly the expression of morphological and/or functional characteristics of the desired cell of a different type. Preferred compounds include those capable of driving directly or indirectly transformation of the cell of the first type into the desired cell of a different type. In preferred embodiment, the reprogramming agent is selected for inducing a direct or indirect endogenous expression of at least one gene regulator as defined herein. There are many compounds that may be helpful in reprogramming a cell according to the invention and these compounds can be used alone or in combinations. In various embodiments, the reprogramming agent is a polynucleotide or polypeptide selected according to TABLE A:











TABLE A








Reprogramming agent















RefSeq/







GenBank ™
UniProt ™ /
UniGene ™





(NCBI) Access.
Swiss-Prot
Accession




Name
No.
Access. No.
No.
Markers





Examples of







Desired Cell







Type







Pluripotent-like
AGR2
NM_006408.3
O95994
Hs.530009
OCT4


cells
AGR3
NM_176813.3
Q8TD06
Hs.100686
Nanog



BRIX1
NM_018321.3
Q8TDN6
Hs.718510
SSEA-4



CRABP2
NM_001878.2
P29373
Hs.405662
TRA1-60



DNMT3B,
NM_006892.3
Q9UBC3
Hs.713611
TRA1-80



isoform 1







DNMT3B,
NM_175848.1
Q9UBC3
Hs.713611
AP



isoform 2







DNMT3B,
NM_175849.1
Q9UBC3
Hs.713611




isoform 3







DNMT3B,
NM_175850.1
Q9UBC3
Hs.713611




isoform 6







DPPA2
NM_138815.3
Q7Z7J5
Hs.351113




DPPA3
NM_199286.2
Q6W0C5
Hs.131358




(STELLA)







DPPA4
NM_018189.3
Q7L190
Hs.317659




DPPA5
NM_001025290.1
A6NC42
Hs.125331




(ESG1)







FOXD3
NM_012183.2
Q9UJU5
Hs.546573




FOXH1
NM_003923.2
O75593
Hs.708365




GABRB3,
NM_000814.5
P28472
Hs.302352




isoform 1







GABRB3,
NM_021912.4
P28472
Hs.302352




isoform 2







GABRB3,
NM_001191320.1
P28472
Hs.302352




isoform 3







GABRB3,
NM_001191321.1
P28472
Hs.302352




isoform 4







GBX2
NM_001485.2
P52951
Hs.184945




GDF3
NM_020634.1
Q9NR23
Hs.86232




GJA1 (CX43)
NM_000165.3
P17302
Hs.74471




GRB7
NM_005310.2
Q14451
Hs.86859





NM_001030002.1
Q14451
Hs.86859




HESRG
NR_027122.1
Q1W209
Hs.720658




IFITM1
NM_003641.3
P13164
Hs.458414




IFITM2
NM_006435.2
Q01629
Hs.709321




KLF2
NM_016270.2
Q9Y5W3
Hs.726356




KLF4
NM_004235.4
O43474
Hs.376206




LEFTY1
NM_020997.2
O75610
Hs.656214




LEFTY2
NM_003240.3
O00292
Hs.520187




(EBAF),







isoform 1







LE FTY2
NM_001172425.1
B4E332
Hs.520187




(EBAF),

(TrEMBL)





isoform 2







LIN28A
NM_024674.4
Q9H9Z2
Hs.86154




MYBL2
NM_002466.2
P10244
Hs.179718




NANOG
NM_024865.2
Q9H9S0
Hs.635882




NODAL
NM_018055.4
Q96S42
Hs.370414




NOG
NM_005450.4
Q13253
Hs.248201




NR0B1
NM_000475.4
P51843
Hs.268490




(DAX1)







NR5A2,
NM_205860.1
O00482
Hs.33446




isoform 1







NR5A2,
NM_003822.3
O00482
Hs.33446




isoform 2







NR6A1,
NM_033334.2
Q15406
Hs.586460




isoform 1







NR6A1,
NM_001489.3
Q15406
Hs.586460




isoform 2







PHC1
NM_004426.2
P78364
Hs.305985




PITX2,
NM_153427.1
Q99697
Hs.643588




isoform a







PITX2,
NM_153426.1
Q99697
Hs.643588




isoform b







PITX2,
NM_000325.5
Q99697
Hs.643588




isoform c







PODXL,
NM_001018111.2
O00592
Hs.726449




isoform 1







PODXL,
NM_005397.3
O00592
Hs.726449




isoform 2







POU5F1
NM_002701.4
Q01860
Hs.249184




(OCT4),







isoform 1*







POU5F1
NM_203289.4
N/A
Hs.249184




(OCT4),
NM_001173531.1






isoform 2







PTEN
NM_000314.4
P60484
Hs.500466




REST
NM_005612.4
Q13127
Hs.307836





NM_001193508.1
Q13127
Hs.307836




REX1
NM_020695.3
Q8N1G1
Hs.192477




SALL4
NM_020436.3
Q9UJQ4
Hs.517113




SEMA3A
NM_006080.2
Q14563
Hs.252451




SFRP2
NM_003013.2
Q96HF1
Hs.481022




SOX2
NM_003106.2
P48431
Hs.518438




TDGF1,
NM_003212.3
P13385
Hs.385870




isoform 1







TDGF1,
NM_001174136.1
P13385
Hs.385870




isoform 2







TERT,
NM_198253.2
O14746
Hs.492203




isoform 1







TERT,
NM_001193376.1
O14746
Hs.492203




isoform 2







TPT1
NM_003295.2
P13693
Hs.374596




UTF1
NM_003577.2
Q5T230
Hs.458406




ZFP42
NM_174900.3
Q96MM3
Hs.335787



Ectoderm-like
ASCL1
NM_004316.3
P50553
Hs.703025
FoxJ3


cells
(MASH1)







CDX1
NM_001804.2
P47902
Hs.1545
Otx2



DLX3
NM_005220.2
O60479
Hs.134194
E-cadherin



DLX5
NM_005221.5
P56178
Hs.99348
TP73L



FOXD3
NM_012183.2
Q9UJU5
Hs.546573




MSI1
NM_002442.2
O43347
Hs.158311




NANOG
NM_024865.2
Q9H9S0
Hs.635882




POU5F1
NM_002701.4
Q01860
Hs.249184




(OCT4),







isoform 1*







POU5F1
NM_203289.4
N/A
Hs.249184




(OCT4),
NM_001173531.1






isoform 2







SOX1
NM_005986.2
O00570
Hs.202526




SOX2
NM_003106.2
P48431
Hs.518438




SP8, isoform
NM_182700.4
Q8IXZ3
Hs.195922




1







SP8, isoform
NM_198956.2
N/A
Hs.195922




2







ZIC1
NM_003412.3
Q15915
Hs.647962



Mesendoderm-
EOMES
NM_005442.2
O95936
Hs.591663
Mixl1


like cells
FOXA2,
NM_021784.4
Q9Y26I
Hs.155651
Mesp1



isoform 1*







FOXA2,
NM_153675.2
Q9Y26I
Hs.155651
Bry



isoform 2







FOXD3
NM_012183.2
Q9UJU5
Hs.546573
Flk1



GATA4
NM_002052.3
P43694
Hs.243987
Pax2



GATA6
NM_005257.3
Q92908
Hs.514746
Six1



MIXL1
NM_031944.1
Q9H2W2
Hs.282079




POU5F1
NM_002701.4
Q01860
Hs.249184




(OCT4),







isoform 1*







POU5F1
NM_203289.4
N/A
Hs.249184




(OCT4),
NM_001173531.1






isoform 2







SOX17
NM_022454.3
Q9H6I2
Hs.98367




T (Brachyury)
NM_003181.2
O15178
Hs.389457



Desired







second cell







type







Neural stem-
CALB1
NM_004929.2
P05937
Hs.65425
Sox2


like cells
DLL1
NM_005618.3
O00548
Hs.379912
Nestin



DLX1,
NM_178120.4
P56177
Hs.407015
GFAP



isoform 1







DLX1,
NM_001038493.1
P56177
Hs.407015
Msi1



isoform 2







DLX2
NM_004405.3
Q07687
Hs.419
Sox1



FOXD3
NM_012183.2
Q9UJU5
Hs.546573
CD133



GJD2 (CX36)
NM_020660.1
Q9UKL4
Hs.283816




HES1
NM_005524.2
Q14469
Hs.250666




HES3
NM_001024598.3
Q5TGS1
Hs.532677




HES5
NM_001010926.3
Q5TA89
Hs.57971




HOXB1
NM_002144.3
P14653
Hs.99992




MNX1 (HB9),
NM_005515.3
P50219
Hs.37035




isoform 1







MNX1 (HB9),
NM_001165255.1
N/A
Hs.37035




isoform 2







MSI1
NM_002442.2
O43347
Hs.158311




NANOG
NM_024865.2
Q9H9S0
Hs.635882




NEUROD1
NM_002500.2
Q13562
Hs.709709




NEUROG1
NM_006161.2
Q92886
Hs.248149




NEUROG2
NM_024019.2
Q9H2A3
Hs.567563




NKX6.1
NM_006168.2
P78426
Hs.546270




PAX6,
NM_000280.3
P26367
Hs.270303




isoform a*







PAX6,
NM_001127612.1
P26367
Hs.270303




isoform a







PAX6,
NM_001604.4
P26367
Hs.270303




isoform b







SFRP2
NM_003013.2
Q96HFI
Hs.481022




SIX3
NM_005413.3
O95343
Hs.567336




SOX1
NM_005986.2
O00570
Hs.202526




SOX2
NM_003106.2
P48431
Hs.518438



Cardiac
BAF60C
NM_001003802.1
Q6STE5
Hs.647067
MLc2α


progenitor-like
(SMARCD3),






cells
isoform 1







BAF60C
NM_003078.3
Q6STE5
Hs.647067
Nkx2.5



(SMARCD3),







isoform 1







BAF60C
NM_001003801.1
Q6STE5
Hs.647067
Isl+



(SMARCD3),







isoform 2*







FOXD3
NM_012183.2
Q9UJU5
Hs.546573
Bry



GATA4
NM_002052.3
P43694
Hs.243987




GATA6
NM_005257.3
Q92908
Hs.514746




HANDI
NM_004821.2
O96004
Hs.152531




HAND2
NM_021973.2
P61296
Hs.388245




ISL1
NM_002202.2
P61371
Hs.505




KDR
NM_002253.2
P35968
Hs.479756




MESP1
NM_018670.3
Q9BRJ9
Hs.447531




MYOCD,
NM_001146312.1
Q6N065
Hs.567641




isoform 1

(TrEMBL)





MYOCD,
NM_153604.2
Q81ZQ8
Hs.567641




isoform 2







MYOCD,
NM_001146313.1
Q81ZQ8
Hs.567641




isoform 3







NKX2.5,
NM_004387.3
P52952
Hs.54473




isoform 1*







NKX2.5,
NM_001166175.1
P52952
Hs.54473




isoform 2*







NKX2.5,
NM_001166176.1
P52952
Hs.54473




isoform 3*







T (Brachyury)
NM_003181.2
O15178
Hs.389457




TBX5, isoform
NM_000192.3
Q99593
Hs.381715




1*







TBX5, isoform
NM_181486.1
Q99593
Hs.381715




1







TBX5, isoform
NM_080718.1
Q99593
Hs.381715




2







TBX5, isoform
NM_080717.2
Q99593
Hs.381715




3







SOX17
NM_022454.3
Q9H612
Hs.98367



Pancreatic
FOXA2,
NM_021784.4
Q9Y261
Hs.155651
PDX1


progenitor-like
isoform 1*






cells
FOXA2,
NM_153675.2
Q9Y261
Hs.155651
Sox17



isoform 2







FOXD3
NM_012183.2
Q9UJU5
Hs.546573
FoxA2



MAFA
NM_201589.2
Q8NHW3
Hs.670866
Ngn3



MIXL1
NM_031944.1
Q9H2W2
Hs.282079
IsI1



NEUROG3
NM_020999.3
Q9Y4Z2
Hs.532682




NKX6.1
NM_006168.2
P78426
Hs.546270




PAX4
NM_006193.2
O43316
Hs.129706




PDX1
NM_000209.3
P52945
Hs.32938




SOX17
NM_022454.3
Q9H612
Hs.98367



Myogenic
FOXC1
NM_001453.2
Q12948
Hs.348883
SMα actin


progenitor-like
FOXC2
NM_005251.2
Q99958
Hs.436448
Calponin


cells
MEF2C,
NM_002397.4
Q06413
Hs.649965
MyoD



isoform 1
NM_001193350.1
Q06413





MEF2C,
NM_001131005.2
Q06413
Hs.649965
MEF2C



isoform 2







MEF2C,
NM_001193347.1
Q06413
Hs.649965
Pax3



isoform 3







MEF2C,
NM_001193348.1
Q06413
Hs.649965
Pax7



isoform 4







MEF2C,
NM_001193349.1
Q06413
Hs.649965




isoform 5







Pax3, isoform
NM_181457.3
P23760
Hs.42146




Pax3







Pax3, isoform
NM_000438.5
P23760
Hs.42146




Pax3a







Pax3, isoform
NM_013942.4
P23760
Hs.42146




Pax3b







Pax3, isoform
NM_181458.3
Q494Z3, Q494Z4
Hs.42146




Pax3d

(TrEMBL)





Pax3, isoform
NM_181459.3
Q494Z3, Q494Z4
Hs.42146




Pax3e

(TrEMBL)





Pax3, isoform
NM_181461.3
Q494Z3, Q494Z4
Hs.42146




Pax3g

(TrEMBL)





Pax3, isoform
NM_ 181460.3
Q494Z3, Q494Z4
Hs.42146




Pax3h

(TrEMBL)





Pax3, isoform
NM_ 001127366.2
Q494Z4
Hs.42146




Pax3i

(TrEMBL)





PAX7,
NM_ 002584.2
P23759
Hs.113253




isoform 1







PAX7,
NM_ 013945.2
P23759
Hs.113253




isoform 2







PAX7,
NM_ 001135254.1
P23759
Hs.113253




isoform 3









In some embodiments, the reprogramming agent is a polypeptide which shares at least 75%, 80%, 85%, 90%, 95%, 97%, 99% or more of the functionality or sequence identity of any one of the reprogramming agents in the table hereinbefore.


Identifying the “sufficient period of time” to allow stable expression of the at least one gene regulator in absence of the reprogramming agent and the “sufficient period of time” in which the cell is to be maintained in culture conditions supporting the transformation of the desired cell is within the skill of those in the art. The sufficient or proper time period will vary according to various factors, including but not limited to, the particular type and epigenetic status of cells (e.g. the cell of the first type and the desired cell), the amount of starting material (e.g. the number of cells to be transformed), the amount and type of reprogramming agent(s), the gene regulator(s), the culture conditions, presence of compounds that speed up reprogramming (ex, compounds that increase cell cycle turnover, modify the epigenetic status, and/or enhance cell viability), etc. In various embodiments the sufficient period of time to allow a stable expression of the at least one gene regulator in absence of the reprogramming agent is about 1 day, about 2-4 days, about 4-7 days, about 1-2 weeks, about 2-3 weeks or about 3-4 weeks. In various embodiments the sufficient period of time in which the cells are to be maintained in culture conditions supporting the transformation of the desired cell and allow a stable expression of a plurality of secondary genes is about 1 day, about 2-4 days, about 4-7 days, or about 1-2 weeks, about 2-3 weeks, about 3-4 weeks, about 4-6 weeks or about. 6-8 weeks. In preferred embodiments, at the end of the transformation period, the number of transformed desired cells is substantially equivalent or even higher than an amount of cells a first type provided at the beginning.


The present invention encompasses various types of compounds that are suitable for increasing in a cell of a first type the intracellular levels of at least one reprogramming agent. Preferably, the compound should also be able to directly or indirectly remodel the chromatin and/or DNA of the cell, thus resulting directly or indirectly in the expression of morphological and functional characteristics of the desired cell of a different type. Preferred compounds are reprogramming agents as defined herein or any other compound having a similar activity and having the ability to activate or enhance the expression of the endogenous version of genes listed in the table of reprogramming agents hereinbefore and which are capable of driving directly or indirectly transformation of the cell of the first type into the desired cell of a different type.


As will be explained hereinafter, the increase in intracellular levels of the at least one reprogramming agent can be achieved by different means. In preferred embodiments the reprogramming agent is a polypeptide and increasing intracellular levels of such polypeptide include transfection (or co-transferction) of an expression vector having a polynucleotide (ex. DNA or RNA) encoding the polypeptide(s), or by an intracellular delivery of polypeptide(s). According to the invention, transient expression is generally preferable. Additional suitable compounds may include compounds capable of increasing the expression of the endogenous version of genes listed in the table of reprogramming agents and gene regulators including, but not limited to, reprogramming factors listed in Table B.










TABLE B





Desired cell of different type
Reprogramming Factor







Pluripotent-like cells
Nodal, ActivinA, Fgf-2, Wnt3a, L-Ascorbic Acid, BIO,



CHIR99021, PD0325901, Thiazovivin, SB431542, Cyclic



Pifithrin-a, Tranylcypromine hydrochloride, Kenpaullone, 5-



Azacytidine, Valproic Acid, BIX01294, R(+)BayK8644,



RG108, Theanine, Sodium butyrate


Ectoderm like cells:
a retinoid compound, L-Ascorbic acid, SHH, Wnt 3a, a


1- Neural stem-like cells
neurotrophic factor, bFGF, EGF, Transforming growth factor



alpha, neuropeptide Y, Estrogen, Noggin, Forskolin, 5-



Azacytidine, Valproic Acid, BIX01294, R(+)BayK8644,



RG108, Sodium butyrate, Lithium


Mesoendoderm like cells:
BMP4, Epidermal growth factor-Cripto/FRL-1/Cryptic



(EGF-CFC) and the TGFβs, Activin, Nodal, SHH,



Vg1/GDF1 (growth and differentiation factor-1)


1- Cardiac progenitor-like
1- BMP4, bFGF, Activin A, VEGF, DKK1 (dickkopf


cells
homologue 1), Insulin-like growth factor 1 (IGF-1) and



hepatocyte growth factor (HGF), 5-Azacytidine,



Valproic Acid, BIX01294, R(+)BayK8644, RG108,



Cardiogenol C hydrochloride, Sodium butyrate


2- Pancreatic progenitor-
2- Activin A, GLP-1, bFGF, Reg1, nicotinamide,


like cells
Betacellulin, SHH, (−)-Indolactam V, a retinoid



compound, Cyclopamine, IDE-1 and 2, 5-Azacytidine,



Valproic Acid, BIX01294, R(+)BayK8644, RG108,



Sodium butyrate


3- Myogenic progenitor-
3- retinoic acid, HGF, FGF, IGF, transforming growth


like cells
factor-beta, Wnt3a, 5-Azacytidine, Valproic Acid,



BIX01294, R(+)BayK8644, RG108, Sodium butyrate









According to the principles of the invention, increasing intracellular levels of at least one reprogramming agent should induce a direct or indirect endogenous expression of at least one gene regulator. As used herein, “gene regulator” refers to a polynucleotide or polypeptide whose expression is associated with a series of intracellular events leading to the transformation of a given cell of a first type into a pluripotent, multipotent and/or unipotent cell. Typically expression of a gene regulator directly or indirectly activates genes necessary for the phenotypical and functional characteristic of pluripotent, multipotent and/or unipotent cells, while repressing genes of the cell of a first type. The gene regulator may be the same or be different than the reprogramming agent. Examples of gene regulators according to the invention include, but are not limited to, the polynucleotides and polypeptides listed herein before in TABLE A.


In some embodiments, the gene regulator is a polypeptide which shares at least 75%, 80%, 85%, 90%, 95%, 97%, 99% or more of the functionality or sequence identity of any one of the gene regulators provided in the Table A hereinbefore.


As used herein, “conditions supporting growth” or “conditions supporting the transformation” when referring to a desired cell refers to various suitable culture conditions (temperature, pH, O2 tension, cell media, factors, compounds, growth substrate (ex. laminin, collagen, fibronectin, Matrigel™, low-bind surface, nanostructured or charged surface, etc.), 3D environment, etc.) favorising growth of the desired cell type and/or favorising transformation towards such desired cell type. Those skilled in the art known that growth or transformation of particular cell types is stimulated under specific conditions, while inhibited by others, and it is within their skill to select suitable conditions (e.g. culture conditions) favorising growth or transformation of desired cell types.


The terms “phenotypical and functional properties”, when referring to a desired cell or to an embryonic stem cell, means the biological, biochemical, physiological and visual characteristics of a cell, including expression of certain genes and cell surface markers, which can be measured or assessed for confirming its identity or function(s).


An example of a suitable reprogramming agent according to preferred embodiments of the invention is MUSASHI1. In some embodiments this polypeptide is preferred for driving a first cell, such as a fibroblast, into a Neural Stem-Like Cell (NSLC). In other embodiments, the at least one reprogramming agent which said intracellular levels is increased is(are) either Musashi1 (Msi1) alone; Musashi1 (Msi1) and Neurogenin 2 (Ngn2); Musashi1 (Msi1) and methyl-CpG binding domain protein 2 (MBD2); or Neurogenin 2 (Ngn2) and methyl-CpG binding domain protein 2 (MBD2). Adequate intracellular levels of these polypeptides are preferred since they tend to be expressed throughout an entire cell lineage, from as early as embryonic stem cells (or even earlier) to pre-somatic cells (or even later).


MBD2 is a member of a family of methyl-CpG-binding proteins that has been reported to be both a transcriptional repressor and a DNA demethylase (dMTase). As used herein, the term “MBD2” generally refers to the human methyl-CpG binding domain protein 2. The GeneBank™ (NCBI) accession number of human MBD2 is NM_003927.3/AF072242, the UniProt™ accession number is NP-003918/Q9UBB5 and the UniGene™ accession number is Hs.25674.


As used herein, the term “Msi1” generally refers to the human musashi homolog 1. The GeneBank™ (NCBI) accession number of human Msi1 is NM_002442.2/AB012851, the UniProt™ accession number is NP-002433/043347 and the UniGene™ accession number is Hs.158311.


As used herein, the term “Ngn2” generally refers to the human neurogenin 2. The GeneBank™ (NCBI) accession number of human Ngn2 is NM_024019.2/BC036847, the UniProt™ accession number is NP-076924/Q9H2A3 and the UniGene™ accession number is Hs.567563.


According to additional aspects, the method of transforming a cell of a first type to a desired cell of a different type comprises the steps of either:

    • 1) contacting the cell of a first type with one or more compounds capable of increasing intracellular levels of at least one reprogramming agent within the cell and directly or indirectly remodeling the chromatin and/or DNA of the cell; or
    • 2) contacting the chromatin and/or DNA of a cell of a first type with an agent capable of remodeling the chromatin and/or DNA of the cell; and increasing intracellular levels of at least one reprogramming agent.


According to various embodiments, step 2) may be carried out consecutively to step 1), simultaneously with step 1), or before step 1).


According to a particular aspect, the invention relates to a method for obtaining a Neural Stem-Like Cell (NSLC), comprising:

    • providing a cell of a first type which is not a NSLC;
    • increasing intracellular levels of at least one neural stem cell specific polypeptide, wherein the polypeptide is capable of driving directly or indirectly transformation of the cell of the first type into a NSLC; and
    • contacting chromatin and/or DNA of a cell of a first type with a histone acetylator, an inhibitor of histone deacetylation, a DNA demethylator, and/or a chemical inhibitor of DNA methylation.


With respect to the second step, the term “remodelling the chromatin and/or DNA” refers to dynamic structural changes to the chromatin. These changes can range from local changes necessary for transcriptional regulation, to global changes necessary for opening up the chromatin structure or chromosome segregation to allow transcription of the new set of genes characteristic of the desired cell of a different type, to closing up of the chromatin structure or chromosome segregation to prevent transcription of certain genes that are not characteristic of the desired cell of a different type. In some embodiments, opening up of the chromatin structure refers more specifically to acetylation of histones, and demethylation of DNA, while closing up of the chromatin structure refers more specifically to deacetylation of histones, and methylation of DNA.


As used herein, “compound” refers to a compound capable of effecting a desired biological function. The term includes, but is not limited to, DNA, RNA, protein, polypeptides, and other compounds including growth factors, cytokines, hormones or small molecules. As used herein, compounds capable of remodeling chromatin and/or DNA include, but are not limited to, histone acetylators, inhibitors of histone deacetylation, DNA demethylators, inhibitors of DNA methylation and combination thereof.


“Inhibitor of DNA methylation” refers to an agent that can inhibit DNA methylation. DNA methylation inhibitors have demonstrated the ability to restore suppressed gene expression. Suitable agents for inhibiting DNA methylation include, but are not limited to 5-azacytidine, 5-aza-2-deoxycytidine, 1-β-D-arabinofuranosil-5-azacytosine, and dihydro-5-azacytidine, and zebularine (ZEB), BIX (histone lysine methytransferase inhibitor), and RG108.


“Inhibitor of histone deacetylation” refers to an agent that prevents the removal of the acetyl groups from the lysine residues of histones that would otherwise lead to the formation of a condensed and transcriptionally silenced chromatin. Histone deacetylase inhibitors fall into several groups, including: (1) hydroxamic acids such as trichostatin (A), (2) cyclic tetrapeptides, (3) benzamides, (4) electrophilic ketones, and (5) aliphatic acid group of compounds such as phenylbutyrate and valporic acid. Suitable agents to inhibit histone deacetylation include, but are not limited to, valporic acid (VPA), phenylbutyrate Trichostatin A (TSA), Na-butyrate, and benzamides. VPA promotes neuronal fate and inhibits glial fate simultaneously through the induction of neurogenic transcription factors including NeuroD.


“Histone Acetylator” refers to an agent that inserts acetyl groups to the lysine residues of histones that opens up the chromatin and turns it into a transcriptionally active state. Suitable Histone Acetylator agents include, but are not limited to, Polyamine, CREB (cAMP element binding protein), and BniP3.


“DNA demethylator” refers to an agent that removes the methyl groups from DNA and possesses the ability to inhibit hypermethylation and restore suppressed gene expression. A demethylase is expected to activate genes by removing the repressive methyl residues. Suitable DNA demethylators include, but are not limited to, MBD2 and Gadd45b.


In some embodiments, the reprogramming agent has one or more of the following functions: it decrease the expression of one or more markers of cells of the first type (ex. see Table C), and/or increase the expression of one or more markers of the desired cell of the different type (ex. see Table A). Cells that exhibit a selectable marker for the desired cell of a different type are then selected and assessed for characteristics of the desired cell of a different type.


According to the invention, transformation into the desired cell results in stable expression of a plurality of secondary genes whose expression is characteristic of phenotypical and/or functional properties of the desired cell. Genes whose expression is characteristic of phenotypical and/or functional properties of the desired cell include, but is not limited to, those listed in Table A.


In some embodiments, expression of secondary genes whose expression is characteristic of phenotypical and functional properties of the desired cell results in the expression of markers defined according to the following table:
















Desired cell type
Markers









Neural stem-like cells
Nestin, Sox2, GFAP, Msi1



Neural-like cells
βIII-tubulin, Map2b, Synapsin, ACHE



Ectoderm-like cells
Sox2, Sox1, Zic1, Nestin, Notch 1, FoxJ3,




Otx2, Cripto1, Vimentin



Mesendoderm-like cells
Sox17, FoxA2, CXCR4, GATA4, Mixl1,




Eomesodermin



Pluripotent-like cells
Oct4, SSEA4, TRA-1-60, TRA-1-81, AP










In some embodiments, transformation of a cell of a first type into the desired cell results in a stable repression of a plurality of genes typically expressed in the cell of the first type. Examples of such suppressed genes include, but are not limited to, those defined in Table C:









TABLE C







Examples of suppressed genes










Cell-type specific genes typically repressed during




Reprogramming















RefSeq/







GenBank ™
UniProt ™ /
UniGene™





(NCBI) Accession
Swiss-Prot
Accession



Cell Type
Name
No.
Accession No.
No.
Markers





Keratinocytes
TP63,
NM_003722.4
Q9H3D4
Hs.137569
Keratin 14



isoform 1







TP63,
NM_001114978.1
Q9H3D4
Hs.137569
Basonuclin



isoform 2







TP63,
NM_001114979.1
Q9H3D4
Hs.137569
P63



isoform 3







TP63,
NM_001114980.1
Q9H3D4
Hs.137569




isoform 4







TP63,
NM_001114981.1
Q9H3D4
Hs.137569




isoform 5







TP63,
NM_001114982.1
Q9H3D4
Hs.137569




isoform 6







BNC1
NM_001717.3
Q01954
Hs.459153




BCN2
NM_017637.5
Q6ZN30
Hs.656581




KRT14
NM_000526.4
P02533
Hs.654380




Involucrin
NM_005547.2
P07476
Hs.516439



Fibroblasts
THY1
NM_006288.3
P04216
Hs.724411
Col5A2



FBN2
NM_001999.3
P35556
Hs.519294
Fibronectin



COL5A2
NM_000393.3
P05997
Hs.445827




DNMT1,
NM_001130823.1
P26358
Hs.202672




isoform a







DNMT1,
NM_001379.2
P26358
Hs.202672




isoform b






CD34+
Isl1
NM_002202.2
P61371
Hs.505
VEGFR



HOXA9
NM_152739.3
P31269
Hs.659350
Cytokeratin



HOXB4
NM_024015.4
P17483
Hs.664706




Klk-1
NM_002257.2
P06870
Hs.123107
CD34



Bry
NM_003181.2
O15178
Hs.389457



Adipose-
ALCAM
NM_001627.2
Q13740
Hs.591293
ALBO


derived stem
VCAM-1
NM_001078.2
P19320
Hs.109225
Adiponectin


cells (ADSC)
VCAM-1,
NM_080682.1
P19320
Hs.109225




isoform b







PROM1,
NM_006017.2
O43490
Hs.614734
Leptin



isoform 1







PROM1,
NM_001145847.1
O43490
Hs.614734




isoform 2
NM_001145848.1






PROM1,
NM_001145852.1
O43490
Hs.614734




isoform 4







PROM1,
NM_001145851.1
O43490
Hs.614734




isoform 5







PROM1,
NM_001145850.1
O43490
Hs.614734




isoform 6







PROM1,
NM_001145849.1
O43490
Hs.614734




isoform 7







FUT4
NM_002033.3
P22083
Hs.390420









In preferred embodiments, stable repression of any one or more of the genes listed in Table C being expressed in the first cell type is also characterized by a disappearance of the corresponding markers (see Table C).


Those skilled in the art will understand that there exist many alternative steps for facilitating cell reprogramming. Those include destabilizing the cell's cytoskeletal structure (for example, by exposing the cell to cytochalasin B), loosening the chromatin structure of the cell (for example, by using agents such as 5 azacytidine (5-Aza) and Valproic acid (VPA) or DNA demethylator agents such as MBD2), transfecting the cell with one or more expression vector(s) containing at least one cDNA encoding a neurogenic transcription factor (for example, Msi1 or Ngn2), using an appropriate medium for the desired cell of a different type and an appropriate differentiation medium to induce differentiation commitment of the desired cell of a different type, inhibiting repressive pathways that negatively affects induction into commitment the desired cell of a different type, growing the cells on an appropriate substrate for the desired cell of a different type (for example, laminin for NSLCs or a low-bind surface for culturing floating neurospheres), and growing the cells in an environment that the desired cell of a different type (or “-like” cell) would be normally exposed to in vivo such as the proper temperature, pH and low oxygen environment (for example about 2-5% O2). In various embodiments, the invention encompasses these and other related methods and techniques for facilitating cell reprogramming.


Accordingly, the method of transforming a cell of a first type into a cell of a second different type may comprise additional facultative steps. In one embodiment, the method of transforming a cell further comprises the step of pretreating the cell of a first type with a cytoskeleton disruptor. As used herein “cytoskeleton” refers to the filamentous network of F-actin, Myosin light and heavy chain, microtubules, and intermediate filaments (IFs) composed of one of three chemically distinct subunits, actin, tubulin, or one of several classes of IF protein. Accordingly, the term “cytoskeleton disruptor” refers to any molecules that can inhibit the cell cytoskeleton to destabilize the cell and consequently remove the feedback mechanisms between the cell's shape and cellular and nuclear function. Suitable cytoskeleton disruptor according to the invention include, but are not limited to, the cytochalasin family of actin cytoskeleton inhibitors, such as Cytochalasin B or D, and myosin inhibitors such as 2,3-butanedione monoxime. Such pretreatment may boost reprogramming. In a preferred embodiment, the cell is cultured in the presence of at least one cytoskeleton inhibitor one day before, during, or after introducing a neurogenic transcription factor(s).


Placing the cell in conditions in conditions for supporting the transformation of the desired cell, and/or maintaining the cell in culture conditions supporting the transformation of the desired cell may comprises culturing the cell in a media comprising one or more factors appropriate for inducing the expression of the morphological and functional characteristics of the desired desired cell of a different type. In some embodiments the one or more factors are reprogramming factors helpful in reprogramming a cell and these reprogramming factors can be used alone or in combinations.


In other embodiments, the step of culturing the cell in a media comprising one or more factors appropriate for inducing the expression of the morphological and functional characteristics of the desired cell of a different type is carried out subsequently or simultaneously to steps iii) or iv), or subsequently or simultaneously to steps 1) or 2), as defined hereinbefore.


Those skilled in the art know many different types of media and many reprogramming factors that may be helpful in reprogramming a cell and these reprogramming factors can be used alone or in combinations. In various embodiments, the reprogramming factor is selected according to TABLE B.


In some embodiments, reprogramming factors have one or more of the following functions: decrease the expression of one or more markers of the first type of cell and/or increase the expression of one or more markers of the desired cell. Cells that exhibit a selectable marker for the desired cell are then selected and assessed for unipotency, multipotency, pluripotency, or similar characteristics (as appropriate).


In particular embodiments, the cells are cultured in serum-free medium before, during or after any one of steps i) to iv) as defined hereinbefore, or during or after steps 1) or 2), as defined hereinbefore.


In some embodiments, Mesendoderm-like cells can be created from cells such as ADSCs by up-regulating the expression of FoxD3, MixI1, Ngn3 and MBD2. Additionally up-regulating the expression of Oct4, Sox17, Brachyury, and/or FoxA2 can be used to create Mesendoderm-like cells. In addition, media compositions detailed in Table 37 can be used to create mesendoderm-like cells from adipocyte-derived stem cells (ADSCs) and similar cells, along with the use of proper cell substrates for mesendoderm cells known in the art (e.g., gelatin-coated plates).


In some embodiments, cells such as ADSCs can be reprogrammed into pancreatic progenitor-like cells and β islet-like cells by up-regulating the expression of Sox17, Pdx1 Ngn3, and Oct4 (e.g., Oct4+Sox17+Pdx1+Ngn3; Sox17+Pdx1+Ngn3; Oct4+Pdx1+Ngn3). Additionally up-regulating the expression of FoxA2 and MBD2 can be used to create these cells. In addition, media compositions detailed in Table 39 can be used to create pancreatic progenitor-like cells and β islet-like cells from adipocyte-derived stem cells (ADSCs) and similar cells, along with the use of proper cell substrates for growing pancreatic progenitor cells and β islet-like cells known in the art (e.g., fibronectin-coated collagen gels).


In some embodiments, cells such as ADSCs can be reprogrammed into cardiac progenitor-like cells and mesoderm-like cells by up-regulating the expression of a combination of Mesp1, FoxD3, Tbx5, Brachyury (T), Nkx2.5, Sox17 and/or Gata4 (e.g., Foxd3+Sox17+Mesp1+Tbx5; Foxd3+Sox17+Mesp1+Nkx2.5; Foxd3+T+Mesp1+Gata4), which increase the expression of mesoderm and cardiac progenitors markers. Additionally up-regulating the expression of Gata6 and Baf60c can be used to create these cells. In addition, media compositions detailed in Table 44 can be used to create cardiac progenitor-like cells from adipocyte-derived stem cells (ADSCs) and similar cells, along with the use of proper cell substrates for growing cardiac progenitor cells known in the art (e.g., matrigel).


In some embodiments, cells such as ADSCs can be reprogrammed into pluripotent-like stem cells up-regulating the expression of Rex1, Oct4 and Klf4, or Sall4, Oct4, Klf4 and Nanog. Reprogramming by transient transfection of a combination of the above genes can be achieved efficiently. These cells can be differentiated into ectoderm-like cells, endoderm-like cells, or mesendoderm-like cells by methods known in the art for differentiating pluripotent stem cells into these lineages. In addition, these pluripotent-like stem cells have protective and/or therapeutic/regenerative effect on other cells (e.g., hepatocytes).


The NSLCs that is a subject of this invention have benefits over native human neural stem/progenitor cells as well as embryonic stem cells. The NSLCs do not readily form tumors or teratomas (as tested in NOD-SCID mice), can be created from easily obtainable somatic cells (e.g., fibroblasts, CD34+ blood cells, keratinocytes) and differentiated towards any specific neuronal lineage, express neurotrophic growth factors (e.g., BDNF and GDNF), express some neuronal differentiation genes while maintaining a stem cell like state allowing a higher proportion of neuronal differentiation when placed in differentiation conditions (compared to native human neural stem/progenitor cells), form functional gap junctions and readily form synapses, and are capable of attaching and surviving on 3D scaffolds and environments.


The stem-like cells of the present invention have numerous advantages over the prior art such as efficient reprogramming without gene integration or constant artificial forced gene expression, greater potency and safety over native cells (esp. stem/progenitor cells), and having the capability to be the cell of interest for a particular application (e.g., a neural stem-like cell for CNS applications; a cardiac progenitor-like cell for cardiac applications, a pancreatic progenitor-like cells or (3 islet-like cells for diabetes, an ectoderm-like cell, a mesoderm-like cells, an endoderm-like cell, etc.). In addition derived cells of the invention can be a relatively homogeneous population of autologous cells of a particular phenotype of interest.


The methods of the present invention allow the ability to create the native types of stem cells for particular applications (e.g., neural stem-like cells for CNS applications; cardiac progenitor-like cells for cardiac applications, pancreatic progenitor-like cells or .beta.-like cells for diabetes, etc.) as well as creating autologous (from the patient's own cells) versions of these stem cells that allow them to graft more appropriately when delivered to the patient as a treatment or for augmenting the health or functionality of a particular tissue or organ, or for diagnostic purposes. The cells can also be used for modeling (e.g., disease modeling, or testing the effects of particular compounds or other molecules (e.g., for personalized medicine purposes)). The cells can also easily be enhanced by specific genes of interest or a defective gene repaired/replaced before delivery to the patient for treatment of a genetic disorder or for enhanced therapeutic value or augmenting the health or functionality of a particular cell, tissue, organ, system, or organism.


Obtaining Neural Stem-Like Cells (NSLCs)


According to preferred embodiments for creating Neural Stem-Like Cells (NSLCs), the methods of the invention are carried out such that cells are treated with selected agents, compounds and factors to promote the reprogramming and/or dedifferentiation towards Stem-Like Cells (SLCs). Such reprogrammed somatic cells can then be further treated with agents and/or cultured under conditions suitable for promoting reprogramming towards Neural Stem-Like Cells (NSLCs), and expansion of the NSLCs for the long-term. NSLCs according to the invention have the potential to differentiate to neuronal-like and/or glial-like cells, as well as neuronal and/or glial cells, for potential treatment of neurological diseases and injuries such as Parkinson's disease and spinal cord injury. The methods described herein are also useful for producing histocompatible cells for cell therapy.


Accordingly, some aspects of the present invention relates to generating neurons from an individual patient, thus making autologous transplantations possible as a treatment modality for many neurological conditions including neurotrauma, stroke, neurodegenerative diseases such as Multiple Sclerosis, Parkinson's disease, Huntington disease, Alzheimer's diseases. Thus, the invention provides for neurological therapies to treat the disease or trauma of interest.


Therefore, another aspect of the invention concerns a method of obtaining a Neural Stem-Like Cell (NSLC), comprising either:

    • 1) contacting the cell of a first type with one or more neural stem cell regulating polypeptide capable of increasing intracellular levels of neural stem cell specific polypeptides within said cell and directly or indirectly remodeling the chromatin and/or DNA of the cell and driving directly or indirectly transformation of the cell of the first type into a NSLC; or
    • 2) contacting the chromatin and/or DNA of a cell of a first type with a histone acetylator, an inhibitor of histone deacetylation, a DNA demethylator, and/or an inhibitor of DNA methylation; and increasing intracellular levels of at least one neural stem cell specific polypeptide driving directly or indirectly transformation of the cell of the first type into a NSLC.


In preferred embodiments, the step 1) comprises increasing intracellular levels of a MUSASHI1 polypeptide. As it will be explained hereinafter this can be achieved by different means including, but not limited to, transient expression of the MUSASHI1 polypeptide, preferably by transfecting an expression vector encoding the polypeptide.


In preferred embodiments, the step 2) comprises increasing intracellular levels of a MBD2 polypeptide or treating the cells with VPA and 5-AZA. As it will be explained hereinafter this can be achieved by different means including, but not limited to, transient expression of the MBD2 polypeptide, preferably by transfecting an expression vector encoding the polypeptide(s), and/or pre-treating and/or treating the cells with VPA and 5-AZA.


In one particular embodiment, reprogramming a cell of a first type to another type of cell that exhibits at least two selectable markers for neural stem cells requires transfecting the cell of a first type with one vector containing a cDNA encoding for a neurogenic transcription factor and one DNA demethylator. To enhance the de-differentiation the cells are exposed or pre-exposed to an agent(s) that inhibits DNA methylation, inhibits histone deacetylation, and/or disrupts the cell cytoskeleton. For example, the dedifferentiation can be enhanced by pre-treating the cells with an agent that disrupts the cell cytoskeleton followed by transfecting the cells with one or more vector(s) containing two neurogenic transcription factors in the presence of a DNA demethylator and/or inhibitor of DNA methylation and histone deacetylation. The histone deacetylator, inhibitor of histone deacetylation, DNA demethylator, and/or an inhibitor of DNA methylation are as defined previously.


As defined previously, the method may further comprise a preliminary step of pre-treating the cell of a first type with a cytoskeleton disruptor, as defined previously, and/or culturing the cell in a media comprising one or more reprogramming factors appropriate for appearance and maintenance of the morphological and functional characteristics of NSLCs as defined previously (e.g. a retinoid compound, a neurotrophic factor, bFGF, EGF, SHH, Wnt 3a, neuropeptide Y, Estrogen). In some embodiment the method further comprises inhibiting cellular BMP signaling pathways (e.g. by NOGGIN, fetuin, or follistatin).


In preferred embodiments, generation of a NSLC from a first cell comprises the use of one or more reprogramming agents. Suitable agents include, but are not limited to, Musashi-1 (Msi1) and Neurogenin 2 (Ngn2). Other potential agents are listed in Table A and B.


The present invention is also directed to the use of DNA expression vectors encoding a protein or transcript which upregulates the expression of neurogenesis. The genetically-engineered DNA sequence, encoding a defined reprogramming agent such as Msi1 and Ngn2, can be introduced into cells by using a mono-, bi-, or poly-cistronic vectors. The expression of an endogenous multipotency gene indicates that the cDNA encodes a protein whose expression in the cell result directly or indirectly in the de-differentiation of the cell. The newly de-differentiated mammalian cells are capable of re-differentiating to neuronal lineages to regenerate said mammalian cells, tissues, and organs.


The present invention is further directed to a method for generating NSLCs by introducing a genetically-engineered DNA sequence into human somatic cells via transient transfection. Since the DNA introduced in the transfection process is not inserted into the nuclear genome, the foreign DNA decreases over time and when the cells undergo mitosis. Nonviral vectors remain in a non-replicative form, have low immunogenicity, and are easy and safe to prepare and to use. Furthermore, plasmids may accommodate large fragments of DNA.


In one particular embodiment, the method starts with obtaining cells from the individual, and reprogramming the cells in vitro to generate NSLCs. The significant aspect of the present invention is the stable reprogramming of a somatic cell or non-neuronal cell into a NSLC that can give rise to different types of, neuronal or glial cells (including neuronal-like or glial-like cells). These can then be implanted back into the same patient from which the cells were obtained, thus making an autologous treatment modality for many neurological conditions including neurotrauma, stroke, and neurodegenerative disease possible. These can also be implanted into a different individual from which the cells were obtained. Accordingly, the cells and methods of the present invention may be helpful to treat, prevent, or to stabilize a neurological disease such as Alzheimer's disease, Parkinson's disease, multiple sclerosis, or spinal cord injury. This technology provides an ample source of neural stem cells, neuro-progenitor cells, neurons and glia for clinical treatment, which can be performed by implantation of NSLCs in vivo or inducing the differentiation in vitro and implantation of neuro-progenitor cells or specific neurons or glia in vivo.


In another embodiment, the method comprises isolating somatic or non-neuronal cells and exposing the cells to one or more agents that alter cell morphology and chromatin structure, and transfecting the cells with one or more genes containing at least one cDNA encoding for a neurogenic transcription factor. The gene transfection step may be replaced with alternative agents that induce the expression of the neurogenic transcription factor(s) in the cell. Inducing epigenetic modifications to DNA and histones (especially DNA demethylation and an open chromatic structure) facilitate true reprogramming of the cells. In another embodiment, the cells are incubated in a low oxygen environment, for example 5% O2, thereby helping in reprogramming the cells.


This methodology allows the reprogramming of a cell into a NSLC. The further course of development and the expansion of the reprogrammed cell depend on the in situ environment cues to which it is exposed. The embodiments of the invention further include growing the reprogrammed cell in an appropriate proliferation medium to expand the generated NSLC, for example Neural Progenitor proliferation Medium (StemCell Technologies) with the presence of epidermal growth factor (EGF) and basic fibroblast growth factor (bFGF), to promote the neural stem cell to proliferate.


The NSLCs obtained according to the invention can be differentiated into neuronal, astrocyte, and/or oligodendrocyte lineages in appropriate differentiation medium, for example NS-A differentiation medium (StemCell, Technologies) or NbActive medium (BrainBits™) including a retinoid compound, such as all-trans-retinoic acid or vitamin A, and BDNF, to induce the differentiation of NSLCs towards neuronal and/or glial cells. Neuronal cells include cells that display one or more neural-specific morphological, physiological, functional and/or immunological features associated with a neuronal cell type. Useful criteria features includes: morphological features (e.g., long processes or neurites), physiological and/or immunological features such as expression of a set of neuronal-specific markers or antigens, synthesis of neurotransmitter(s) such as dopamine or gamma aminobutyric acid (GABA), and functional features such as ion channels or action potentials characteristic of neurons.


In accordance with the method, reprogrammed cells can be selected based on differential adherence properties as compared to untransfected cells; for example, reprogrammed cells can form floating neurospheres or grow well on laminin while untransfected fibroblasts attach and grow well on regular cell culture treated plates. Reprogrammed cells include cells that exhibit one or more neural stem specific markers and morphology and the loss of some or all of the specific markers related to the original cells. Furthermore, some of the functionality of the neural-like cells (NLCs) can be assessed at different time points by, for example, patch-clamping, immunostaining for synaptophysin and MAP2b, and by immunochemical means such as by enzyme-linked immunosorbent assay (ELISA).


In certain embodiments, the present invention provides NSLCs that are able to initiate and direct central nervous system regeneration at a site of tissue damage and can be customized for individual patients using their own cells as the donor or starting cell. The present invention can be used to generate cells from an individual patient, thus making autologous transplantations possible as a treatment modality for many neurological conditions. Thus, this technology eliminates the problems associated with transplantations of non-host cells, such as, immunological rejection and the risk of transmitted disease. The great advantage of the present invention is that it provides an essentially limitless supply for autologous grafts suitable for transplantation. Therefore, it will obviate some significant problems associated with current source of materials and methods of transplantation.


Delivery of Polynucleotides


In certain embodiments, the invention concerns the use of polynucleotides, e.g. a polynucleotide encoding a MBD2 polypeptide, a MUSASHI1 polypeptide and/or a Ngn2 polypeptide. Means for introducing polynucleotides into a cell are well known in the art. Transfection methods of a cell such as nucleofection and/or lipofection, or other types of transfection methods may be used. For instance a polynucleotide encoding a desired polypeptide can be cloned into intermediate vectors for transfection in eukaryotic cells for replication and/or expression. Intermediate vectors for storage or manipulation of the nucleic acid or production of protein can be prokaryotic vectors, (e.g., plasmids), shuttle vectors, insect vectors, or viral vectors for example. A desired polypeptide can also be encoded by a fusion nucleic acid.


To obtain expression of a cloned nucleic acid, it is typically subcloned into an expression vector that contains a promoter to direct transcription. Suitable bacterial and eukaryotic promoters are well known in the art and described, e.g., in Sambrook and Russell (Molecular Cloning: a laboratory manual, Cold Spring Harbor Laboratory Press). The promoter used to direct expression of a nucleic acid of choice depends on the particular application. For example, a strong constitutive promoter is typically used for expression and purification. In contrast, when a dedifferentiation protein or compound is to be used in vivo, either a constitutive or an inducible promoter or compound is used, depending on the particular use of the protein. In addition, a weak promoter can be used, such as HSV TK or a promoter having similar activity. The promoter typically can also include elements that are responsive to transactivation, e.g., hypoxia response elements, Ga14 response elements, lac repressor response element, and small molecule control systems such as tet-regulated systems and the RU-486 system.


In addition to a promoter, an expression vector typically contains a transcription unit or expression cassette that contains additional elements required for the expression of the nucleic acid in host cells, either prokaryotic or eukaryotic. A typical expression cassette thus contains a promoter operably linked, e.g., to the nucleic acid sequence, and signals required, e.g., for efficient polyadenylation of the transcript, transcriptional termination, ribosome binding, and/or translation termination. Additional elements of the cassette may include, e.g., enhancers, and heterologous spliced intronic signals.


Expression vectors containing regulatory elements from eukaryotic viruses are often used in eukaryotic expression vectors, e.g., SV40 vectors, papilloma virus vectors, and vectors derived from Epstein-Barr virus. Other exemplary eukaryotic vectors include pMSG, pAV009/A+, pMTO10/A+, pMAMneo-5, baculovirus pDSVE, and any other vector allowing expression of proteins under the direction of the SV40 early promoter, SV40 late promoter, metallothionein promoter, murine mammary tumor virus promoter, Rous sarcoma virus promoter, polyhedrin promoter, or other promoters shown effective for expression in eukaryotic cells.


Standard transfection methods can be used to produce bacterial, mammalian, yeast, insect, or other cell lines that express large quantities of dedifferentiation proteins, which can be purified, if desired, using standard techniques. Transformation of eukaryotic and prokaryotic cells is performed according to standard techniques.


Any procedure for introducing foreign nucleotide sequences into host cells can be used. These include, but are not limited to, the use of calcium phosphate transfection, DEAE-dextran-mediated transfection, polybrene, protoplast fusion, electroporation, lipid-mediated delivery (e.g., liposomes), microinjection, particle bombardment, introduction of naked DNA, plasmid vectors, viral vectors (both episomal and integrative) and any of the other well known methods for introducing cloned genomic DNA, cDNA, synthetic DNA or other foreign genetic material into a host cell (see, e.g., Sambrook et al., supra). It is only necessary that the particular genetic engineering procedure used be capable of successfully introducing at least one gene into the host cell capable of expressing the protein of choice.


Conventional viral and non-viral based gene transfer methods can be used to introduce nucleic acids into mammalian cells or target tissues. Such methods can be used to administer nucleic acids encoding reprogramming polypeptides to cells in vitro. Preferably, nucleic acids are administered for in vivo or ex vivo gene therapy uses. Non-viral vector delivery systems include DNA plasmids, naked nucleic acid, and nucleic acid complexed with a delivery vehicle such as a liposome. Viral vector delivery systems include DNA and RNA viruses, which have either episomal or integrated genomes after delivery to the cell.


Methods of non-viral delivery of nucleic acids include lipofection, microinjection, ballistics, virosomes, liposomes, immunoliposomes, polycation or lipid-nucleic acid conjugates, naked DNA, artificial virions, and agent-enhanced uptake of DNA. Lipofection reagents are sold commercially (e.g., Transfectam™ and Lipofectin™). Cationic and neutral lipids suitable for efficient receptor-recognition lipofection of polynucleotides are known. Nucleic acid can be delivered to cells (ex vivo administration) or to target tissues (in vivo administration). The preparation of lipid:nucleic acid complexes, including targeted liposomes such as immunolipid complexes, is well known to those of skill in the art.


The use of RNA or DNA virus-based systems for the delivery of nucleic acids take advantage of highly evolved processes for targeting a virus to specific cells in the body and trafficking the viral payload to the nucleus. Viral vectors can be administered directly to patients (in vivo) or they can be used to treat cells in vitro, wherein the modified cells are administered to patients (ex vivo). Conventional viral based systems for the delivery include retroviral, lentiviral, poxviral, adenoviral, adeno-associated viral, vesicular stomatitis viral and herpesviral vectors, althoughntegration in the host genome is possible with certain viral vectors, including the retrovirus, lentivirus, and adeno-associated virus gene transfer methods, often resulting in long term expression of the inserted transgene. Additionally, high transduction efficiencies have been observed in many different cell types and target tissues.


pLASN and MFG-S are examples of retroviral vectors that have been used in clinical trials. In applications for which transient expression is preferred, adenoviral-based systems are useful. Adenoviral based vectors are capable of very high transduction efficiency in many cell types and are capable of infecting, and hence delivering nucleic acid to, both dividing and non-dividing cells. With such vectors, high titers and levels of expression have been obtained. Adenovirus vectors can be produced in large quantities in a relatively simple system.


Gene therapy vectors can be delivered in vivo by administration to an individual patient, typically by systemic administration (e.g., intravenous, intraperitoneal, intramuscular, subdermal, or intracranial infusion) or topical application. Alternatively, vectors can be delivered to cells ex vivo, such as cells explanted from an individual patient (e.g., lymphocytes, bone marrow aspirates, tissue biopsy) or universal donor hematopoietic stem cells, followed by reimplantation of the cells into a patient, usually after selection for cells which have been reprogrammed.


Ex vivo cell transfection for diagnostics, research, or for gene therapy (e.g., via re-infusion of the transfected cells into the host organism) is well known to those of skill in the art. In a preferred embodiment, cells are isolated from the subject organism, transfected with a nucleic acid (gene or cDNA), and re-infused back into the subject organism (e.g., patient). Various cell types suitable for ex vivo transfection are well known to those of skill in the art.


Vectors (e.g., retroviruses, adenoviruses, liposomes, etc.) containing therapeutic nucleic acids can be also administered directly to the organism for transfection of cells in vivo. Alternatively, naked DNA can be administered. Administration is by any of the routes normally used for introducing a molecule into ultimate contact with blood or tissue cells. Suitable methods of administering such nucleic acids are available and well known to those of skill in the art, and, although more than one route can be used to administer a particular composition, a particular route can often provide a more immediate and more effective reaction than another route.


Pharmaceutically acceptable carriers are determined in part by the particular composition being administered, as well as by the particular method used to administer the composition. Accordingly, there is a wide variety of suitable formulations of pharmaceutical compositions of the present invention.


Delivery of Polypeptides


In most, if not all the methods described herein, an alternative possibility consists of bypassing the use of a polynucleotide and contacting a cell of a first type cell directly with a compound (e.g. a polypeptide) for which an increased intracellular level is desired. In other embodiments, for example in certain in vitro situations, the cells are cultured in a medium containing one or more functional polypeptides.


An important factor in the administration of polypeptides is ensuring that the polypeptide has the ability to traverse the plasma membrane of a cell, or the membrane of an intra-cellular compartment such as the nucleus. Cellular membranes are composed of lipid-protein bilayers that are freely permeable to small, nonionic lipophilic compounds and are inherently impermeable to polar compounds, macromolecules, and therapeutic or diagnostic agents. However, proteins, lipids and other compounds, which have the ability to translocate polypeptides across a cell membrane, have been described. For example, “membrane translocation polypeptides” have amphiphilic or hydrophobic amino acid subsequences that have the ability to act as membrane-translocating carriers. Polypeptides for which an increased intracellular level is desired according to the invention can be linked to suitable peptide sequences for facilitating their uptake into cells. Other suitable chemical moieties that provide enhanced cellular uptake can also be linked, either covalently or non-covalently, to the polypeptides. Other suitable carriers having the ability to transport polypeptides across cell membranes may also be used.


A desired polypeptide can also be introduced into an animal cell, preferably a mammalian cell, via liposomes and liposome derivatives such as immunoliposomes. The term “liposome” refers to vesicles comprised of one or more concentrically ordered lipid bilayers, which encapsulate an aqueous phase. The aqueous phase typically contains the compound to be delivered to the cell.


In certain embodiments, it may be desirable to target a liposome using targeting moieties that are specific to a particular cell type, tissue, and the like. Targeting of liposomes using a variety of targeting moieties (e.g., ligands, receptors, and monoclonal antibodies) has been previously described.


Cells and Cell Lines


The invention encompasses the cells, cell lines, stem cells and purified cell preparations derived from any of the methods described herein. In some embodiments, the cells, cells lines, stem cells and purified cells preparations of the invention are of mammalian origins, including but not limited to human, primates, rodent, dog, cat, horse, cow, or sheep. In preferred embodiments, they originate from a human.


Accordingly, another aspect of the invention relates to modified cells, cell lines, pluripotent, multipotent or unipotent cells and purified cell preparations, wherein any of these cells comprise an exogenous polynucleotide encoding Musashi1 (Msi1); Msi1 and Ngn2; Msi1 and MBD2; and Ngn2 and MBD2; Msi1, Ngn2 and MBD2; Msi1, Ngn2, Nestin and MBD2; and other potential combinations from Table A preferably including Msi1 and Ngn2 and MBD2. In preferred embodiments the cell according to the invention is a stem-like cell, more preferably a Neural Stem-Like Cell (NSLC), the cell possessing one or more of the following characteristics:

    • expression of one or more neural stem cell marker selected from the group consisting of Sox2, Nestin, GFAP, Msi1, and Ngn2;
    • decreased expression of one or more genes specific to the cell that the NSLC was obtained from (e.g. see Table C);
    • forms neurospheres in the neurosphere colony formation assay;
    • capable of being cultured in suspension or as an adherent culture;
    • capable of proliferating without the presence of an exogenous reprogramming agent for over 1 month, preferably over 2 months, over 3 monthths, over 5 months and even for more than a year;
    • capable of dividing every 36 hours at low passage;
    • positive for telomerase activity;
    • capable of differentiation into a neuronal-like cell, astrocyte-like cell, oligodendrocyte-like cell and combinations thereof;
    • decreased expression of telomerase and one or more neural stem cell markers after differentiation;
    • having one or more morphological neurite-like processes (axons and/or dendrites) greater than one cell diameter in length after differentiation into a neuronal-like cell;
    • expression of at least one neural-specific antigen selected from the group consisting of neural-specific tubulin, microtubule associated protein 2, NCAM, and marker for a neurotransmitter after differentiation into a neuronal-like cell;
    • expression of one or more functional neural markers (e.g. synapsin) after differentiation into a neuronal-like cell;
    • capable of releasing one or more neurotrophic factors (e.g. BDNF) after differentiation into a neuronal-like cell;
    • negative in a tumor colony forming assay;
    • negative for tumor growth in SCID mice;
    • negative for teratoma growth in SCID mice;
    • capable of significantly improving one or more functional measures after placement of an adequate number of NSLCs into the void in a brain ablation model;
    • capable of significantly improving or maintaining one or more functional measures after injecting an adequate number of NSLCs into an EAE model; and
    • capable of improving one or more functional measures more significantly than hNPCs in CNS injury or neurodegenerative models.


Examples of all of the above items can be found in the Examples section of this application.


In preferred embodiments, a NSLC according to the inventions possesses all of the following characteristics:

    • ability to self-renew for significantly longer than a somatic cell;
    • is not a cancerous cell;
    • is stable and not artificially maintained by forced gene expression or by similar means and may be maintained in standard neural stem cell media;
    • can differentiate to a progenitor, precursor, somatic cell or to another more differentiated cell type of the same lineage;
    • has the characteristics of a stem cell and not just certain markers or gene expression or morphological appearance; and
    • does not exhibit uncontrolled growth, teratoma formation, and tumor formation in vivo.


In one particular embodiment, the reprogrammed cells (NSLCs) according to the invention are capable of proliferating for several months without losing their neural stem cell markers and their ability to differentiate towards neuron-like, astrocyte-like, and oligodendrocyte-like cells. The generation of the neural lineages is characterized based on morphology, phenotypic changes and functionality.


In some embodiments, the cells of the invention may have one or more of the following characteristics and properties: self-renewal, multilineage differentiation in vitro and in vivo, clonogenicity, a normal karyotype, extensive proliferation in vitro under well defined culture conditions, and the ability to be frozen and thawed, as well as any of the commonly known and/or desired properties or characteristics typical of stem cells. The cells of the invention may further express molecular markers of multipotent or pluripotent cells (i.e. gene and surface markers as defined previously).


Another aspect of the invention relates to the production of tissue specific autologous (self) stem and/or progenitor cells. These stem and/or progenitor cells may be used in cell therapy applications to treat diseases of cellular degeneration. Diseases of cellular degeneration include, for example, neurodegenerative diseases such as stroke, Alzheimer's disease, Parkinson's disease, multiple sclerosis, Amyotrophic lateral sclerosis, macular degeneration, osteolytic diseases such as osteoporosis, osteoarthritis, bone fractures, bone breaks, diabetes, liver injury, degenerative diseases, myocardial infarct, burns and cancer. It is envisioned that cells according to the invention may be implanted or transplanted into a host. An advantage of the invention is that large numbers of autologous stem cells can be produced for implantation without the risk of immune system mediated rejection. Those cells can lead to production of tissue suitable for transplant into the individual. Since the tissue is derived from the transplant recipient, it should not stimulate an immune response, as would tissue from an unrelated donor. Such transplants can constitute tissues (e.g. vein, artery, skin, muscle), solid organ transplants (e.g., heart, liver, kidney), neuronal cell transplants, or bone marrow transplants such as are used in the treatment of various malignancies such as, for example, leukemias and lymphomas. Neural stem cell, neuroprogenitor, or neuronal cell (as well as NSLCs and derivations thereof) transplants can also be used in the treatment of, for example, neurological disorders, stroke, spinal cord injury, Parkinson's disease, and the like, as well as potentially some non-neurological disorders such as a cardiac infarct.


Another aspect of the invention relates to a method to produce ex vivo engineered tissues for subsequent implantation or transplantation into a host, wherein the cellular components of those engineered tissues comprise cells according to the invention, or cells derived therefrom. For example, expanded cultures of the cells of the invention may be differentiated by in vitro treatment with growth factors and/or morphogens. Populations of differentiated cells are then implanted into the recipient host near the site of injury or damage, or cultured in vitro to generate engineered tissues, as described.


The methods and cells of the invention described herein can be used to immortalize cells, for example to generate a cell line. Using the methods disclosed herein, a somatic cell can be transformed into one possessing a dedifferentiated phenotype, thereby facilitating the generation of cell lines from a variety of tissues. Therefore, the invention encompasses such immortalized cells.


In addition, the methods of deriving the cells according to the invention, may be helpful in scientific and therapeutic applications including, but not limited to, (a) scientific discovery and research involving cellular development and genetic research (e.g. uses in lieu of human stem cells as a model cell line to study the differentiation, dedifferentiation, or reprogramming of human cells), (b) drug development and discovery (e.g., screening for efficacy and toxicity of certain drug candidates and chemicals, screening for prospective drugs or agents which mediate the differentiation, dedifferentiation, or reprogramming of cells), (c) gene therapy (e.g., as a delivery device for gene therapy), and (d) treatment of injuries, trauma, diseases and disorders including, but not limited to, Parkinson's, Alzehimer's, Huntington's, Tay-Sachs, Gauchers, spinal cord injury, stoke, burns and other skin damage, heart disease, diabetes, Lupus, osteoarthritis, liver diseases, hormone disorders, kidney disease, leukemia, lymphoma, multiple sclerosis, rheumatoid arthritis, Duchenne's Musclar Dystrophy, Ontogenesis Imperfecto, birth defects, infertility, pregnancy loss, and other cancers, degenerative and other diseases and disorders.


Additional aspects concern therapeutic methods, methods of treatment and methods of regenerating a tissue or organ in a mammal (e.g. a human subject). One particular method concerns a method of regenerating a mammalian tissue or organ which comprises contacting the tissue or organ to be regenerated with a SLC, NSLC, or other desired cell or artificial tissue construct as defined herein. The SLC, NSLC, desired cell or artificial tissue construct may be placed in proximity to the tissue or organ to be regenerated by administering to the subject using any suitable route (e.g. injecting the cell intrathecally, directly into the tissue or organ, or into the blood stream).


Another method for repairing or regenerating a tissue or organ in a subject in need thereof comprises administering to the subject a compound inducing a direct or indirect endogenous expression of at least one gene regulator in cells of the tissue or organ and/or a compound inducing a direct or indirect endogenous expression of at least one gene regulator in cells capable of transformation or dedifferentiation in vivo in the subject. Accordingly, the expression of the at least one gene regulator reprograms the cells into desired cells of a different type (e.g. neural stem-like cells), and these cells of a different type are effective in repairing or regenerating said tissue or organ.


Another method comprises obtaining cells or tissue from a patient (e.g. hematopoietic stem cells, fibroblasts, or keratinocytes), reprogramming a plurality of such cells or the tissue, and reintroducing the reprogrammed cells or tissue into the patient. A related aspect concerns pharmaceutical compositions comprising a plurality of a desired cell, SLC and/or Neural Stem-Like Cell (NSLC) or reprogrammed tissue as defined herein.


The therapeutic methods of the invention may be applicable to the regeneration or repair of various tissues and organs including, but not limited to, the brain, the spine cord, the heart, the eye, the retina, the cochlea, the skin, muscles, intestines, pancreas (including beta cells), kidney, liver, lungs, bone, bone marrow, cartilage, cartilage discs, hair follicles, teeth, blood vessels, glands (including endocrine and exocrine glands), ovaries, reproductive organs, mammary and breast tissue.


A related aspect concerns pharmaceutical compositions comprising a plurality of desired cell, SLC and/or Neural Stem-Like Cell (NSLC) as defined herein.


Tissues


Another aspect of the invention relates to a tissue containing reprogrammed cells as defined herein that can be implanted into a subject in need thereof.


In some embodiments the present invention provides for the reprogramming of cells within a tissue, for example an in vitro produced 3D tissue construct comprising cells and extracellular matrix produced by these cells. In addition, transfected cells can be seeded on top of these 3D tissue constructs that can be made completely autologously, thus preventing host rejection, making it completely immunocompatible and as carrier for reprogrammed cells to be transplanted in vivo. Advantageously, these newly created cells can be used in their undifferentiated and/or differentiated state within these tissues for in vitro diagnostic purposes or transplanted into a patient in need of such a construct in cell therapy/tissue replacement approaches.


The invention further encompasses 3D Neuronal-Like multilayer tissue. Cells within CDM reprogrammed to Neural Stem-Like Cells according to the invention readily differentiate into neuronal-like cells, astrocyte-like cells, and oligodendrocyte-like cells within the CDM. It is thus possible to use CDM and reprogramming methods of the invention to reprogram the cells within the CDM to form 3D Neuronal-Like multilayer tissue (up to >30 cell layers). Such 3D tissue comprises neurons (or specifically, neuron-like cells), astrocytes (or specifically, astrocyte-like cells), and oligodendrocytes (or specifically, oligodendrocyte-like cells) and it can be made completely autologously, can be manually handled and implanted with relative ease, or can used as an in vitro CNS tissue model.


One particular aspect concerns an artificial tissue construct which comprises a 3D assembly of in vitro cultured cells and extracellular matrix produced by these cells. The cells may be desired cells, SLC and/or a plurality of Neural Stem-Like Cell (NSLC) obtained using any one of the methods described herein.


Screening Methods


Another aspect of the invention relates to methods for identifying new compounds (e.g. small molecules, drugs, etc) capable of transforming a cell of a first type to a desired cell of a different type. These new compounds may be useful for research purposes or as medicaments for use in repairing or regenerating tissues in a subject.


The Examples section provides principles, methods and techniques useful for screening and identifying such desirable active compounds. For instance, those skilled in the art will understand that it is conceivable to screen for compounds that will induce transformation of a cell of a first type to a NSLC by replacing the “induction” or “biological activity” provided by the transient increase of Musashi1, NGN2 or MBD2 in the cell by a candidate compound to be tested (e.g. a library of small molecules or compounds) and measuring activity or efficacy of the candidate compound in generating the NSLC. Individual or mixture of active compounds would be selected if they have the same activity and/or if they can provide the same or similar effects as these polypeptides (e.g. cell transformation and/or appearance of any desirable markers or desirable characteristics as defined hereinbefore). For example, a compound or mixture of compounds capable of transforming a fibroblast into a NSLC could be identified by:

    • (i) Setting up, culturing and transforming the fibroblasts into NSLC as in Example 1;
    • (ii) Screening a library of compounds by replacing Msi1, Ngn2 and/or MBD2 with each candidate compound in a different well;
    • (iii) Identify a compound ‘hit’ when the candidate compound is able to transform the fibroblasts into NSLCs approximately as well as the replaced Msi1, Ngn2 and/or MBD2;
    • (iv) If compound from part (iii) did not replace all of Msi1, Ngn2 and MBD2, and is not able to transform the fibroblasts into NSLCs by itself, then by including the compound from (iii) in each well, screening a library of compounds by replacing the Msi1, Ngn2 and/or MBD2 that was not removed in part (ii) with each candidate compound in a different well;
    • (v) Identify a compound ‘hit’ when the candidate compound is able to transform, along with the compound from part (iii), the fibroblasts into NSLCs approximately as well as the replaced Msi1, Ngn2 and/or MBD2;
    • (vi) If compound from part (V) did not replace all of Msi1, Ngn2 and/or MBD2, and is not able to transform the fibroblasts into NSLCs together with the compound from part (iii), then by including the compound from (iii) and (v) in each well, screening a library of compounds by replacing the Msi1, Ngn2 or MBD2 that was not removed in part (ii) and (iv) with each candidate compound in a different well;
    • (vii) Identify a compound ‘hit’ when the candidate compound is able to transform, along with the compound from part (iii) and (v), the fibroblasts into NSLCs approximately as well as the replaced Msi1, Ngn2 or MBD2;
    • (viii) A combination of the compounds from part (iii), (v) and (vii) will be able to transform the fibroblasts into NSLC; modifications to these compounds can be made and further screened to identify more effective or safe versions of these compounds.


The same principles are applicable for other desired types of stem-like cells including pluripotent-like cells, mesendoderm-like cells, pancreatic progenitor-like cells, etc. Tables A and B, and the Examples section provides, for each of these types of cells, a list of potential genes and/or compounds to be considered in such screening methods.


Accordingly, the present invention encompasses these and any equivalent screening methods where candidate compounds are tested for their efficacy in transforming a cell of a first type to a desired cell of a different type when compared to the efficacy of the reprogramming factor and/or gene regulator as defined herein.


Delivery of Neurotrophic Factors


Local delivery of neurotrophic factors has been suggested as a method to treat several neurological conditions. Strategies using neurotrophic molecules focus on preventing the progressive loss of neurons, maintaining neuronal connections and function (neuroprotection), and inducing additional regenerative responses in neurons such as increased neurotransmitter turnover and/or axonal sprouting (neuroregeneration). Up to date, several therapeutic strategies to deliver neurotrophic-factors in animal models have been explored, but so far testing of the effects of growth factors on the brain and nervous system have been limited to direct peripheral injection of large doses of these factors, which carries a significant risk of side effects. Accordingly, a related aspect of the invention relates to overcoming these problems by using NSLC cells and cell lines according to the invention which can stably express and secrete growth factors of potential interest after transplantation.


To summarize, the present invention provides a plentiful source of Neural Stem-Like Cells, Neuron-Like Cells, Astrocyte-Like Cells or Oligodendrocyte-Like Cells for potential clinical treatments which require transplantation of neural stem cells, neurons, astrocytes or oligodendrocytes 1) to compensate for a loss of host cells (ex. neurons) or 2) as vehicles to deliver genetically-based drugs. Further, the invention provides a novel neurological tool for use in basic research and drug screening.


Those skilled in the art will recognize, or be able to ascertain using no more than routine experimentation, numerous equivalents to the specific procedures, embodiments, claims, and examples described herein. Such equivalents are considered to be within the scope of this invention and covered by the claims appended hereto. The invention is further illustrated by the following examples, which should not be construed as further limiting.


EXAMPLES

The examples set forth herein below provide exemplary methods for obtaining Reprogrammed and Dedifferentiated cells, including Neural Stem-Like Cells (NSLCs). Also provided are exemplary protocols, molecular tools, probes, primers and techniques.


Example I

Preparation of Human Fibroblast Cells


Human Foreskin fibroblast (HFF) cells were purchased from American Type Culture Collection (ATCC, Manassas, Va.) and expanded in cell culture flasks with Dulbecco's Modified Eagle's Medium (DMEM, Invitrogen), supplemented with 10% heat-inactivated fetal calf serum (FCS, Hyclone Laboratories), 0.1 mM non-essential amino acids, and 1.0 mM sodium pyruvate (Invitrogen) at 37° C., 5% CO2. The medium was changed twice per week. Cells were trypsinized using Trypsin 0.25% for 4 minutes at 37° C., followed by adding trypsin inhibitor solution, pelleting the cells by centrifugation, washing the cells once with PBS, and plating the cells at a ratio of 1:2 onto tissue culture flasks until a suitable number of cells was reached.


Cells were then trypsinized and plated (8×104 cells/well) in cell culture plates pre-coated with Laminin (10 μg/ml, Sigma) in two different composition of CDM medium: CDM I Medium consisting of a 3:1 ratio of Dulbecco's modified Eagle medium (DMEM, high glucose (4.5 g/L) with L-glutamine and sodium pyruvate) and Ham's F-12 medium supplemented with the following components: EGF (4.2×10−10M), bFGF (2.8×10−10M), ITS (8.6×10−5M), dexamethasone (1.0×10−7M), L-3,3′,5-triiodothyronine (2.0×10−10M), ethanolamine (10−4M), GlutaMAX™ (4×10−3M), and glutathione (3.3×10−6M), but without the presence of L-ascorbic acid.


CDM II Medium consisting of a 3:1 ratio of Dulbecco's modified Eagle medium (DMEM, high glucose (4.5 g/L) with L-glutamine and sodium pyruvate) and Ham's F-12 medium supplemented with the following components: EGF (2.5 ng/ml), bFGF (10 ng/ml), ethanolamine (2.03 mg/ml), insulin (10 mg/ml), Selenious acid (2.5 μg/ml), dexamethasone (19.7 μg/ml), L-3,3′,5-triiodothyronine (675 ng/ml), GlutaMAX™ (4×10−3M), and glutathione (3.3×10−6M).


Transient Transfection of HFF by Lipofectamine Using Constructed Vectors


After two days in culture, cells were transfected with pCMV6-XL5-MBD2 (2 g) (a DNA demethylator) using lipofectamine reagent (Invitrogen) as per the manufacturer's protocol. The DNA-lipid complex was added to cells and incubated for 24 h at 372C, 5% CO2. After 24 hours of transfection with the DNA demethylator, the medium was changed and cells were transfected by pCMV6-XL5-Musashi1 (2 g, Origene) or pCMV6-XL4-Ngn2 (2 g, Origene) for 24 h. After 24 hours, the medium was changed to Neural Progenitor Basal Medium (NPBM, Lonza) supplemented with Noggin (20 ng/ml, Peprotech), EGF (20 ng/ml, Peprotech), and bFGF (20 ng/ml, Peprotech) and cultured in this Proliferation Medium. Cells were retransfected after three days and incubated at 372C, 5% CO2 and 5% O2. After 7 days in proliferation conditions, 50% of the Proliferation Medium was changed to Differentiation Medium (NbActive, BrainBits™) supplemented with Forskolin (10 μM, Tocris), all-trans-Retinoic Acid (ATRA, 5 μM, Spectrum), bFGF (20 ng/ml, Peprotech), NGF (20 ng/ml, Peprotech), and BDNF (20 ng/ml, Peprotech); medium was changed every day by increasing the percentage of Differentiation Medium over Proliferation Medium, and the cells were cultured for 20 days.


Visual observation of reprogrammed cells was performed by light microscopic observation every day following transfection using bright field at 10× magnification. Samples were collected at different time points (6, 12, and 20 days) to analyze neuronal gene expression and protein levels by gene array and immunohistochemistry. Following transfection, reprogramming cells displayed a rapid change in cellular morphology within 3 days post-transfection (FIG. 1). The cells were more rounded and the cell's cytoplasm retracted towards the nucleus forming contracted cell bodies with extended cytoplasmic extensions and exhibiting neuronal perikaryal appearance at day 6 and 12, which was maintained until day 20. However, this morphology was not observed in untransfected cells at day 6 and 12.


Gene Array Analysis


Characterization of the newly engineering cells after transfection was performed using a neuronal gene-array containing 48 partial cDNAs coding for these genes and controls.


RNA was isolated from samples using QIAshredder™ (Qiagen) and RNeasy™ Plus mini Kit (Qiagen) as per manufacturer's instructions. DNase I treatment was performed on the RNeasy™ Column to further remove the transfected plasmid DNA using Rnase-Free DNase Set (Qiagen). RNA was eluted in 35 μl of RNase-free water. Before cDNA synthesis, all RNA samples were quantified using the NanoDrop 1000™ (ThermoScientific). cDNA was prepared using the High Capacity cDNA archive kit (Applied Biosystems) as per the manufacturer's instructions. 400 ng of RNA was used in each 50 μl RT reaction. The resulting cDNA samples were used immediately for TLDA analysis. For each card of the Taqman™ low-density array (TLDA), there are eight separate loading ports that feed into 48 separate wells for a total of 384 wells per card. Each 2 μl well contains specific, user-defined primers and probes, capable of detecting a single gene. In this study, a customized Neuronal Markers 2 TLDA was configured into eight identical 48-gene sets, i.e. 1 loading port for each 48-gene set. Genes were chosen based on literature. Each set of 48 genes also contains three housekeeping genes: ACTIN, GAPDH, and PPIA.


A sample-specific master mix was made for each sample by mixing cDNA (160 ng for each loading port), 2× Taqman™ Gene Expression Master Mix (Applied Biosystems) and nuclease-free water (USB) for a total of 100 μl per loading port. After gentle mixing and centrifugation, the mixture was then transferred into a loading port on a TLDA card. The array was centrifuged twice for 1 minute each at 1200 rpm to distribute the samples from the loading port into each well. The card was then sealed and PCR amplification was performed using Applied Biosystems 7900HT™ Fast Real-time PCR system. Thermal cycler conditions were as follows: 2 minutes at 50° C., 10 minutes at 94.5° C., and 30 seconds at 97° C., 1 minute at 59.7° C. for 40 cycles. 1 TLDA's was prepared for 8 samples.


Relative Expression values were calculated using the Comparative CT method. Briefly, this technique uses the formula 2−ΔΔCT to calculate the expression of target genes normalized to a calibrator. The threshold cycle (CT) indicates the cycle number at which the amount of amplified target reaches a fixed threshold. CT values range from 0 to 40 (the latter representing the default upper limit PCR cycle number that defines failure to detect a signal). ΔCT values [ΔCT=CT (target gene)−CT (Average of 3 Housekeeping genes)] were calculated for HFF Ctrl, and subsequently used as the calibrator for the respective samples. All gene expression values were assigned a relative value of 1.00 for the calibrator, which is used to determine comparative gene expression such that ΔΔCT=ΔCT (Treated)−ΔCT (HFF Ctrl). Relative Expression is calculated using the formula 2−ΔΔCT.


Quantitative comparison of astrocyte, neuron, and oligodendrocyte gene expression allowed identification of the majority of the genes that are differentially expressed in reprogrammed cells. Data in Table 1 were analyzed by using a significance analysis algorithm to identify genes that were reproducibly found to be enriched in reprogrammed cells compared to untransfected cells. After the transfection with Msi1 or Ngn2 in the presence of MBD2, the expression of oligodendrocytes progenitors such as NKx2.2, olig2, and MAG and two markers for astrocytes (GFAP and AQP4) were highly increased. Also, several markers of early neuronal cells were enhanced after the transfection of HFF. TDLA data revealed a remarkable increase in specific markers for interneurons, such as somatostatin and calbindin1. The induction of Doublecortin (DCX), which is expressed by migrating immature cells during development, and acetylcholine (ACHE) mRNA, an early marker of neuronal cells, were highly expressed in the reprogrammed cells (Table 1). Transfection increased the expression of dihydropyrimidinase-like 3 (DPYSL3), an early marker of newborn neurons, to fivefold with Msi1 and seven fold with Ngn2. Expression of Microtubule-Associated Protein 2 (MAP2), an essential marker for development and maintenance of early neuronal morphology and neuronal cell adhesion molecule, were highly expressed with Msi1 and Ngn2 (Table 1). The expression of enolase-2, a marker of mature neurons, was 20-fold enhanced by Msi1 and Ngn2. Member of the NeuroD family NeuroD1 was highly expressed after transfection with Msi1 to 84 fold and to 34 by Ngn2.


Gene expression of growth factors such as IGF-1, IGF2, NPY and CSF-3 was also enhanced in reprogrammed cells. The expression of VEGF and GDNF genes were up-regulated to almost five fold and seven fold by Msi1 and Ngn2, respectively. However, the expression of BDNF, EGF, and bFGF were not activated and even down-regulated as compared to untransfected cells. The expression of growth associated protein (GAP-43), a growth- and regeneration-associated marker of neurons, and expression of netrin, implicated in neuronal development and guidance, were highly enriched in reprogrammed cells. Expression of receptors for growth and neurotrophin factors was increased, such as type III receptor tyrosine kinase, neurotrophic tyrosine kinase, and neurotrophic tyrosine kinase receptor. Vimentin and fibronectin, markers for fibroblasts, were down-regulated in reprogrammed cells compared to the untransfected control fibroblast cultures.









TABLE 1







Gene array of transfected human fibroblast cells by Msi1/MBD2 and Ngn2/MBD2.


Gene array was performed on samples after two weeks of differentiation. Expression values are


given relative to untransfected fibroblasts.














Relative
Relative




Company
expression
expression


Symbol
Common name and description
Gene ID
to Msi1
to Ngn2





Astrocytes and






oligodendrocytes






markers






NKx2-2
Markers for oligodendrocyte
NM_002509.2
very high
very high


OLIG2
progenitors
NM_005806.2
47.511
8.38


MAG
Oligodendrocyte lineage transcription
NM-080600.1
212.61
4.51


GFAP
factor 2
NM_002055.4
very high
very high


AQP4
Myelin-associated glycoprotein
NM_001650.4
83.77
56.86



Glial fibrillary acidic protein






Aquaporin 4





NC markers






SST
Somatostatin, specific marker for
NM_001048.3
32.73
35.34


CALB1
interneurons
NM_004929.2
18.21
13.22


Tubulin1A
Calbindin 1, interneuron marker
NM_006009.2
7.45
9.32


NES
Are necessary for axonal growth
NM_006617.1
1.61
1.54


DCX
Precursor neurons (nestin)
NM_178151.1
very high
very high


ACHE
An early neuronal marker
NM_015831.2
9.02
13.22


ENO2
(Doublecortin)
NM_001975.2
22.62
20.68


NEUROD1
Acetylcholinesterase, marker of early
NM_002500.2
84.22
34.27


DPYSL3
neuronal development
NM_001387.2
5.33
7.02


MAP2
A marker for neurons cells, enolase
NM_002374.3
86.38
89.67


NCAM
Neural marker; expression gradually
NM_18135.3
very high
very high


CEND1
increased from neural precursor to
NM_016564.3
4.80
5.57



fully differentiated neuron






Dihydropyrimidinase-1ike3, marker






of immature neurons






Microtubule-associated protein 2,






essential for development of early






neuronal morphology and






maintenance of adult neuronal






morphology






Neural cell adhesion molecule 1






Cell cycle exit & neuronal






differentiation, early marker of






proliferating precursor cells that will






differentiate to neurons





Neuroregeneration






and survival genes






FGF2
Fibroblast growth factor
NM_002006.4
0.06
0.11


EGF
Epidermal growth factor,
Hs00153181_m1
0.99
0.56


IGF-1
Insulin growth factor-1,
NM_000618.2
58.92
21.21


IGF-2
Insulin growth factor-2
NM_0000612.3
very high
very high


CSF3
Granulocyte colony-stimulating
NM_2219.1
very high
42.60


BDNF
factor
NM-199231.1
0.05
0.03


GDNF
Brain derived growth factor,
NM-000614.2
4.77
6.89


CNTF
neurogenesis
NM_001025366.1
1.86
1.09


VEGF
Glial derived neurotrophic factor
NM_130850.1
6.67
7.32


BMP-4
Ciliary neurotrophic factor
NM_002253.1
5.96
8.57


KDR
Vascular endothelial growth factor
NM_006180.3
31.78
6.83


NTRK2
Bone morphogenetic protein 4
NM_00905.2
10.31
13.37


NPY
Type III receptor tyrosine kinase)
NM_002649.2
very high
very high


PIK3CG
Neurotrophic tyrosine kinase receptor
NM_213662.1
very high
very high


STAT3
(TrkB)
NM_002045.2
2.14
3.65


Gap43
Neuropeptide factors
_NM_006180.3
very high
very high


NTN1
phosphoinositide-3-kinase,
NM_024003.1
26.84
23.98


NTRk2
Signal transduction transcription 3
NM_003061.1
10.31
13.37


Slit
Growth associated protein 43
Hs00185584
very high
very high


Vimentin
Netrinl, implicated in neuronal
NM_212474.1
0.11
0.13


Fibronectin
development and guidance

0.15
0.23



Neurotrophic tyrosine kinase,






receptor, type 2






Axonal guidance molecules






Radial glia and fibroblast marker






fibronectin is a marker for fibroblasts










Immunohistochemical Analysis


Cells were fixed with a 4% formaldehyde/PBS solution for 10 min at room temperature and subsequently permeabilized for 5 min with 0.1% Triton X-100™ in 4% formaldehyde/PBS. After two brief washes with PBS, unspecific antibody binding was blocked by a 30 min incubation with 5% normal goat serum in PBS. Then primary antibodies were added in 5% normal goat serum/PBS as follows: Mouse anti-Nestin (1:100, BD) as an intermediate microfilament present in neural stem cells and mouse anti-NCAM (1:100, Neuromics) as neuronal adhesion molecule. After a 2 h incubation the cells were washed 4 times for 5 min each with 0.1% Tween™/PBS. Appropriate fluorescence-tagged secondary antibody was used for visualization; Goat anti-mouse 546 (1:200, invitrogen) prepared in 5% normal goat serum/PBS was used. After incubation for one hour, cells were washed in 0.1% Tween™/PBS three times for 5 min each. The DNA stain Hoechst33342 (Invitrogen) was used as a marker of nuclei (dilution 1:5000 in PBS, 10 min incubation). Fluorescence images were taken with a Cellomics™ ArrayScan HCS Reader microscopy system. To determine an estimate of the percentage of cells adopting neuronal or glial phenotypes, random fields were selected and for each field the total number of cells (as determined by counting Hoechst stained nuclei) and the total number of cells positive for neuronal or glial markers were determined.


To confirm that these cells exhibited markers of neuronal lineages, cells were immunostained for nestin and NCAM. This analysis revealed that reprogrammed cells expressed both proteins. As shown in FIG. 2, NCAM was present in cells during the 6 days post-transfection and increase at day 12 and 20 following differentiation, while the inverse pattern was observed for the nestin staining.


This study showed the ability to reprogram HFF cells using one neurogenic transcription factor with the presence of a DNA demethylator towards cells that expressed neuronal genes and proteins specific to neural stem cells and neuronal cells. These reprogrammed cells were stable in culture for at least 2 weeks.


Example II

Comparison of Reprogramming Efficiency of Three Different Neurogenic Genes


HFF cells were cultured as described in Example I and plated in CDM I medium. Cells were transfected using the Amaxa Nucleofector™™ Device (Lonza). The HFFs were harvested with TrypLE™ (Gibco), resuspended in CDM Medium and centrifuged for 10 min at 90×g (1×106 cells/tube). The supernatant was discarded and gently resuspended in 100 μl of Basic Nucleofector™ Solution (basic Nucleofector™ kit for primary mammalian fibroblasts, Lonza). Each 100 μl of cell suspension was combined with a different mix of plasmid DNA (for example, sample 1 was mixed with 2 μg of pCMV6-XL5-Pax6 and 2 μg pCMV6-XL5-MBD2). Cell suspension was transferred into an Amaxa certified cuvette and transfected with the appropriate program (U023). The sample was transferred without any further resuspension into a coated culture plate with LAS-Lysine/Alanine (BrainBits™, 50 μg/ml) and the cells were incubated at 37° C., 5% CO2. These steps were repeated for each sample that was transfected. After 24 hours, the medium was changed to Proliferation Medium. After two days, cells were retransfected using lipofectamine as described in Example I and incubated at 37° C., 5% CO2 and 5% O2. After 6 days, differentiation was induced with Differentiation Medium that gradually replaced the Proliferation Medium over several days. Cells were collected at day 14 for RT-PCR and imunohistochemistry analysis.


Gene Expression Analysis


RNA isolation and quantification was performed as previously described in Example I. cDNA was prepared using the High Capacity cDNA RT kit (Applied Biosystems) as per the manufacturer's instructions with a final cDNA concentration of 2 ng/l. Real-time PCR was then performed for each gene of interest using the FAST PCR master mix (Applied Biosystems) and the Taqman™® Gene Expression Assays (Applied Biosystems) listed below:
















Gene Name
Assay ID









ACHE
Hs00241307_m1



NES
Hs00707120_s1



TUBB3
Hs00964962_g1



GFAP
Hs00157674_m1



PAX6
Hs00240871_m1



MSI1
Hs01045894_m1



NGN2
Hs00702774_s1



MAP2
Hs00258900_m1



GAPDH (housekeeping gene)
Hs99999905_m1



PPIA (housekeeping gene)
Hs99999904_m1










The FAST 96-well reaction was performed with 8 ng cDNA per well in a 101 reaction with 40 cycles. Thermal cycler conditions were as follows: 20 seconds at 95° C., and 1 second at 95° C., 20 seconds at 60° C. for 40 cycles.


Relative Expression values were calculated as previously described in Example I, except the Average of 2 Housekeeping genes (GAPDH & PPIA) was used for normalization instead of the Average of 3 Housekeeping genes. Identification of neuronal lineage genes was investigated following the transfection with three independent vectors containing Msi1, Ngn2, and Pax6.


As shown in Table 2, after 14 days following transfection, relative expression of mRNA of neuronal lineage was undetectable in untransfected cells (HFF), while the cells transfected with Msi1 or Ngn2 in the presence of MBD2 expressed neural stem cell markers (Nestin and Sox2), however the expression of Sox2 was much more highly expressed than nestin following transfection with Ngn2 or Msi1. Neuronal and astrocyte specific genes (βIII-Tubulin, MAP2b, GFAP, and ACHE) was increased as well. mRNAs level of the tripotent-associated genes βIII-tubulin, MAP2b, acetycholine, and GFAP were undetectable in Pax6 transfected cells, indicating that Pax6 alone was not implicated in the reprogramming process toward neuronal lineage.









TABLE 2





Relative expression of gene expression of different neuronal lineage performed by RT-PCR following the


transfection of HFF by Msi1, Ngn2, or Pax6 in the presence of MBD2 and cultured for 14 days.





















MSI1
NGN2
PAX6
NES
TUBB3


















Rel.
Std.
Rel.
Std.
Rel.
Std.
Rel.
Std.
Rel.
Std.



Exp.
Dev.
Exp.
Dev.
Exp.
Dev.
Exp.
Dev.
Exp.
Dev.





#1 Control
1.00
0.07
1.08
0.57
1.11
0.67
1.00
0.02
1.00
0.01


Untransfect.












#2
4077.82
248.02
1.18
0.66
487.09
69.58
8.62
0.00
6.58
0.11


MSI1/MBD2












#3
14.16
0.63
47803.26
192.78
624.31
91.27
8.62
0.02
8.33
0.02


NGN2/MBD2












#4
1.70
0.36
0.27
0.01
29564.43
357.89
0.46
0.00
0.49
0.02


PAX6/MBD2



























ACHE
GFAP
MAP2
SOX2


















Rel.
Std.
Rel.
Std.
Rel.
Std.
Rel.
Std.




Exp.
Dev.
Exp.
Dev.
Exp.
Dev.
Exp.
Dev.






#1 Control
1.02
0.29
1.00
0.06
1.00
0.01
1.00
0.09



Untransfect.











#2
6.58
0.64
215.71
20.65
5.50
0.46
3499.53
184.85



MSI1/MBD2











#3
8.33
0.97
365.60
5.11
5.42
0.00
4039.03
8.65



NGN2/MBD2











#4
1.98
0.48
1.15
0.13
0.55
0.04
1.00
0.03



PAX6/MBD2


















Immunohistochemical Analysis


Fluorescent imunohistochemical staining was performed as previously described in Example I. In agreement with the RT-PCR data, immunohistochemical analysis of these cultures revealed that reprogrammed cells (with Msi1 or Ngn2) generated morphologically complex neurons that were positive for MAP2b, indicating the differentiation of NSLCs to neuron-like cells (NLCs) (FIG. 3). However, the positive staining for these markers was undetectable after transfection with Pax6/MBD2. Moreover, the newly formed neurons expressed the markers for and developed long neurites with growth cones at their ends, expressed neural specific genes, and ceased to proliferate when they were exposed to differentiation conditions.


Example III

Transfection of HFF by Various Combinations of Vectors and Disruption of Cell Cytoskeleton


Various combinations of neurogenic regulators and cytokines for epigenetic modifications were tested to ascertain their effect on reprogramming efficiency. Starting one day before transfection, cells were treated with or without cytochalasin B (Calbiochem), with the concentration decreased every day over five days during media changes (starting with 10 μg/ml Cytochalasin B on day 1 to 7.51 μg/ml, 51 μg/ml, 2.51 μg/ml, and 0 μg/ml over the subsequent four days) in order to investigate the effect of disrupting the cell cytoskeleton on the process of reprogramming. Cells were transiently transfected as described in Example II with one or two vectors containing one neurogenic transcription factors by nucleofection. Cells were co-transfected with either of two DNA demethylators, MBD2 or GAdd45B, (e.g. 2×106 cells were transfected with pCMV6-XL5Msi1 (2 μg) and pCMV6-XL5-MBD2 (2 μg)). After 24 hours, the medium was changed to Neural Proliferation Medium (NeuroCult™ proliferation Kit, StemCell Technologies) consisting of DMEM/F12 (1:1), glucose (0.6%), sodium bicarbonate (0.1%), glutamine (20 mM), HEPES (5 mM), insulin (230 μg/ml), transferrin (100 μg/ml), progesterone (200 nM), putrescine (90 μg/ml), and sodium selenite (300 nM) and supplemented with Noggin (20 ng/ml, Peprotech), recombinant hFGF (20 ng/ml, Peprotech), and recombinant hEGF (20 ng/ml, Peprotech) and cells were cultured for two weeks at 37° C., 5% CO2 and 5% O2. Cells were then analyzed for neural stem cell markers.


Gene Expression Analysis


Gene expression analysis was performed for neural stem-specific markers (Sox2, Nestin, GFAP) and a fibroblast-specific marker (Col5A2) by RT-PCR as previously described in Example I. RT-PCR analysis showed that the relative expression of Sox2, nestin and GFAP was enhanced after transfecting the cells with the neurogenic transcription factors. As shown in Table 3, transfecting the cells with one transcription factor Msi1 in the presence of Gadd45b was associated with up-regulation of relative expression of Sox2 (22.3±5.26) and GFAP (10.14±0.15) and the expression of the these genes was highly increase when transfecting the cells with Ngn2 by 20 fold and 10 fold respectively. Combining the two neurogenic factors (Msi1 and Ngn2) with Gadd45b enhanced further the expression of Sox2 and GFAP. Transfecting the cells with one transcription factor (Msi1 or Ngn2) in the presence of MBD2 was associated with up-regulation of relative expression of Sox2, Nestin, and GFAP and down-regulation of Col5A2, while co-transfection with Gadd45b did not increased the expression of nestin and the expression of Col5A2 was not regulated. The enhancement of neural stem cells relative expression was observed when transfecting the cells with two neurogenic genes in combination with MBD2; a small increase in the expression was noticed in the presence of cytochalasin B under certain conditions. An increase in the relative expression of the neural stem-specific markers (Sox2, Nestin, GFAP) and a decrease in the fibroblast-specific gene (COL5A2) was observed after transfection with Msi1/Ngn2/MBD2, Msi1/Ngn2/Gadd45b, Msi1/MBD2 or Ngn2/MBD2 (Table 3). This study demonstrated that MBD2 increased more reprogramming efficiency then GDA45b and showed that cytochalasin B had no effect of its own in the control cultures.









TABLE 3







RT-PCR analysis of relative expression of neuronal precursor cell markers such as nestin, Sox2, and GFAP after transfection of


fibroblast cells with different combinations with or without the co-treatment with cytochalasin B. Relative expression of Sox2,


nestin, and GFAP in NSLCs was increased after transfection with both transcription factors (Ngn2 and Msi1) with MBD2 as the


DNA demethyaltor. As demonstrated, this upregulation of neural stem cell gene expression was associated with a


decrease of CoL5A2, a specific gene for fibroblast cells.
















COL5A2
FBN2
NES
MAP2
TUBB3
SOX2
ACHE
GFAP
























Rel.
Std.
Rel.
Std.
Rel.
Std.
Rel.
Std.
Rel.
Std.
Rel.
Std.
Rel.
Std.
Rel.
Std.



Exp.
Dev.
Exp.
Dev.
Exp.
Dev.
Exp.
Dev.
Exp.
Dev.
Exp.
Dev.
Exp.
Dev.
Exp.
Dev.


























 #1, +CytoB,
1.00
0.07
1.00
0.01
1.00
0.04
1.00
0.05
1.00
0.05
1.00
0.05
1.00
0.10
1.00
0.11


Control


















 #2, −CytoB,
1.00
0.03
1.00
0.08
1.00
0.00
1.00
0.09
1.00
0.09
1.15
0.80
1.01
0.18
1.00
0.01


Control


















 #3, +CytoB,
0.85
0.04
0.75
0.02
0.60
0.01
0.29
0.01
0.44
0.00
22.39
5.26
0.81
0.19
10.14
0.15


Msi1, GAD45b


















 #4, −CytoB,
0.87
0.03
1.81
0.09
1.84
0.04
2.31
0.00
2.09
0.03
20.28
5.33
1.99
0.74
6.03
0.05


Msi1, GAD45b


















 #5, +CytoB,
0.84
0.04
0.77
0.03
0.44
0.00
0.24
0.00
0.36
0.01
470.84
13.43
0.63
0.05
103.22
0.80


Ngn2, GAD45b


















 #6, −CytoB,
0.75
0.07
1.97
0.02
1.83
0.00
4.40
0.16
2.02
0.10
789.33
60.35
1.70
0.13
110.48
4.90


Ngn2, GAD45b


















 #7, +CytoB,
0.74
0.12
1.08
0.00
0.89
0.01
0.51
0.00
0.63
0.04
1.64
0.98
0.86
0.12
2.49
0.21


Pax6, GAD45b


















 #8, −CytoB,
0.66
0.04
2.41
0.09
2.70
0.03
4.96
0.30
3.48
0.07
0.46
0.33
2.97
1.04
0.43
0.09


Pax6, GAD45b


















 #9, +CytoB,
0.14
0.01
0.28
0.01
1.30
0.03
4.07
0.11
0.84
0.00
54768.27
6709.56
0.81
0.24
3391.96
64.63


Msi1, Ngn2,


















GAD45b


















#10, −CytoB,
0.12
0.00
0.73
0.03
5.28
0.21
50.84
1.23
4.93
0.28
17400.66
822.88
3.58
0.10
1255.76
5.27


Msi1, Ngn2


















GAD45b


















#11, +CytoB,
0.10
0.00
0.26
0.01
1.11
0.01
3.69
0.09
0.76
0.00
55588.41
1331.20
0.55
0.14
2849.96
261.51


Msi1, Ngn2


















MBD2


















#12, −CytoB,
0.44
0.01
1.47
0.06
5.49
0.14
47.30
0.11
5.50
0.31
14587.46
789.19
3.90
0.13
1424.04
39.29


Msi1, Ngn2


















MBD2


















#13, +CytoB,
1.11
0.04
1.09
0.06
0.92
0.08
0.68
0.01
0.82
0.03
63.93
2.81
1.19
0.17
17.43
1.86


GAD45b


















#14, −CytoB,
0.94
0.01
2.22
0.00
2.82
0.02
6.49
0.30
4.01
0.05
6.12
0.61
2.34
0.17
1.42
0.10


GAD45b


















#15, +CytoB,
0.83
0.00
0.83
0.05
0.36
0.01
0.16
0.01
0.36
0.00
3.42
3.74
0.63
0.37
2.18
0.12


MBD2


















#16, −CytoB,
0.68
0.02
1.55
0.04
1.57
0.05
1.47
0.01
2.00
0.00
0.52
0.29
1.45
0.15
0.55
0.04


MBD2


















#17, +CytoB,
1.10
0.01
1.16
0.03
1.37
0.01
1.12
0.06
0.86
0.06
5.59
1.48
1.07
0.27
1.70
0.46


Msi1, Ngn2


















#18, −CytoB,
0.93
0.04
2.52
0.10
3.48
0.01
9.01
0.02
4.55
0.18
1.78
1.46
3.83
0.42
0.59
0.01


Msi1, Ngn2


















#19, +CytoB,
0.20
0.03
0.36
0.01
1.25
0.05
6.68
0.31
0.72
0.02
66592.29
3481.89
2.57
0.03
4450.08
131.85


Msi1, MBD2


















#20, −CytoB,
0.12
0.00
0.64
0.03
4.70
0.22
77.51
0.11
4.12
0.11
19128.03
1542.00
8.14
0.13
999.22
24.75


Msi1, MBD2


















#21, +CytoB,
0.17
0.01
0.28
0.00
1.16
0.04
5.73
0.06
0.62
0.00
67945.51
3000.74
2.15
0.04
4736.83
11.92


Ngn2, MBD2


















#22, −CytoB,
0.17
0.00
0.78
0.03
4.32
0.08
68.89
5.26
4.01
0.04
16570.91
92.96
7.04
0.53
1427.13
13.19


Ngn2, MBD2


















#23, +CytoB,
0.71
0.05
0.79
0.06
0.87
0.01
0.63
0.06
0.67
0.04
2.86
0.70
1.08
0.08
2.08
0.11


Msi1


















#25, −CytoB,
0.66
0.04
1.92
0.17
2.03
0.02
2.77
0.02
2.68
0.02
0.32
0.12
1.85
0.65
0.58
0.04


Msi1


























Immunohistochemical Analysis


Fluorescent imunohistochemical staining was performed as previously described in Example I. Table 4 shows the percentage of Nestin and Sox2 in each condition, with the highest percentage of Sox2 (38.18±1.75%) and nestin (28.18±2.77%) positive cells observed after transfecting the cells simultaneously with both neurogenic transcription factors and in the presence of a DNA demethylator and cytochalasin B. A slight increase of Sox2 positive cells (10.42±10.27%) and nestin positive cells (4.85±1.10%) was detected following transfection with one transcription factor Msi1 and MBD2. Same tendency of nestin and Sox2 positive cells was observed following transfection with Ngn2 and MBD2. Disrupting the cell cytoskeleton with Cytochalasin B significantly enhanced reprogramming, but had no reprogramming effect on its own (Table 4).









TABLE 4







Percentage of positive cells for Sox2 and nestin after transfection of fibroblast cells


with different expression vectors with or without the presence of cytochalasin B. After


transfection the cells were cultured in proliferation medium (StemCell Technologies)


supplemented by EGF (20 ng/ml, Peprotech) and FGF (20 ng/ml, Peprotech) for two weeks at


37° C./5% CO2/5% O2. The percentage of immunopositive cells was determined by Cellomics ™


and represented as mean ± SD (n = 3-5).










% of Sox2 positive cells
% of Nestin positive cells












+CytoB
−CytoB
+CytoB
−CytoB





Untransfected cells
0.02 ± 0.01
0.01 ± 0.00
0.14 ± 0.04
0.11 ± 0.09


Ngn2
0.35 ± 0.36
0.15 ± 0.05
2.34 ± 0.99
1.04 ± 0.21


Msi1
0.23 ± 0.15
0.12 ± 0.09
1.95 ± 0.11
1.11 ± 0.18


Gadd45b
0.30 ± 0.17
0.29 ± 0.11
4.94 ± 0.25
2.33 ± 0.42


MBD2
0.22 ± 0.13
0.22 ± 0.11
 2.8 ± 0.11
1.53 ± 0.6 


Msi1/Ngn2
0.19 ± 0.13
0.32 ± 0.05
1.91 ± 0.56
2.59 ± 1.28


Msi1/MBD2
10.42 ± 10.27
 8.84 ± 11.63
4.85 ± 1.10
2.06 ± 0.08


Msi1/Gadd45b
0.06 ± 0.01
0.14 ± 0.17
0.55 ± 0.06
0.24 ± 0.11


Ngn2/MBD2
11.17 ± 0.08 
 9.07 ± 11.31
 5.7 ± 0.10
2.18 ± 0.23


Ngn2/GAdd45b
0.29 ± 0.11
0.95 ± 0.17
1.17 ± 0.54
0.98 ± 0.25


Msi1/Ngn2/MBD2
38.18 ± 1.75 
22.03 ± 1.90 
28.18 ± 2.77 
14.54 ± 0.45 


Msi1/Ngn2/Gadd45b
22.65 ± 5.03 
18.54 ± 9.40 
18.72 ± 6.26 
8.70 ± 4.51









Various DNA demethylators were tested as well for their effect on reprogramming efficiency. Cells were co-transfected with one vector (MSI1/NGN2) containing two neurogenic pCMV6-Msi1-Ngn2 factors with various DNA demethylators. Simultaneously another neurogenic factor was tested for its effect on cells de-differentiation towards NSCs, pCMV-XL-Nestin individually or in combination with pCMV-Msi1-Ngn2, pCMV-XL5-Msi1, or pCMV-XL4-Ngn2 in the presence of MBD2 as previously described in Example II.


Cells were co-transfected pCMV-Msi1-Ngn2 with different DNA demethylators (MBD1, MBD2, MBD3, MBD4, MeCP2, AICDA). Another assay was performed to assess the effect of nestin on the reprogramming efficiency; therefore cells were transfected with nestin individually or in combination with one vector containing one neurogenic factor (Msi1 or Ngn2) or both neurogenic factors in the presence of MBD2. Cells were cultured following transfection in the presence of proliferation medium supplemented with EGF (20 ng/ml), FGF (20 ng/ml), and Noggin (20 ng/ml) with and without VPA (1 mM) treatment for 12 days at 37° C., 5% CO2 and 5% O2.


Gene expression analysis and immunohitochemistry was performed to analyse neural specific gene and protein expression (βIII-tubulin, GFAP, Sox2, Nestin) as described in Example II. Transfecting cells with Msi1 and Ngn2 in the presence of various DNA demethylators revealed and confirm previous data showing that the among various DNA demethylators used in this study, MBD2 promotes the expression of neural stem genes (Sox2, GFAP, Nestin) as shown in Table 5. Furthermore, transfecting cells with nestin with and without the presence of one neurogenic factor had no effect on the reprogramming efficiency into neural stem-like cells. However co-transfection with nestin and Msi1/Ngn2/MBD2 enhanced the expression of neural stem cells genes and this increase was more pronounced in the presence of VPA.









TABLE 5







RT-PCR analysis of relative expression of neuronal precursor cell markers such as nestin, Sox2, βIII-tubulin,


and GFAP after transfection of fibroblast cells with various combinations of pCMV-Msi1-Ngn2 (MSI1/NGN2),


pCMV-XL5-Msi1, pCMV-XL4-Ngn2, pCMV-XL-Nestin with different combinations of DNA demethylators,


with and without the co-treatment with VPA.












TUBB3
GFAP
SOX2
NES
















Rel.
Std.
Rel.
Std.
Rel.
Std.
Rel.
Std.



Exp.
Dev.
Exp.
Dev.
Exp.
Dev.
Exp.
Dev.


















Day 12, Untransfected
1.00
0.11
1.00
0.05
1.01
0.16
1.00
0.13


(−VPA)










Day 12, Untransfected
1.00
0.03
1.00
0.06
1.00
0.00
1.00
0.02


(+VPA)










Day 12,
0.96
0.06
2.69
0.13
1.15
0.49
0.46
0.02


MSI1/NGN2/MBD1 (−VPA)










Day 12,
1.10
0.06
2.22
0.06
0.80
0.01
0.84
0.02


MSI1/NGN2/MBD1 (+VPA)










Day 12,
123.52
0.06
1638.53
99.86
61467.29
1487.21
31.77
0.17


MSI1/NGN2/MBD2 (−VPA)










Day 12,
232.00
0.08
1889.30
42.39
72022.15
7894.41
42.69
0.14


MSI1/NGN2/MBD2 (+VPA)










Day 12,
0.92
0.07
3.98
0.59
28.05
4.67
0.56
0.01


MSI1/NGN2/MBD3 (−VPA)










Day 12,
1.23
0.05
1.66
0.18
11.31
2.35
0.87
0.02


MSI1/NGN2/MBD3 (+VPA)










Day 12,
0.85
0.01
4.80
0.23
5.42
5.20
0.62
0.00


MSI1/NGN2/MBD4 (−VPA)










Day 12,
0.95
0.01
1.57
0.16
2.27
0.04
0.79
0.03


MSI1/NGN2/MBD4 (+VPA)










Day 12,
1.11
0.06
3.80
0.38
6.54
6.45
0.69
0.01


MSI1/NGN2/MeCP2










(−VPA)










Day 12,
1.37
0.09
1.63
0.45
10.53
10.49
1.07
0.01


MSI1/NGN2/MeCP2










(+VPA)










Day 12,
1.07
0.04
4.59
0.02
0.65
0.01
0.74
0.02


MSI1/NGN2/AICDA (−VPA)










Day 12,
1.10
0.01
2.37
0.29
1.21
0.16
0.91
0.04


MSI1/NGN2/AICDA










(+VPA)










Day 12, Msi1/MBD2
1.31
0.17
3.78
0.49
0.70
0.02
0.78
0.00


(−VPA)










Day 12, Msi1/MBD2
1.36
0.07
1.75
0.31
1.26
0.03
1.15
0.03


(+VPA)










Day 12, Ngn2/MBD2
0.85
0.06
2.93
0.51
0.79
0.05
0.58
0.02


(−VPA)










Day 12, Ngn2/MBD2
1.41
0.05
1.60
0.11
2.30
0.06
1.03
0.03


(+VPA)










Day 12, Nes/Msi1 (−VPA)
0.84
0.03
3.21
0.72
0.76
0.01
0.51
0.01


Day 12, Nes/Msi1 (+VPA)
0.86
0.09
1.82
0.30
2.14
1.02
0.94
0.01


Day 12, Nes/Ngn2 (−VPA)
0.69
0.05
2.88
0.32
0.99
0.10
0.57
0.02


Day 12, Nes/Ngn2 (+VPA)
0.88
0.01
1.53
0.19
2.71
0.02
0.83
0.03


Day 12,
111.58
0.04
1423.56
82.87
72069.27
624.51
51.52
0.12


Nes/MSI1/NGN2/MBD2










(−VPA)










Day 12,
321.00
0.04
2600.14
1.90
88932.00
708.72
82.74
0.18


Nes/MSI1/NGN2/MBD2










(+VPA)










Day 12, Nes/MSI1/NGN2
0.74
0.11
2.60
0.28
1.98
0.97
0.55
0.01


(−VPA)










Day 12, Nes/MSI1/NGN2
0.86
0.00
1.70
0.49
1.70
0.04
0.88
0.05


(+VPA)










Day 12, Nes/MBD2 (−VPA)
0.76
0.12
3.15
0.17
0.87
0.03
0.44
0.00


Day 12, Nes/MBD2
0.87
0.03
2.05
0.07
2.66
1.64
0.91
0.00


(+VPA)










Day 12, Nes/Msi1/MBD2
0.81
0.05
3.41
0.66
1.11
0.01
0.58
0.01


(−VPA)










Day 12, Nes/Msi1/MBD2
1.01
0.13
2.43
0.07
3.27
0.26
0.93
0.02


(+VPA)










Day 12, Nes/Ngn2/MBD2
1.19
0.07
5.71
1.30
4.11
0.07
0.91
0.04


(−VPA)










Day 12, Nes/Ngn2/MBD2
1.29
0.03
2.98
0.66
21.20
0.42
1.65
0.02


(+VPA)

















Immunohistochemistry analysis performed in parallel with RT-PCR data indicated that positive Sox2 cells were undetectable when transfecting the cells with Msi1/Ngn2 in the presence of MBD1, MBD3, MBD4, MeCP1, or AICADA (Table 6) and that among the different types of DNA demethylator genes tested only MBD2 plays a significant positive role in the reprogramming efficiency of HFF towards NSLCs when using the above neurogenic genes. Immunohsitochemistry analysis revealed a small increase of immunopositive Sox2 cells (89.49±3.18) after co-transfecting the cells with nestin and Msi1/Ngn2 in the presence of MBD2 (Table 6).









TABLE 6







Percentage of positive cells for Sox2 after transfection of fibroblast


cells with different expression vectors with or without the presence of


various DNA demethylators. After transfection the cells were cultured


in proliferation medium (StemCell Technologies) supplemented by EGF


(20 ng/ml, Peprotech) and FGF (20 ng/ml, Peprotech) for two weeks at


37° C./5% CO2/5% O2. The percentage of immunopositive cells was


determined by Cellomics ™ and represented as mean ± SD (n = 3-5).









% Sox2 positive ± stdv











HFF untransfected
0.13 ± 0.12


Msi-Ngn2 + MBD1
0.92 ± 0.13


Msi-Ngn2 + MBD2
79.44 ± 9.86 


Msi-Ngn2 + MBD3
1.22 ± 0.82


Msi-Ngn2 + MBD4
0.59 ± 0.03


Msi-Ngn2 + MeCP2
1.10 ± 0.25


Msi-Ngn2 + AICDA
0.69 ± 0.28


Msi + MBD2
0.79 ± 0.28


Ngn2 + MBD2
1.74 ± 1.01


Nestin + Msi
0.91 ± 0.01


Nestin + Ngn2
2.16 ± 1.44


Nestin + MSI1/NGN2 + MBD2
89.49 ± 3.18 


Nestin + MSI1/NGN2
10.20 ± 0.21 


Nestin + MBD2
0.00 ± 0.00


Nestin + Msi + MBD2
8.45 ± 0.08


Nestin + Ngn2 + MBD2
5.71 ± 0.66









Another study was designed to test the effect of various neurogenic genes on the reprogramming efficiency towards neural stem-like cells. HFF cells were cultured as described in Example I, and transfected using the Nucleofector™® 96-well Shuttle® Device (Lonza) following procedure described in Example IV, except for the untreated HFF control and the untransfected HFF control (for determining the effect of the complete media & compound treatments on the cells). The cells that had been pre-treated with VPA and 5-Aza and the untreated cells were transfected with the mixes of DNA as described in Table 7. The cells were plated on Laminin-coated plates and incubated at 37° C., 5% CO2. Media was changed daily according to Table 7. Cells were analysed at day 3, 7, 12 by immunohistochemistry analysis and at Day 9 by gene array for multipotent and pluripotent gene expression.


Gene Array Analysis


An additional batch of cells treated according to 0a and 1a in Table 7 was analyzed at Day 9, along with HFFs, hNPCs, and passage 5 NSLCs (frozen from previous experiments from Example III) by the Pluripotency Gene Array (ABI) (Tables 8a and b) and a set of genes (Table 8c) to determine the gene expression profile of select pluripotency, ectoderm, endoderm, mesoderm, and neural lineage genes in passage 1 and passage 5 NSLCs compared to HFFs (from which they were created) and normal human neuroprogenitor cells (hNPCs). The results in Table 8 indicate that all the genes related to neural stem cells (some of the significantly expressed pluripotency markers and mesendoderm markers are also expressed in neural stem cells) and the neural lineage were significantly expressed in NSLCs as opposed to HFFs, and the expression pattern was a bit different from hNPCs indicating that NSLCs are similar to, but not identical, to the hNPCs tested. Passage 5 NSLCs 5 had a higher expression of stemness genes than Passage 1 NSLCs. hNPCs had a higher expression of neuronally committed genes than NSLCs, indicting their neuroprogenitor status versus the greater stemness status of NSLCs.









TABLE 7







Plasmids and media composition from day 1 to day 12.













From day −2
Plasmids transfected






to day 0
at day 0
From day 1 to day 3
From day 3 to day 4
From day 4 to day 12





 0a
Untreated
No plasmid
Neural proliferation medium +
Neural proliferation
Neural proliferation





Egf + Fgf-2
medium + Egf + Fgf-2
medium + Egf + Fgf-2


 1a
Untreated
Msi1/Ngn2 + pCMV6-
Neural proliferation medium +
Neural proliferation
Neural proliferation




XL5-MBD2
Egf + Fgf-2
medium + Egf + Fgf-2
medium + Egf + Fgf-2


 1b
Untreated
Msi1/Ngn2 + pCMV6-
Neural proliferation medium +
Neural proliferation
Neural proliferation




XL5-MBD2
Egf + Fgf-2
medium + Egf + Fgf-2
medium + Egf + Fgf-2 +







SHH


 1c
Untreated
Msi1/Ngn2 + pCMV6-
Neural proliferation medium +
Neural proliferation
Neural proliferation




XL5-MBD2
Egf + Fgf-2 + Noggin
medium + Egf + Fgf-2 +
medium + Egf + Fgf-2 +






Noggin
Noggin







(day 1 today 7)/Forskolin







(day 7 to day 12)


 1d
Untreated
Msi1/Ngn2 + pCMV6-
Neural proliferation medium +
Neural proliferation
Neural proliferation




XL5-MBD2
Egf + Fgf-2
medium + Egf + Fgf-2
medium + Egf + Fgf-2


 1e
Untreated
Msi1/Ngn2 + pCMV6-
Neural proliferation medium +
Neural proliferation
Neural proliferation




XL5-MBD2
Egf + Fgf-2
medium + Egf + Fgf-2
medium + Egf + Fgf-2


 1f
Untreated
Msi1/Ngn2 + pCMV6-
Neural proliferation medium +
Neural proliferation
Neural proliferation




XL5-MBD2
Egf + Fgf-2
medium + Egf + Fgf-2
medium + Egf + Fgf-2


 2
Untreated
Msi1/Ngn2 + pCMV6-
Neura l proliferation medium +
Neural proliferation
Neural proliferation




XL5-MBD2
Egf + Fgf-2 + CytoB
medium + Egf + Fgf-2 +
medium + Egf + Fgf-2






CytoB



 3
Untreated
Msi1/Ngn2
Neural proliferation medium +
Neural proliferation
Neural proliferation





Egf + Fgf-2 + VPA + 5-Aza
medium + Egf + Fgf-2
medium + Egf + Fgf-2


 4
Untreated
Msi1/Ngn2
Neural proliferation medium +
Neural proliferation
Neural proliferation





Egf + Fgf-2
medium + Egf + Fgf-2
medium + Egf + Fgf-2


 5
Untreated
pCMV6-XL5-Musashi
Neural proliferation medium +
Neural proliferation
Neural proliferation





Egf + Fgf-2
medium + Egf + Fgf-2
medium + Egf + Fgf-2


 6
Untreated
pCMV6-XL5-Musashi
Neural proliferation medium +
Neural proliferation
Neural proliferation





Egf + Fgf-2 + Noggin
medium + Egf + Fgf-2 +
medium + Egf + Fgf-2 +






Noggin
Noggin + Forskolin


 7
Untreated
pCMV6-XL5-Musashi
Neural proliferation medium +
Neural proliferation
Neural proliferation





Egf + Fgf-2 + VPA + 5-Aza
medium + Egf + Fgf-2
medium + Egf + Fgf-2


 8
Untreated
pCMV6-XL5-Musashi
Neural proliferation medium +
Neural proliferation
Neural proliferation





Egf + Fgf-2 + Noggin + VPA +
medium + Egf + Fgf-2 +
medium + Egf + Fgf-2 +





5-Aza
Noggin
Noggin + Forskolin


 9
Untreated
pCMV6-XL5-ZIC1 +
Neural proliferation medium +
Neural proliferation
Neural proliferation




pCMV6-XL4-Ngn2 +
Egf + Fgf-2
medium + Egf + Fgf-2
medium + Egf + Fgf-2




pCMV6-XL5-MBD2





10
Untreated
pCMV6-XL5-SOX1 +
Neural proliferation medium +
Neural proliferation
Neural proliferation




pCMV6-XL4-Ngn2 +
Egf + Fgf-2
medium + Egf + Fgf-2
medium + Egf + Fgf-2




pCMV6-XL5-MBD2





11
Untreated
pCMV6-XL5-Sox2 +
Neural proliferation medium +
Neural proliferation
Neural proliferation




pCMV6-XL4-Ngn2 +
Egf + Fgf-2
medium + Egf + Fgf-2
medium + Egf + Fgf-2




pCMV6-XL5-MBD2





12
Untreated
pCMV6-XL5-Nanog +
Neural proliferation medium +
Neural proliferation
Neural proliferation




pCMV6-XL4-Ngn2 +
Egf + Fgf-2
medium + Egf + Fgf-2
medium + Egf + Fgf-2




pCMV6-XL5-MBD2





13
Untreated
pCMV6-XL4-Oct4 +
Neural proliferation medium +
Neural proliferation
Neural proliferation




pCMV6-XL4-Ngn2 +
Egf + Fgf-2
medium + Egf + Fgf-2
medium + Egf + Fgf-2




pCMV6-XL5-MBD2





14
VPA + 5-Aza
Msi1/Ngn2
Neural proliferation medium +
Neural proliferation
Neural proliferation



pre-treated

Egf + Fgf-2
medium + Egf + Fgf-2
medium + Egf + Fgf-2


15
VPA + 5-Aza
pCMV6-XL5-Musashi
Neural proliferation medium +
Neural proliferation
Neural proliferation



pre-treated

Egf + Fgf-2 + VPA + 5-Aza
medium + Egf + Fgf-2
medium + Egf + Fgf-2


16
VPA + 5-Aza
pCMV6-XL5-Musashi
Neural proliferation medium +
Neural proliferation
Neural proliferation



pre-treated

Egf + Fgf-2 + Noggin + VPA +
medium + Egf + Fgf-2 +
medium + Egf + Fgf-2 +





5-Aza
Noggin
Noggin + Forskolin


17
VPA + 5-Aza
pCMV6-XL5-Musashi +
Neural proliferation medium +
Neural proliferation
Neural proliferation



pre-treated
pCMV6-XL5-MBD2
Egf + Fgf-2 + Noggin + VPA +
medium + Egf + Fgf-2 +
medium + Egf + Fgf-2 +





5-Aza
Noggin
Noggin + Forskolin


18
VPA + 5-Aza
pCMV6-XL4-Ngn2
Neural proliferation medium +
Neural proliferation
Neural proliferation



pre-treated

Egf + Fgf-2 + Noggin + VPA +
medium + Egf + Fgf-2 +
medium + Egf + Fgf-2 +





5-Aza
Noggin
Noggin + Forskolin


19
VPA + 5-Aza
pCMV6-XL5-MBD2
Neural proliferation medium +
Neural proliferation
Neural proliferation



pre-treated

Egf + Fgf-2 +Noggin + VPA +
medium + Egf + Fgf-2 +
medium + Egf + Fgf-2 +





5-Aza
Noggin
Noggin + Forskolin


20
VPA + 5-Aza
Ngn2 + pCMV6-XL5-
Neural proliferation medium +
Neural proliferation
Neural proliferation



pre-treated
MBD2
Egf + Fgf-2 + Noggin + VPA +
medium + Egf + Fgf-2 +
medium + Egf + Fgf-2 +





5-Aza
Noggin
Noggin+ Forskolin


21
VPA + 5-Aza
No plasmid
Neural proliferation medium +
Neural proliferation
Neural proliferation



pre-treated

Egf + Fgf-2 + Noggin + VPA +
medium + Egf + Fgf-2 +
medium + Egf + Fgf-2 +





5-Aza
Noggin
Noggin + Forskolin





* Immunohistochemistry analysis performed in parallel with RT-PCR data indicated among all the combinations in this experiment where no cytochalasin B was used, positive Sox2 cells were detectable only in cells transfected with Msi1/Ngn2 with and without MBD2.













TABLE 8a







Results for Human Stem Cell Pluripotency Array (n = 4 for each sample)-Embryonic Stem Cell Markers, Germ Cell Markers


and Trophoblast Markers. For Relative Expression calculations, each sample was normalized to the average Ct of the 6 housekeeping genes


(ACTB, 18S, CTNNB1, EEF1A1, GAPD, RAF1), and calibrated to the Untreated HFF (Passage 8) control. Relative Expression values with


asterisk (*) indicate values with significant up or down-regulation (>2-fold or <0.5-fold). For these samples, for Ct values <35 is considered that


the expression of the gene is adequate for quantification. For the Relative Expression values that are >2-fold or <0.5-fold but without asterisk,


the values could have significant error due to the low expression of the gene (Ct >35), and thus the up or down-regulation could be merely a


result of the high standard deviation of the high Ct values of the genes, or fluctuations in the housekeeping genes. As for the Relative


Expression values that are between 0.5-fold and 2-fold, it indicates no significant change in the expression of the gene for these samples.






















MSI1/NGN2/MBD2-


















Untransfected


transfected HFF
Neural stem-like cells,













Untreated HFF
HFF
hNPC neurospheres
(Day 9)
NSLC



(Passage 8)
(Day 9)
(Passage 4)
(NSLC, Passage 1)
(Passage 5)


















Rel.
Std.
Rel.
Std.
Rel.
Std.
Rel.
Std.
Rel.
Std.


Gene
Exp.
Dev.
Exp.
Dev.
Exp.
Dev.
Exp.
Dev.
Exp.
Dev.




















Embryonic Stem












cell markers












BRIX
1.03
0.30
0.47
0.10
0.78
0.22
0.78
0.25
0.83
0.10


CD9
1.01
0.18
2.46*
0.62
1.86
0.29
2.24*
0.19
1.00
0.39


COMMD3
1.08
0.53
0.94
0.36
0.94
0.40
0.98
0.40
1.05
0.59


DNMT3B
1.07
0.50
0.34*
0.14
2.96*
0.84
1.90
0.41
0.35
0.34


EBAF/LEFTY2
1.00
0.00
2.10
0.00
7.95
4.60
7.79
4.88
70.56*
26.12


FGF4
1.00
0.00
2.10
0.00
1.44
0.00
1.54
0.00
1.37
0.00


FOXD3
1.00
0.00
2.10
0.00
1.44
0.00
7.13
11.18
222.41*
63.43


GABRB3
1.06
0.38
4.22*
0.71
66.65*
12.52
40.01*
4.54
1.62
0.98


GAL
1.00
0.04
9.73*
0.32
0.03*
0.01
4.25*
0.46
2.89*
0.83


GBX2
1.00
0.09
0.04
0.05
45.28*
4.59
90.92*
12.14
55.22*
2.36


GDF3
1.00
0.00
2.10
0.00
1.44
0.00
1.54
0.00
1.37
0.00


GRB7
1.02
0.24
0.30*
0.16
0.05*
0.04
0.29*
0.08
0.06*
0.08


IFITM1
1.01
0.17
63.96*
6.04
0.04*
0.01
21.80*
4.31
3.35*
0.63


IFITM2
1.00
0.12
3.84*
0.89
0.02*
0.00
0.65
0.11
0.43*
0.09


IL6ST
1.01
0.21
2.19*
0.39
0.85
0.14
1.59
0.26
0.75
0.06


IMP2
1.11
0.66
1.65
0.92
1.06
0.48
0.78
0.26
1.96
0.97


KIT
1.02
0.26
1.15
0.30
0.02*
0.00
0.31*
0.09
0.00*
0.00


LEFTB
1.61
1.15
12.28*
7.84
5.45
3.15
5.58
2.65
8.96*
4.12


LIFR
2.29
3.57
13.51
16.55
6.31
7.24
12.98
9.81
2.85
4.31


LIN28
4.69
8.62
5.25
8.88
28.38*
19.25
26.97*
8.68
32.13*
14.32


NANOG
1.71
1.97
18.61
16.43
64.94*
28.32
70.87*
9.88
5.87
3.52


NOG
1.03
0.27
0.18*
0.08
0.18*
0.06
0.22*
0.06
0.02*
0.00


NR5A2
2.04
2.05
6.85
8.80
0.38
0.00
3.89
4.36
0.36
0.00


NR6A1
1.11
0.66
1.37
0.31
5.08*
0.37
2.71*
0.63
2.04*
0.17


PODXL
1.00
0.07
0.01*
0.01
0.80
0.11
2.09*
0.04
6.49*
0.64


POU5F1
1.01
0.13
0.27*
0.17
0.89
0.09
0.71
0.09
0.19*
0.06


PTEN
1.00
0.02
2.68*
0.29
0.87
0.04
1.07
0.12
0.80
0.14


RESET
1.01
0.12
1.53
0.17
0.94
0.18
1.04
0.21
1.10
0.24


SEMA3A
1.00
0.11
1.99
0.19
0.66
0.05
1.05
0.11
0.90
0.16


SFRP2
1.11
0.56
122.57*
14.57
3480.98*
702.37
1500.84*
272.46
2.75
2.85


SOX2
1.00
0.00
2.45
0.70
127594.46*
11326.91
88615.76*
15003.70
137424.37*
26622.02


TDGF1
1.41
1.28
2.92
0.68
6.13
1.52
5.46
1.95
2.20
1.51


TERT
1.00
0.00
2.10
0.00
10.81
18.75
10.74
18.41
6506.88*
893.84


TFCP2L1
1.00
0.00
2.10
0.00
7.84
12.80
32.49
10.01
1.37
0.00


UTF1
1.00
0.00
8.21
12.23
27.86
19.24
1.54
0.00
30.68
25.94


XIST
1.00
0.00
2.10
0.00
24609.46*
4337.83
22637.95*
3988.10
1.37
0.00


ZFP42
1.24
1.06
12.38
12.58
1.41
0.78
2.01
1.85
1.76
0.93


Germ cell markers












DDX4
1.00
0.00
2.10
0.00
1.44
0.00
5.84
8.60
19.11
20.49


SYCP3
1.58
1.95
11.97
8.01
11.12
3.46
15.46
11.65
2.25
2.85


Trophoblast












markers












CDX2
1.00
0.00
2.10
0.00
1.44
0.00
1.54
0.00
1.37
0.00


CGB
1.02
0.24
2.08*
0.74
0.15*
0.16
0.57
0.41
0.09*
0.17


EOMES
1.51
1.14
0.33
0.00
0.71
0.97
0.24
0.00
0.77
1.12


GCM1
2.61
2.80
0.42
0.00
3.25
5.92
5.68
1.44
1.47
2.38


KRT1
1.00
0.00
2.10
0.00
1.44
0.00
1.54
0.00
1.37
0.00
















TABLE 8b







Results for Human Stem Cell Pluripotency Array (n = 4 for each sample)-Ectoderm, Endoderm and Mesoderm Markers. For


Relative Expression calculations, each sample was normalized to the average Ct of the 6 housekeeping genes (ACTB, 18S, CTNNB1, EEF1A1,


GAPD, RAF1), and calibrated to the Untreated HFF (Passage 8) control. Relative Expression values with asterisk (*) indicate values with


significant up or down-regulation (>2-fold or <0.5-fold). For these samples, for Ct values <35 is considered that the expression of the gene is


adequate for quantification. For the Relative Expression values that are >2-fold or <0.5-fold but without asterisk, the values could have significant


error due to the low expression of the gene (Ct >35), and thus the up or down-regulation could be merely a result of the high standard deviation of


the high Ct values of the genes, or fluctuations in the housekeeping genes. As for the Relative Expression values that are between 0.5-fold and


2-fold, it indicates no significant change in the expression of the gene for these samples.























MSI1/NGN2/MBD2-


















Untransfected
hNPC
transfected HFF
Neural stem-like













Untreated HFF
HFF
neurospheres
(Day 9)
cells, NSLC



(Passage 8)
(Day 9)
(Passage 4)
(NSLC, Passage 1)
(Passage 5)


















Rel.
Std.
Rel.
Std.
Rel.
Std.
Rel.
Std.
Rel.
Std.


Gene
Exp.
Dev.
Exp.
Dev.
Exp.
Dev.
Exp.
Dev.
Exp.
Dev.




















Ectoderm markers












CRABP2
1.04
0.35
26.14*
4.28
0.01*
0.01
21.11*
2.80
0.21*
0.05


FGF5
1.01
0.15
0.21*
0.07
0.00*
0.00
0.10*
0.02
0.00*
0.00


GFAP
1.22
0.84
9.89*
5.46
798.04*
162.37
487.99*
79.84
12052.09*
2984.71


ISL1
1.01
0.12
2.19*
0.27
0.02*
0.02
0.42*
0.08
0.00*
0.00


NES
1.10
0.58
3.19*
0.95
6.78*
0.95
3.84*
0.19
7.47*
0.54


NEUROD1
1.00
0.00
2.10
0.00
1.44
0.00
2.32
1.57
25.54
6.42


OLIG2
1.00
0.00
2.10
0.00
124181.50*
14735.13
80826.42*
27820.32
36172.45*
3145.67


PAX6
1.11
0.48
0.06*
0.00
533.31*
120.59
326.02*
33.14
371.42*
77.50


SYP
1.02
0.25
5.22*
2.10
229.40*
22.54
143.94*
17.41
16.48*
4.47


TH
1.00
0.00
9.52
14.86
1218.08*
186.74
217.79*
45.71
348.31*
150.50


Endoderm markers












AFP
1.00
0.00
2.10
0.00
1.44
0.00
1.54
0.00
1.37
0.00


FN1
1.00
0.06
1.41
0.10
0.02*
0.00
1.96
0.19
0.00*
0.00


FOXA2
1.00
0.00
150.00*
55.92
1.44
0.00
1.54
0.00
1.37
0.00


GATA4
1.00
0.00
11.93
19.67
7.22
11.56
9.14
12.35
1.37
0.00


GATA6
1.00
0.09
0.37*
0.17
0.00*
0.00
0.44*
0.04
0.02*
0.01


GCG
1.00
0.00
7.96
11.74
1.44
0.00
33.59*
22.17
1.37
0.00


IAPP
1.00
0.00
2.10
0.00
1.44
0.00
1.54
0.00
1.37
0.00


INS
1.00
0.00
2.10
0.00
1.44
0.00
12.67
22.26
1.37
0.00


IPF1
1.00
0.00
2.10
0.00
1.44
0.00
1.54
0.00
1.37
0.00


LAMA1
1.00
0.11
4.42*
0.86
78.49*
6.82
43.99*
2.79
46.49*
16.59


LAMB1
1.02
0.26
12.51*
2.40
0.29*
0.09
2.27*
0.77
3.89*
1.12


LAMC1
1.00
0.10
2.82*
0.10
1.54
0.33
3.01*
0.94
1.31
0.30


NODAL
1.00
0.00
12.16
11.62
16.27
11.25
1.54
0.00
1.37
0.00


PAX4
1.00
0.00
6.77
9.35
1.44
0.00
1.54
0.00
1.37
0.00


PTF1A
1.00
0.00
2.10
0.00
1.44
0.00
1.54
0.00
1.37
0.00


SERPINA1
1.03
0.30
0.79
0.53
0.24
0.00
1.52
1.17
0.99
0.68


SOX17
1.00
0.00
2.10
0.00
1.44
0.00
1.35
5.63
1.37
0.00


SST
1.25
1.00
52.58*
10.67
0.55
0.36
48.97*
8.70
0.92
0.42


TAT
1.00
0.00
2.10
0.00
255.86*
84.52
106.04*
45.87
1.37
0.00


Mesoderm markers












ACTC
1.04
0.35
0.01*
0.00
0.02*
0.01
0.05*
0.01
0.01*
0.01


OD34
1.67
1.69
501.85*
61.88
45.17*
27.01
113.96*
39.39
13203.40*
5385.80


CDH5
1.00
0.00
4.12
4.06
16.69
8.07
32.41*
20.31
13447.65*
3220.80


COL1A1
1.01
0.12
2.28*
0.41
0.00*
0.00
0.50*
0.05
0.02*
0.00


COL2A1
3.56
6.27
103.52*
37.78
1813.86*
236.76
873.19*
259.80
3815.72*
839.02


DES
1.00
0.07
1.94
0.33
1.09
0.33
0.87
0.07
0.22*
0.08


FLT1
1.01
0.15
0.68
0.29
0.00
0.00
0.46*
0.05
0.00*
0.00


HBB
3.08
4.01
0.39
0.00
0.27
0.00
0.29
0.00
0.26
0.00


HBZ
1.14
0.63
3.53
1.32
0.25
0.22
0.61
0.63
2.88
1.20


HLXB9
1.00
0.00
2.10
0.00
59.80*
16.35
24.94
3.14
35.12
40.50


MYF5
1.77
1.87
0.69
0.00
0.47
0.00
0.51
0.00
0.45
0.00


MYOD1
1.71
2.27
1.22
0.00
0.83
0.00
0.89
0.00
0.80
0.00


NPPA
1.00
0.00
2.10
0.00
96.60*
76.23
18.97
26.98
32.37
10.96


PECAM1
1.00
0.00
1041.24*
150.95
31.30*
24.22
964.70*
200.82
7305.03*
1127.69


RUNX2
1.01
0.12
1.76
0.37
0.09*
0.02
0.78
0.23
1.18
0.27


T
1.00
0.00
2.10
0.00
1.44
0.00
1.54
0.00
1.37
0.00


WT1
2.09
3.13
1.11
0.00
0.76
0.00
2.72
3.82
4.24
4.21
















TABLE 8c







Results for relative expression of Embryonic Stem Cell, Ectoderm, Endoderm/mesoderm, and neuronal markers in untransfected


and transfected HFF with Msi1/Ngn2/MBD2 calibrated to untreated HFF (passage 8). Genes with asterisk (*) indicate that the Ct


values of the test samples are within the quantifiable range (Ct <35), suggesting the expression of the gene in the test sample is


adequate for quantification. For genes without asterisk, the values may be inaccurate due to the low expression of the gene (Ct >35)


and thus the up or down-regulation is merely a result of the high standard deviation of the high Ct values of the genes, or fluctuation


of the housekeeping genes; the trend for these samples may be correct, but the absolute relative expression values may not.


Expression of NGN3 and LIN28 were also tested but these two genes were not expressed in any of the test samples (data not shown).


RT-PCR revealed a significant increase of ectoderm and neuronal markers.
















Untransfected
hNPC
MSI1/NGN2/MBD2-
Neural stem-like cells,













Untreated HFF
HFF
neurospheres
transfected HFF (Day 9)
NSLC



(Passage 8)
(Day 9)
(Passage 4)
(NSLC, Passage 1)
(Passage 5)


















Rel.
Std.
Rel.
Std.
Rel.
Std.
Rel.
Std.
Rel.
Std.


Gene
Exp.
Dev.
Exp.
Dev.
Exp.
Dev.
Exp.
Dev.
Exp.
Dev.




















Embryonic Stem Cell












Markers












OCT4*
1.04
0.38
7.27
0.81
6.26
0.05
6.63
0.51
3.15
0.58


OCT4 (5′UTR)
1.04
0.41
0.08
0.00
2.07
0.11
1.82
0.53
0.55
0.59


NANOG (5′UTR)
1.02
0.32
19.29
2.23
11.27
0.89
16.73
6.86
9.94
6.32


FBXO15*
1.05
0.46
2.58
0.45
3.57
0.23
5.89
1.22
1.13
0.39


ALPL*
1.03
0.33
0.57
0.73
652.20
46.60
194.23
10.82
13.04
4.04


SALL4*
1.02
0.25
9.20
1.35
9.76
0.62
15.84
0.92
2.35
0.55


NR0B1 (DAX1)*
1.01
0.19
18.62
4.70
2.64
0.11
11.59
3.17
0.06
0.00


Ectoderm Markers












ZIC1*
1.01
0.24
2.01
0.25
1889.80
93.48
1158.21
80.43
156.40
12.64


SOX1*
1.00
0.01
2.05
0.06
1776.83
128.63
1052.75
243.07
47.98
2.12


CDH1 (E-cadherin)*
1.00
0.01
2.05
0.06
264.59
6.22
59.14
7.57
18.20
3.73


p63
1.00
0.01
68.37
72.49
18.01
5.33
39.72
12.76
37.83
6.76


MSX1
1.00
0.05
4.19
0.56
0.10
0.01
1.53
0.35
0.09
0.00


NOTCH1*
1.00
0.07
1.26
0.08
7.38
1.20
4.51
0.54
4.75
0.26


SOX2*
1.00
0.01
2.50
0.57
340909.59
5659.15
194495.82
17929.15
219269.76
31399.68


SOX2 (3′UTR)*
1.00
0.01
7.74
8.11
864191.09
60204.44
452684.80
26457.70
618245.01
7107.48


Mesoderm/Endoderm












Markers












CXCR4*
1.05
0.46
12.45
5.64
5048.23
172.14
2763.82
30.29
3773.11
78.89


Neuronal markers












MAP2*
1.01
0.17
2.98
0.20
155.33
9.08
88.82
6.48
27.38
0.13


TUBB3*
1.00
0.04
0.38
0.02
1.15
0.05
0.89
0.05
0.98
0.09


ASCL1 (MASH1)*
1.29
1.16
11.19
0.22
42618.46
68.52
23554.16
1588.45
31358.79
2301.26


NGN2*
1.00
0.01
2.05
0.06
19.45
6.64
247883.48
16409.80
968.11
191.73


NGN2 (3′UTR)*
1.83
2.17
1.17
0.76
13.39
5.10
8.45
1.75
539.02
59.72


MSI1*
1.00
0.01
263.87
70.10
100376.36
81.45
479098.05
2281.62
116105.29
2745.03


MSI1 (3′UTR)*
1.01
0.20
13.61
2.00
3601.96
345.79
2163.87
59.84
3698.14
160.78


ACHE*
1.00
0.00
2.00
0.26
25.00
3.71
12.84
0.84
21.30
0.30


Glia markers












CNP*
1.01
0.18
1.43
0.10
3.48
0.58
2.69
0.12
1.93
0.07


SOX9*
1.00
0.04
3.54
0.06
88.25
9.71
41.11
2.70
26.96
0.53





Note that custom primers (5′UTR) for detecting endogenous gene expression are generally not as sensitive and/or effective as standard primers (from the supplier's (Origene) catalog) that dtect overall gene expression (both endogenous and exogenous) of a particular gene.






In another part of the experiment, another batch of cells that were transfected with Msi1/Ngn2+pCMV6-XL5-MBD2 were plated on Poly-Ornithine (30 min at RT) and Laminin (1 h at RT) coated plates in CDM II medium in 5 different wells. On day 1, medium in two of the wells was switched to the same medium as in condition 1a (Table 7) until day 12. Medium was changed daily until day 12, at which point it was switched to either NS-A Differentiation Medium (StemCell Technologies) or NbActive4 (BrainBits™) medium that were both supplemented with BDNF (20 ng/ml), NT-3 (20 ng/ml), NGF (20 ng/ml), Retinoic acid (5 μM), Noggin (20 ng/ml) and Forskolin (10 μM). These cells showed a typical neural stem-like cell morphology by day 7, and proliferated until day 12. During the exposure to either of the two differentiation media, these NSLC changed to a more neuronal and glial phenotype as shown in the bright field pictures (FIG. 21), but only expressed GFAP by Day 17 (FIG. 22).


For the other three wells, on day 1 medium was switched to either NS-A Differentiation Medium (StemCell Technologies), NbActive4 (BrainBits), or CDM II medium; these first two were supplemented with the same cytokines as previously described but with the addition of Fgf-2 (20 ng/ml). On day 12, Fgf-2 was removed from the first two differentiation media while cells in the CDM II medium were switched to the NS-A Differentiation Medium (StemCell Technologies) supplemented with cytokines without Fgf-2. Between day 12 and day 17, media was changed every two to three days. During the first 12 days of culture, cells in all 3 media developed into a mix of more spindle shaped cells compared to untransfected fibroblasts and some into cells with a NSLC morphology; upon removal of Fgf-2 cell morphology turned into a very pronounced neuronal shape as well as glial cells with a network established between cells as shown in the bright field pictures (FIG. 21) that expressed GFAP and 1111-tubulin by Day 17 (FIG. 22).


An additional study was designed to assess the effect of Msi1, Ngn2 and MBD2 on their endogenous proteins levels in reprogrammed cells. Cells were transfected with the MSI1/NGN2 vector and MBD2 as previously described and cultured in proliferation condition at 37° C., 5% CO2 and 5% O2. Samples were collected at various time points from Day 2-10 and analyzed by RT-PCR to investigate the expression of endogenous genes and the expression of neural stem cell and neuronal genes at different time points. RT-PCR revealed a gradual loss of total Msi1, Ngn2 and MBD2 gene expression starting from Day 2 to Day 10, with the increase in MBD2 expression relative to control having been almost completely lost by Day 5. This decrease was associated with a significant activation of endogenous Msi1 and Ngn2 on Day 5, with another jump in endogenous gene expression at Day 9 (Table 9). A significant increase in Sox2 expression was detected at Day 4, and the expression of this ectoderm/neural stem cell/neuronal gene continued to increase with each subsequent timepoint (Table 10). GFAP (a neural stem cell and astrocyte marker) was slightly elevated already from Day 2 onwards, but significantly increased on Day 5 with a large jump in gene expression at Day 7 analysis timepoint and stayed at this expression level for the rest of the study period. Expression of the neural stem cell marker Nestin also started to slowly increase from Day 5 onwards. Expression of the neuronal genes βIII-tubulin (TUBB3) and Map2b were slightly elevated already from Day 2 onwards, but significantly increased on Day 5 onwards. Expression of a marker for acetylcholine receptors (found in neurons), acetylcholine esterase (ACHE), was also slightly elevated from Day 2 onwards, but did not significantly increased until Day 7 onwards. It should be noted that among the neural stem cell markers that were analyzed, the relative expression of Sox2 was highly and early expressed which could then be directly or indirectly interact with the exogenous Msi/Ngn2 and/or other genes in the activation of Nestin, GFAP, and endogenous Msi1 and Ngn2 and other genes that promote the reprogramming and cell fate change, as well as the activation of neuronal genes like βIII-tubulin (TUBB3), Map2b, and ACHE.









TABLE 9







RT-PCR analysis of exogenous and endogenous relative expression of Msi1, Ngn2 and MBD2 from Day 2-10 after transfection of


fibroblast cells with pCMV-Msi1-Ngn2(Msi1/Ngn2) and MBD2 and cultured for 10 days in proliferation medium. Cells were


collected at different time point to analyse endogenous gene expression.


















Endogenous


Endogenous

Endogenous














MSI1
MSI1
NGN2
NGN2
MBD2
MBD2




















Rel.
Std.
Rel.
Std.
Rel.
Std.
Rel.
Std.
Rel.
Std.
Rel.
Std.



Exp.
Dev.
Exp.
Dev.
Exp.
Dev.
Exp.
Dev.
Exp.
Dev.
Exp.
Dev.






















#1 Day 12 Untransfected HFF
1.01
0.18
1.04
0.38
1.01
0.15
1.01
0.15
1.01
0.21
1.00
0.14


#2 Day 12 HFF Msi1/Ngn2 +
1102.17
91.80
620.56
19.49
2208.36
375.09
51.09
14.69
1.09
0.00
0.83
0.06


MBD2














#3 Day 18 HFF Msi1/Ngn2 +
1470.36
164.35
950.07
152.50
71.57
52.59
122.66
39.63
1.21
0.02
0.73
0.08


MBD2














#4 Untransfected Keratinocytes
1.49
N/A
1.01
N/A
1.00
N/A
1.00
N/A
1.02
N/A
1.00
N/A


#5 Day 12 Keratinocytes
4142.78
872.87
364.20
60.90
4656.42
232.63
102.01
3.18
0.40
0.14
0.74
0.30


Msi1/Ngn2 + MBD2














#6 Day 18 Keratinocytes
4830.20
291.17
486.38
19.59
50.01
6.99
43.08
13.78
0.40
0.01
0.67
0.01


Msi1/Ngn2 + MBD2














#7 Untransfected CD34 +
1.01
0.19
1.00
0.01
1.01
0.16
1.17
0.87
1.00
0.02
1.00
0.07


#8 Day 18 CD34 +
3969.52
286.36
147.99
7.08
2.03
0.55
3.72
1.23
0.43
0.06
0.90
0.18


Msi1/Ngn2 + MBD2














hNPC (14 Oct. 09, EXP0067)
7574.57
234.74
1141.14
49.15
8.18
5.64
6.27
5.19
0.58
0.00
2.35
0.03
















TABLE 10







RT-PCR analysis of relative expression of Nestin, Map2b, TUBB3, ACHE, GFAP, and Sox2 from Day 2-10 after


transfection of fibroblast cells with pCMV-Msi1-Ngn2 (Msi1/Ngn2) and MBD2 and cultured for 10 days


in proliferation medium. Cells were collected at different time point to analyse endogenous gene expression.














NES
MAP2
TUBB3
ACHE
GFAP
SOX2




















Rel.
Std.
Rel.
Std.
Rel.
Std.
Rel.
Std.
Rel.
Std.
Rel.
Std.



Exp.
Dev.
Exp.
Dev.
Exp.
Dev.
Exp.
Dev.
Exp.
Dev.
Exp.
Dev.






















 #1 Untransfected Day 2
1.00
0.04
1.00
0.01
1.00
0.03
1.00
0.08
1.01
0.23
1.17
0.87


 #2 Msi1/Ngn2 + MBD2/
0.88
0.01
8.59
0.18
1.38
0.03
5.71
1.06
4.56
0.08
1.26
0.82


+Noggin Day 2














 #3 Untransfected Day 3
1.38
0.07
0.66
0.03
0.40
0.02
1.36
0.06
1.95
0.38
2.34
2.29


 #4 Msi1-Ngn2 + MBD2/
1.39
0.08
4.31
0.24
0.79
0.09
6.03
0.60
4.66
0.02
0.96
0.10


+Noggin Day 3














 #5 Untransfected Day 4
2.43
0.23
1.78
0.11
0.44
0.01
2.70
0.02
3.76
0.86
0.93
0.01


 #6 Msi1/Ngn2 + MBD2/
1.91
0.06
2.81
0.20
0.64
0.02
6.76
0.64
8.67
1.06
5.37
6.06


+Noggin Day 4














 #7 Untransfected Day 5
1.40
0.05
1.13
0.04
0.41
0.03
1.17
0.37
5.44
0.02
15.03
8.77


 #8 Msi1-Ngn2 + MBD2/
4.31
0.08
71.60
6.43
1.34
0.01
7.60
0.18
42.28
2.94
66377.25
4089.77


+Noggin Day 5














 #9 Untransfected Day 7
2.24
0.00
4.02
0.15
1.22
0.05
1.10
0.48
7.61
1.24
1.34
0.02


#10 Msi1/Ngn2 + MBD2/
3.07
0.11
48.10
2.85
2.70
0.05
13.11
1.30
3271.10
149.81
44255.59
2004.08


+Noggin Day 7














#11 Untransfected Day 9
4.37
0.23
14.55
0.96
1.75
0.14
3.35
0.36
15.95
0.23
429.09
119.98


#12 Msi1-Ngn2 + MBD2/
7.97
0.16
123.55
3.27
2.79
0.12
16.59
0.03
3152.25
3.31
114149.70
3372.20


+Noggin Day 9














#13 Untransfected Day 10
3.48
0.44
10.03
0.37
1.63
0.21
3.20
0.81
5.64
1.92
14.66
5.03


#14 Msi1/Ngn2 + MBD2/
7.48
0.22
100.25
6.66
2.87
0.03
17.49
1.35
3374.03
22.47
101105.49
3996.44


+Noggin Day 10





















Example IV

Comparison of the Nucleofector™® II Device and the Nucleofector™® 96-Well Shuttle® Device in the Reprogramming of HFF into NSLC in Adherent and Floating Conditions.


HFF cells were cultured as described in Example I, and transfected using the Nucleofector™® II Device (Lonza) as previously described in Example II or the Nucleofector™® 96-well Shuttle® Device (Lonza). The HFFs were harvested with TrypLE™ (Gibco), and 1×106 cells/transfection with the Nucleofector™® II Device for 10 min at 90 g and 6×105 cells/transfection with the Nucleofector™® 96-well Shuttle® Device for 5 min at 80×g. After centrifugation, the cell pellet was gently resuspended in either 100 μl of Basic Nucleofector™ Solution for the Nucleofector™® II or 20 μl of SE Solution (Cell line kit SE, Lonza) for the Nucleofector™® 96-well Shuttle®. For the Nucleofector™® II Device, each 100 μl of cell suspension was combined with 2 different mixes of plasmid DNA (sample 1 was mixed with 2 μg of pCMV6-XL5-Msi1 and 2 μg pCMV6-XL5-MBD2, and sample 2 with 2 μg of Msi1/Ngn2 and 2 μg pCMV6-XL5-MBD2). Each cell suspension was transferred into an Amaxa certified cuvette and transfected with the appropriate program (U-023). Right after transfection, 900 μl of warm CDM1 medium was added to each cuvette and the sample was transferred into a culture plate coated with Laminin (Stemgent, 10 μg/ml) at a cell density of 1×105 to 1.5×105 cells per cm2 or into non-cell culture treated Petri dishes for neurosphere formation. The cells were incubated at 37° C., 5% CO2 overnight. However for the Nucleofector™® 96-well Shuttle® Device, the steps described before were similar with the following exceptions: the cell suspension was mixed with 0.6 μg of each DNA of the same 2 DNA mixes, the cell suspension was transferred to a well of a 96-well Nucleoplate™ (Lonza) and transfected with the program FF-130™. After transfection, 80 μl of warm CDM1 medium was added to each well and the samples were left for 10 min in the incubator prior to being transferred into a laminin coated plate or non-cell culture treated Petri dishes at the same cell density as previously mentioned. For both devices, these steps were repeated for each sample that was transfected. Prior to transfection cells were cultured in CDM1 as described in Example I. After 24 hours, the medium was switched to a mix of 75% CDM medium and 25% Proliferation Medium which was supplemented with EGF (20 ng/ml), FGF-2 (20 ng/ml), Noggin (20 ng/ml) and Cytochalasin B (10 μg/ml) and the cells were incubated at 37° C., 5% CO2 and 5% O2. The medium was changed daily with an increased proportion of Neural proliferation medium up to 100% by Day 4 and a decreased proportion of Cytochalasin B that was completely omitted by Day 5. Forskolin (10 PM) was added to the medium from Day 4 onwards. The cells in floating conditions were pelleted by centrifugation and their medium changed daily as described for the adherent condition. Cells were collected at Day 3, 7, and 12 for imunohistochemistry analysis.


Fluorescence images were taken with a Cellomics™ ArrayScan HCS Reader™ microscopy system to determine an estimate of the percentage of cells positive for Sox2, a neural stem cell marker. This analysis revealed that in untransfected controls and at 3 days after transfection, no nuclear Sox2 staining was detectable. However, at Day 7 and Day 12 the percentage of Sox2 positive cells increased progressively under all transfection conditions except the pCMV6-XL5-Musashi and pCMV6-XL5-MBD2 Nucleofector™® II condition. The highest percentage at Day 12 was obtained with Msi1/Ngn2 and pCMV6-XL5-MBD2 transfected with the Nucleofector™® 96-well Shuttle® Device (80%). The same combination transfected with the Nucleofector™® II yielded only ˜35% positive cells. The pCMV6-XL5-Musashi and pCMV6-XL5-MBD2 with the Shuttle® produced ˜20% positive cells, while generally none were observed with the Nucleofector™® II. The percentage of positive cells varied strongly between wells. The staining indicated that the cell population was not homogenous, since fields of densely arranged Sox2 positive cells and complete fields with only negative cells could be found in all cases. In general the Shuttle® was initially more toxic to cells than the Nucleofector™® II, however at least in the case of Msi1/Ngn2 and pCMV6-XL5-MBD2 shuttle, the Sox2 positive population rapidly expanded from Day 7 to Day 12 to have twice as many Sox2 positive cells as compared to the Nucleofector™® II. The cells in floating conditions did not form spheres during the 12 day experiment in any of the conditions, suggesting that the formation of neurospheres requires either the generation of neural stem-like cells in adherent conditions first or more time.


Table 11 shows the percentage of Sox2 positive cells with a typical neural stem cell morphology using both the Nucleofector™® II Device and the Nucleofector™® 96-well Shuttle® Device. The latter had the advantages of requiring a smaller starting material (less cells and less DNA required) and in addition gave rise to a higher number of Sox2 positive cells. Moreover a very small population of Sox2 positive cells was observed with the Shuttle® Device only upon transfection with only one neurogenic transcription factor (Msi) in the presence of the DNA demethylator MBD2.









TABLE 11







Percentage of positive cells for Sox2 after transfection of fibroblast cells with different expression


vectors. After transfection the cells were cultured in proliferation medium (StemCell Technologies)


supplemented by EGF (20 ng/ml, Peprotech) and FGF (20 ng/ml, Peprotech) for two weeks at


37° C./5% CO2/5% O2. The percentage of immunopositive cells was determined by Cellomics ™


and represented as mean ± SD (n = 3-5).


% Sox2 positive cells













Day 3
Day 7
Day 12

















Total

Total

Total





Cell

Cell

Cell




Sox2
count
Sox2
count
Sox2
count





Shuttle
MSI1/NGN2 + MBD2
1.34 ±
6430 ±
31 ± 20 ±
10683 ±
78.17 ±
29341 ±




0.10
566
8.03
1112
3.10
2527



Msi + MBD2
1.08 ±
8253 ±
3.19 ±
8953 ±
19.05 ±
11082 ±




0.61
399
3.57
672
17.88
2999


Nucleofector ™
MSI1/NGN2 + MBD2
0.87 ±
21870 ±
14.30 ±
37321 ±
35.93 ±
33009 ±




0.30
4476
1.83
6877
7.10
1567



Msi + MBD2
0.64 ±
46793 ±
0.35 ±
34854 ±
0.51 ±
32095 ±




0.07
8808
0.16
2186
0.25
3236









Example V

Neurosphere Formation Assay and Cell Differentiation Analysis


Based on previous studies showing that greater proportional reprogramming is achieved by transfecting two neurogenic genes, this study was designed to evaluate the number of reprogramming cells by using the vector Msi1/Ngn2, containing two neurogenic transcription factors (Msi1 and Ngn2) and the role of DNA demethylator or DNA methylation inhibitor (5-azacytidine) and histone deacetylation inhibitor (VPA) in the reprogramming process.


HFFs were cultured and treated with cytochalasin B as described in Example III, and treated simultaneously with VPA (1 mM) and 5-Azacytidine (0.5 μM). After two days of treatment, cells were transfected by Nucleofection as described in Example II with the constructed vector Msi1/Ngn2. After preparing the cells, they were mixed with 2 μg of total DNA (Msi1/Ngn2) and cells that had not been treated with chemical inhibitors (VPA and 5-Aza) were co-transfect with MBD2 (2 μg), using the appropriate program (U023). The samples were transferred into a coated culture plate with Laminin (10 μg/ml, Sigma) and incubated in a humidified 37° C./5% O2/5% CO2 incubator. The medium was changed to the proliferation basal media, Neural Proliferation Medium (NeuroCult™ proliferation Kit, StemCell Technologies), with the presence of Noggin (20 ng/ml, Peprotech), recombinant hFGF (20 ng/ml, Peprotech), and recombinant hEGF (20 ng/ml, Peprotech). Following 6 days of transfection, cells were harvested using Accutase™ (Millipore), centrifuged (300×g, 5 min, RT) and plated in uncoated cell culture dishes in NeuroCult™ NSC Proliferation medium to investigate the capacity to grow cells in suspension as neurospheres or on Laminin coated-plates for adherent culture. To prevent loss of floating spheres during media changes, cells were sedimented by centrifugation at 150×g for 3 min at room temperature (RT). The pellet was then resuspended in fresh medium and plated into new uncoated, low-bind cell culture dishes. Cultures were incubated at 37° C., 5% CO2, 5% O2 and were fed daily for at least two months.


To investigate whether a single cell from human neural precursor cells (hNPCs) and human NSLCs was able to generate a neurosphere (a standard test for proving that a cell is a neural stem cell), neurospheres were dissociated into single cells and these single cells were isolated and cultured in proliferation medium in suspension, and neurosphere formation was monitored by taking bright field images using light microscope (Nikon, 10×) and by Cellomics™. These cells started to proliferate and grew as spheres starting day 6 to day 10 (FIG. 4A). Immunohistochemistry analysis of these spheres (Table 12 and FIG. 4) on Day 20, revealed immunopositive staining for the neural stem cells markers Sox2, Musashi, CD133, Nestin, and GFAP. Cells also stained positive for 1111-tubulin (a marker for neurons), 04 (a marker for oligodendrocytes), and GFAP (a marker for astrocytes), indicating the tri-potent differentiation potential of both sets of cells (NSLC and hNPC), and negative for NGFrec and NeuN (markers for differentiated neurons) indicating that the cells were not terminally differentiated.









TABLE 12







Percentage of positive cells for neural stem cells, and neuronal,


astrocyte and oligodendrocyte lineage markers in neurospheres


formed from single NSLCs and hNPCs cultured in


proliferation medium (StemCell Technologies) supplemented


by EGF (20 ng/ml, Peprotech) and FGF (20 ng/ml, Peprotech)


for 20 days at 37° C./ 5% CO2 / 5% O2. The percentage


of positive cells was determined by


Cellomics ™ and represented as mean ± SD.











% of positive cells
NSLCs
hNPCs







Musashi
91.8 ± 6.8
88.6 ± 7.9



Nestin
78.6 ± 5.7
 75.4 ± 12.0



GFAP
69.2 ± 7.4
78.6 ± 8.4



βIII-tubulin
85.6 ± 6.4
76.6 ± 8.4



P75
0
0



NeuN
0
0



O4
65.4 ± 6.6
71.4 ± 7.5



CD133
0
0










HFF cells were cultured as described in Example I, and transfected using the Nucleofector™ II device (Lonza) as described in Example II. Cells were co-transfected with pCMV6-XL5-Msi/pCMV6-XL4-Ngn2, pCMV-Msi1-Ngn2 with MBD2 or pre-treated with VPA/5aza. Cells were cultured in proliferation medium as suspension or adherent cultures. Gene expression analysis on 8 samples was performed as previously described in Example I with the customized Neuronal Markers 2 TLDA (Table 13) which profiled the expression of 48 genes (including three housekeeping genes: ACTIN, GAPDH and PPIA) in four major categories; 1) fibroblast specific genes; 2) neuronal lineage specific genes; 3) Neural stem cell marker specific genes; and 4) Genes for growth factors and their receptors.









TABLE 13





Neuronal Markers 2 TLDA Layout (Applied Biosystems)


Gene Symbols




























1
2
3
4
5
6
7
8
9
10
11
12





 1
ACTB
PPIA
COL3A1
LOX
S100A4
SYT1
SNAP25
NEUROD1
MBP
NKX2-2
GAPDH
OLIG2


 2
VIM
SOX3
SOX9
PROM1
SOX1
SOX2
KLF4
POUSF1
STAT3
PIK3CG
GDNF
NGF


 3
ACTB
PPIA
COL3A1
LOX
S100A4
SYT1
SNAP25
NEUROD1
MBP
NKX2-2
GAPDH
OLIG2


 4
VIM
SOX3
SOX9
PROM1
SOX1
SOX2
KLF4
POUSF1
STAT3
PIK3CG
GDNF
NGF


 5
ACTB
PPIA
COL3A1
LOX
S100A4
SYT1
SNAP25
NEUROD1
MBP
NKX2-2
GAPDH
OLIG2


 6
VIM
SOX3
SOX9
PROM1
SOX1
SOX2
KLF4
POUSF1
STAT3
PIK3CG
GDNF
NGF


 7
ACTB
PPIA
COL3A1
LOX
S100A4
SYT1
SNAP25
NEUROD1
MBP
NKX2-2
GAPDH
OLIG2


 8
VIM
SOX3
SOX9
PROM1
SOX1
SOX2
KLF4
POUSF1
STAT3
PIK3CG
GDNF
NGF


 9
ACTB
PPIA
COL3A1
LOX
S100A4
SYT1
SNAP25
NEUROD1
MBP
NKX2-2
GAPDH
OLIG2


10
VIM
SOX3
SOX9
PROM1
SOX1
SOX2
KLF4
POUSF1
STAT3
PIK3CG
GDNF
NGF


11
ACTB
PPIA
COL3A1
LOX
S100A4
SYT1
SNAP25
NEUROD1
MBP
NKX2-2
GAPDH
OLIG2


12
VIM
SOX3
SOX9
PROM1
SOX1
SOX2
KLF4
POUSF1
STAT3
PIK3CG
GDNF
NGF


13
ACTB
PPIA
COL3A1
LOX
S100A4
SYT1
SNAP25
NEUROD1
MBP
NKX2-2
GAPDH
OLIG2


14
VIM
SOX3
SOX9
PROM1
SOX1
SOX2
KLF4
POUSF1
STAT3
PIK3CG
GDNF
NGF


15
ACTB
PPIA
COL3A1
LOX
S100A4
SYT1
SNAP25
NEUROD1
MBP
NKX2-2
GAPDH
OLIG2


16
VIM
SOX3
SOX9
PROM1
SOX1
SOX2
KLF4
POUSF1
STAT3
PIK3CG
GDNF
NGF






13
14
15
16
17
18
19
20
21
22
23
24





 1
ALDHIL1
DIO2
GFAP
NCAM1
FOXJ1
PDGFRA
MK167
NES
CSPG4
DLX2
MSI1
CROCC


 2
BDNF
CNT-
GAP43
NRG1
NPY
CSF3
BMP4
TGFB1
VEGFA
NGFR
EGFR
KDR




ZFP91-














CNTF












 3
ALDHIL1
DIO2
GFAP
NCAM1
FOXJ1
PDGFRA
MK167
NES
CSPG4
DLX2
MSI1
CROCC


 4
BDNF
CNT-
GAP43
NRG1
NPY
CSF3
BMP4
TGFB1
VEGFA
NGFR
EGFR
KDR




ZFP91-














CNTF












 5
ALDHIL1
DIO2
GFAP
NCAM1
FOXJ1
PDGFRA
MK167
NES
CSPG4
DLX2
MSI1
CROCC


 6
BDNF
CNT-
GAP43
NRG1
NPY
CSF3
BMP4
TGFB1
VEGFA
NGFR
EGFR
KDR




ZFP91-














CNTF












 7
ALDHIL1
DIO2
GFAP
NCAM1
FOXJ1
PDGFRA
MK167
NES
CSPG4
DLX2
MSI1
CROCC


 8
BDNF
CNT-
GAP43
NRG1
NPY
CSF3
BMP4
TGFB1
VEGFA
NGFR
EGFR
KDR




ZFP91-














CNTF












 9
ALDHIL1
DIO2
GFAP
NCAM1
FOXJ1
PDGFRA
MK167
NES
CSPG4
DLX2
MSI1
CROCC


10
BDNF
CNT-
GAP43
NRG1
NPY
CSF3
BMP4
TGFB1
VEGFA
NGFR
EGFR
KDR




ZFP91-














CNTF












11
ALDHIL1
DIO2
GFAP
NCAM1
FOXJ1
PDGFRA
MK167
NES
CSPG4
DLX2
MSI1
CROCC


12
BDNF
CNT-
GAP43
NRG1
NPY
CSF3
BMP4
TGFB1
VEGFA
NGFR
EGFR
KDR




ZFP91-














CNTF












13
ALDHIL1
DIO2
GFAP
NCAM1
FOXJ1
PDGFRA
MK167
NES
CSPG4
DLX2
MSI1
CROCC


14
BDNF
CNT-
GAP43
NRG1
NPY
CSF3
BMP4
TGFB1
VEGFA
NGFR
EGFR
KDR




ZFP91-














CNTF












15
ALDHIL1
DIO2
GFAP
NCAM1
FOXJ1
PDGFRA
MK167
NES
CSPG4
DLX2
MSI1
CROCC


16
BDNF
CNT-
GAP43
NRG1
NPY
CSF3
BMP4
TGFB1
VEGFA
NGFR
EGFR
KDR




ZFP91-














CNTF










Sample Information

















Sample

TLDA



ID
Sample Name
Port









1
HFF Ctrl
1



2
ReNcell Undifferentiated Ctrl
2



3
Msi1-Ngn2/MBD2
3



4
Msi1-Ngn2/MBD2
4



5
Msi1-Ngn2/VPA + AZA
5



6
Msi1-Ngn2
6



7
Msi1-Ngn2/MBD2, neurospheres
7



8
Msi1-Ngn2/MBD2, neurospheres
8










As shown in Table 14, fibroblast-specific genes (Col3A1, Lox, S100A4) were down-regulated in reprogrammed cells, indicating the loss of fibroblast-specific genes following transfection (note that not all cells got transfected and reprogrammed, so the presence of fibroblast-specific gene expression in the cultures is mostly from the un-programmed fibroblasts left in the culture). The expression of these genes is observed to increase when HFFs were transfected in the absence of DNA demethylator or the DNA methylation inhibitor, indicating that down-regulation of differentiated markers of fibroblast cells requires DNA demethylation. The expression of ectoderm genes such as Msi1, Sox2, and Nestin was remarkably increased following transfection in conjunction with DNA demethylation. The expression of neuronal markers, such as synaptogamini (a synaptic vesicle protein) and NeuroD1 was up-regulated in transfected cells with Msi1/Ngn2/MBD2, and slightly increased in transfected cells with Msi1/Ngn2/VPA and 5-AZA. The selected three markers of oligodendrocytes were detected in the transfected cells with a strong increase of Olig2. Two markers for astrocytes, GFAP and ALDH1L1, were enhanced following transfection. The results support the idea that neurospheres are composed of heterogeneous progenitor subtypes.


Among the neurotrophic factors, expression of CNTF was slightly increased in the reprogrammed cells. The expression of GAP-43 and neuropeptide Y (NPY) were the most annotated genes. GAP-43 has long been acknowledged to play a pivotal role in axonal plasticity and is used as a marker of regenerating neurite outgrowth and synaptogenesis, both in embryonic development and in neuronal regeneration in injured brain and spinal cord. Expression of receptors for growth and neurotrophic factors was increased, such as neurotrophic receptor tyrosine kinase expression.









TABLE 14







Gene array analysis was performed after one month of transfection of human fibroblast cells with Msi1/Ngn2, in the presence MBD2 or


VPA and 5-Aza. Cells were cultured on coated culture plates as adherent cells or on untreated culture plates as neurospheres in proliferation


medium (StemCell Technologies) supplemented with EGF (20 ng/ml) and FGF (20 ng/ml). Untransfected cells were considered as negative


control and ReNcell (Millipore) as positive control.











Relative expression to #1 HFF Ctrl


















#3
#4


#7
#8




#2
Msi1-
Msi1-
#5
#6
Msi1-Ngn2/
Msi1-Ngn2




ReNcell
Ngn2/
Ngn2/
Msi1-Ngn2/
Msi1-
MBD2,
NPA + AZA,


Symbol
Common name and description
Undiff
MBD2
MBD2
VPA + AZA
Ngn2
neurospheres
neurospheres


















Fibroblast/ECM










component










COL3A1
Collagen, type III, alpha 1, fibroblast
0.00
0.03
0.02
0.02
11.92
0.00
0.00



marker









LOX
Lysyl oxidase, ECM component
0.01
0.03
0.01
0.01
2.38
0.00
0.00


FSP1
Fibroblast transcription site-1,
0.04
0.04
0.06
0.05
3.22
0.05
0.05



enzyme for ECM remodeling









Neuron markers










SYT1
Synaptotagmin1, a synaptic vesicle
106.49
108.40
78.66
26.72
22.42
37.61
16.80



protein in neurons









SNAP25
SNAP25, mature neuron marker
4.72
6.10
7.89
3.11
3.19
6.47
4.00


NEUROD1*
Neurogenic differentiation 1, neuron
2.32
93.35
100.84
2.02
3.11
271.11
10.23



marker









Oligodendrocyte










markers










MBP*
Myelin Basic Protein, mature
2.32
48.53
18.11
6.94
667.56
16.67
1.67



oligodendrocyte marker









NKX2-2*
NK2 homeobox 2, remyelination
2.32
75.31
54.65
1.66
3.11
1.67
1.74


OLIG2*
Oligodendrocyte lineage transcription
2856.4
15594
67369
38733
3.11
92420
101733



factor 2, oligodendrocyte progenitor









Astrocyte markers










ALDH1L1*
Aldehyded ehydrogenase 1 family
6.20
3.77
4.65
1.66
0.02
5.87
9.59



member L1, astrocyte









DIO2*
Deiodinase iodothyronine type II,
23.20
0.00
0.00
0.00
0.51
0.00
0.00



astrocyte marker









GFAP
Glial fibrillary acidic protein,
3342.1
6899.0
6291.0
4800.9
1.27
3118.7
3222.0



astrocyte marker









NSCS markers










NCAM1
NCAM1, neuroblast marker
23.21
43.90
24.45
12.72
1.13
31.93
36.70


PDGFRA
Plate-derived growth factor receptor
0.05
0.01
0.01
0.00
4.42
0.00
0.01



alpha, oligodendrocyte progenitor










cells









NES
Nestin, neural progenitor
5.76
19.84
19.56
3.46
4.23
16.57
8.36


MSI1*, **
Musashi I, neuroblast marker
5120.3
5985.2
5262.7
5645.1
204.34
3179.6
4113.6


SOX1*
Sox1, neural progenitor
679.21
223.59
373.14
361.67
3.11
287.82
323.23


SOX2*
Sox2, NSCs
1924084
2265299
1889166
1014816
3.11
1313765
1103212


Neurotrophic/










Growth Factor










GDNF*
Glial cell derived neurotrophic factor
0.01
0.02
0.02
0.00
1.69
0.00
0.00


NGF*
Nerve growth factor
0.00
0.00
0.00
0.00
1.48
0.00
0.00


BDNF
Brain derived neurotrophic factor
0.03
0.09
0.09
0.05
0.82
0.02
0.01


CNTF*
Ciliary neurotrophic factor
9.25
4.32
3.11
2.90
64.05
2.31
3.39


GAP43
Growth associated protein 43, neural
917.52
3506.5
1530.8
452.75
584.00
746.25
578.52



regeneration









NRG1*
Neuregulin 1, neural regeneration
0.01
0.00
0.00
0.00
0.40
0.00
0.00


NPY*
Neuropeptide Y, interneuron
2.32
675.69
465.04
153.54
3.11
1244.0
130.38


CSF3*
Colony stimulating factor 3, neural
0.50
0.03
0.02
0.58
18.62
0.02
0.02



regeneration









BMP4
Bone morphogenetic protein 4,
0.83
0.26
0.74
0.45
11.03
0.09
0.07



remyelination marker









TGFB1
Transforming growth factor, beta 1
0.85
2.39
0.92
0.83
0.65
0.45
0.58


Angiogenesis










VEGFA
Vascular endothelial growth factor
2.77
14.93
15.01
2.67
3.82
2.80
3.21


Neurotrophin/










Growth Factor










Receptors










NGFR/P75
NGFR, neurotrophin receptor
5.35
3.29
5.78
9.10
7.53
7.26
17.51


EGFR
Epidermal growth factor receptor
0.89
0.77
0.86
0.79
1.63
1.44
1.25


KDR*
Kinase insert domain receptor,
210.87
259.42
263.45
51.85
0.07
11.23
17.50



growth factor receptor
















Further analysis and quantification of the adherent population of NSLCs showed that cells were positively stained for Sox2 (93.43±1.9%), nestin (60.76±5.7%), and GABA (37.48±4.9), while these markers were undetectable in untransfected cells (FIG. 5, Table 15). Furthermore, these cells stained positive for p75NTR (31.15±1.6), βIII-tubulin (37.55±0.6%) and GFAP (16.47±0.9). However, untransfected HFFs only stained positive for HFF markers (FIG. 5), such as fibronectin and fibroblast protein marker, while these markers were undetectable in reprogrammed cells, demonstrating that the reprogrammed cells lost markers of the original cells and adopted morphology and markers of neural stem cells and a neuronal lineage.









TABLE 15







The percentage of cells stained positive for neural stem cell markers and fibroblast


markers in untransfected cells and transfected cells with pMsi1/Ngn2/MBD2. Transfected


cells (NSLCs) possess a high percentage of neural stem markers but a very low percentage


of fibroblast markers as compared to untransfected cells. The percentage of immunopositive


cells was determined by Cellomics ™ and represented as mean ± SD (n = 5).










Transfected fibroblast cells
Untransfected fibroblast cells


Marker
(% of average
(% of average


protein
positive cells ± stdv)
positive cells ± stdv)





Sox2
93.43 ± 1.9
1.90 ± 0.5


Nestin
60.76 ± 5.7
0.84 ± 0.2


p75NTR
31.15 ± 1.6
3.95 ± 1.7


NCAM
26.84 ± 3.8
0.87 ± 0.2


S100
41.80 ± 0.6
1.60 ± 0.3


GFAP
16.47 ± 0.9
3.84 ± 0.9


βIII-Tubulin
37.55 ± 0.6
1.90 ± 0.9


GABA
37.48 ± 4.9
2.54 ± 0.5


Fibronectin
 1.05 ± 0.7
94.19 ± 0.9 


Fibroblast
 4.81 ± 1.0
50.30 ± 7.8 


marker protein









This study showed as well that NSLCs have the capacity to proliferate in culture and exhibit stable morphology, gene and protein expression that were maintained for the entire study period, which was for over five month in culture (Table 16).









TABLE 16







Doubling time of NSLCs over serial passages. NSLCs were


maintained in proliferation conditions for 35 passages in a


37° C., 5% CO2 and 5% O2 incubator. The time required for


the cell population to double (g) was calculated for each


passage, and was defined as g = (In2)/k, where k was


the number of generations that occured per unit time (t)


defined as, k = (In Nf − In N0)/t, where Nf was the final


cell number and N0 the initial seeded cell number. The


average generation time was 25.4 h over 35 passages.












Passage
Time
LN
LN
k
g


number
(h)
N0
Nf
(h−1)
(h)















2
168
11.513
15.577
0.024
38.655


3
216
11.513
16.195
0.022
31.977


4
192
11.513
18.258
0.035
39.730


5
144
11.513
16.258
0.033
21.036


6
144
11.513
16.258
0.033
21.036


7
144
11.513
15.702
0.029
33.824


8
168
11.513
15.870
0.026
26.729


9
120
11.513
16.811
0.031
32.548


10
144
11.513
15.415
0.027
35.580


11
120
13.122
15.895
0.023
30


12
120
11.513
15.747
0.035
19.645


13
168
11.513
15.870
0.026
26.729


14
168
12.429
15.870
0.020
23.847


15
168
11.513
15.520
0.024
29.059


16
192
11.513
16.167
0.024
28.596


17
144
11.513
15.239
0.026
36.791


18
168
11.513
15.790
0.025
37.229


19
120
13.122
15.870
0.023
30.276


20
144
13.122
16.249
0.022
31.922


21
96
13.122
15.761
0.027
25.214


22
120
13.122
15.870
0.023
30.276


23
120
13.122
15.761
0.022
31.518


24
96
13.122
15.687
0.027
25.943


25
96
13.122
16.013
0.030
23.022


26
96
13.122
16.067
0.031
22.599


27
96
13.122
16.300
0.033
20.938


28
120
13.122
16.482
0.028
24.752


29
96
13.122
16.380
0.034
20.424


30
96
13.122
16.300
0.033
19.938


31
120
13.122
16.483
0.028
22.752


32
96
13.122
16.062
0.031
20.640


33
96
13.122
16.300
0.033
20.938


34
96
13.122
16.077
0.031
15.519


35
96
13.122
16.077
0.031
15.519










Gene Expression Microarray


Microarray expression analysis was performed to get a global overview to compare the gene expression profile of passage 7 NSLC to both HFF (the cells that the NSLC were created from) and hNPCs. NSLC (n=3), HFF (n=2), and hNPC (n=3) were resuspended in RNAlater™ (Qiagen) and shipped to Genotypics (India) where the samples were processed and the Gene Expression Microarray was performed.


In brief, Genotypics extracted RNA from the samples and performed Quality Control using an Agilent Bioanalyzer™. Labelling was done using Agilent's Quick Amp™ kit (cDNA synthesis and in vitro transcription), followed by Labelling QC. Hybridization was then performed using the 8×60K array, and scanning was done using high throughput Agilent scanner with SureScan™ technology. The Agilent Feature Extraction software was used for automated feature extraction, followed by Raw Data QC and Image QC. Advanced Data Analysis was then performed, including Pathway and Gene Ontology analysis using Agilent's GeneSpring GX™ v10.0 and Genotypic's Biointerpreter Software. The NSLC samples were compared to the HFF samples (Set 1) and hNPC samples (Set 2) The NSLC samples had a global gene expression pattern that was much closer to the hNPCs than the HFFs from which the NSLCs were created (FIG. 23). Pearson correlation analysis revealed that NSLCs are closely related to hNPCs, including in terms of neuronal lineage markers, regenerative genes and migration genes. These data confirm that NSLCs are similar, but not identical, to hNPCs.


Microarray analysis revealed an up-regulation of neural precursor genes in the NSLC samples as compared to the HFF samples. ACTL6A and PHF10, which both belong to the neural progenitors-specific chromatin remodelling complex (npbaf complex) and are required for the proliferation of neural progenitors, were up-regulated by 2.9-fold and 2.3 fold respectively. MSI2, which plays a role in the proliferation and maintenance of stem cells in the central nervous system, was up-regulated by 6-fold (Table X1). Glia genes were up-regulated in the NSLC samples as compared to the HFF samples. GFAP, is a neural stem cell- and astrocyte-specific marker that, during the development of the central nervous system, distinguishes astrocytes from other glial cells, is highly up-regulated in the NSLC sample as compared to HFF (690-fold). OLIG1, which promotes formation and maturation of oligodendrocytes, especially within the brain, is also highly up-regulated in NSLC sample as compared to HFF (370-fold) (Table X2).


Table X3 lists a subset of regenerative genes that are up-regulated in the NSLC samples as compared to the HFF samples. SOX2, a gene critical for early embryogenesis and for embryonic stem cell pluripotency as well as neural stem cells, is highly up-regulated in the NSLC samples as compared to the HFF samples (5000-fold). CCND2, which is essential for the control of the cell cycle at the G1/S (start) transition, is also up-regulated in NSLC samples (70-fold as compared to HFF samples). As shown in Table X4, numerous fibroblast genes were down-regulated in the NSLC samples as compared to the HFF samples. This shows that the NSLC lose the expression of numerous fibroblast genes as it gets reprogrammed from HFF to NSLC.


Table X5 show that neural precursor genes were also up-regulated in the NSLC samples as compared to the hNPC samples. BDNF, which promotes the survival and differentiation of selected neuronal populations of the peripheral and central nervous systems during development, is even more highly expressed in NSLC samples than in hNPC samples (34-fold up-regulation). Table X6 shows that a subset of Glia genes are also up-regulated in the NSLC samples as compared to the hNPC samples. GFAP, a neural stem cell- and astrocyte-specific marker that, during the development of the central nervous system, distinguishes astrocytes from other glial cells, is more highly expressed in NSLC samples than hNPC samples (13-fold). PLP1, the major myelin protein of the central nervous system which plays an important role in the formation or maintenance of the multilamellar structure of myelin, is also more highly expressed in NSLC samples than in hNPC samples (20-fold).


Regenerative genes were also up-regulated in the NSLC samples as compared to the hNPC samples (Table X7). BMP2, a neural crest marker, but which induces growth especially of cartilage and bone formation and BMP4, which in turn induces cartilage and bone formation and acts in mesoderm induction, tooth development, limb formation and fracture repair, but also in neural stem cells, were both more highly expressed in NSLC samples than in hNPC samples (18-fold and 20-fold respectively). GAP43, which is a major component of the motile growth cones that form the tips of elongating axons was more highly expressed in NSLC samples than hNPC samples (4-fold). This suggests the regenerative potential of NSLC. HOXB4, a transcription factor that is involved in development and also in the expansion of neural stem cells as well as hematopoietic stem and progenitor cells in vivo and in vitro making it a potential candidate for therapeutic stem cell expansion, was also more highly expressed in NSLCs than in hNPCs. This data indicates that NSLCs are more ‘stem-like’ or have more ‘stemness’ than hNPCs.









TABLE X1







Up-regulated Neural Precursor genes (NSLC vs. HFF)











Accession
Fold change of NSLC



GeneSymbol
Number
compared to HFF1
p-value













ACTL6A
NM_178042
2.90
0.000


ADAM9
NM_001005845
2.64
0.004


AIFM1
NM_004208
2.45
0.000


BCAT1
NM_005504
3.23
0.000


BMP2
NM_001200
17.49
0.000


DLL1
NM_005618
40.32
0.000


EDNRB
NM_003991
933.03
0.000


ERBB4
NM_005235
53.22
0.006


GMNN
NM_015895
4.42
0.000


HES5
BC087840
102.33
0.000


KIF1B
NM_015074
9.45
0.002


LIMK1
NM_002314
2.44
0.002


MAPK8IP1
NM_005456
5.88
0.001


MCHR1
NM_005297
68.19
0.001


MEF2C
NM_002397
2.91
0.000


MSI2
NM_170721
6.76
0.000


NMB
NM_021077
3.65
0.000


NOS2A
NM_000625
279.45
0.000


NOTCH1
NM_017617
6.75
0.000


NPAS3
NM_022123
187.85
0.000


PHF10
NM_018288
2.28
0.001


PHLPP
NM_194449
8.84
0.000


SMAD1
NM_005900
4.74
0.000


SNTG1
AL161971
34.05
0.000


SP8
NM_198956
1392.67
0.000


STAU2
AK002152
3.35
0.000


STIL
NM_003035
4.94
0.003






1Fold change represents the up-regulation of the gene in the NSLC samples as compared to the HFF samples. (n = 2 for HFF samples, n = 3 for NSLC samples).














TABLE X2







Up-regulated Glia genes (NSLC vs. HFF)












Fold change of





NSLC compared



GeneSymbol
Accession Number
to HFF1
p-value













ASTN1
NM_004319
51.44
0.000


ATP1B2
NM_001678
186.64
0.000


B3GAT1
NM_018644
1784.49
0.000


BCL2
NM_000633
2.65
0.002


BMP7
NM_001719
41.35
0.000


CA14
NM_012113
43.44
0.000


CLCN2
NM_004366
4.18
0.000


CNDP1
NM_032649
4.39
0.010


CP
NM_000096
93.08
0.002


CXCR4
NM_001008540
4124.29
0.000


ERBB4
NM_005235
53.22
0.006


FABP7
NM_001446
18702.36
0.000


GAB1
NM_207123
2.44
0.001


GFAP
NM_002055
696.51
0.000


GJB2
NM_004004
13.89
0.001


ITGB8
NM_002214
8.48
0.005


KCNJ10
NM_002241
263.42
0.000


LMO3
NM_018640
194.32
0.000


MAP6D1
NM_024871
3.99
0.000


MAPT
NM_016835
2.38
0.001


NDE1
NM_017668
2.21
0.002


NEFL
NM_006158
10.30
0.001


NKX6-2
NM_177400
10.83
0.026


NOVA2
NM_002516
7.51
0.000


NTN1
NM_004822
5.29
0.015


NTRK3
NM_001012338
15.32
0.000


OLIG1
NM_138983
372.11
0.000


OLIG2
NM_005806
163.20
0.000


PARD6A
NM_016948
4.12
0.001


PASK
NM_015148
3.89
0.001


PAX6
NM_001604
28.53
0.001


PDCD11
ENST00000369797
2.23
0.001


PDE6B
NM_000283
5.55
0.001


PER1
NM_002616
2.43
0.001


PLP1
M54927
351.09
0.000


PTK2
NM_153831
4.22
0.000


QKI
NM_206855
8.75
0.003


S100B
NM_006272
456.00
0.000


SLC1A3
NM_004172
49.49
0.000


SORL1
NM_003105
27.61
0.000


SOX9
NM_000346
27.82
0.000


SPRY2
NM_005842
15.83
0.000


TARDBP
NM_007375
2.69
0.005


TSPAN12
NM_012338
259.78
0.000
















TABLE X3







Up-regulated Regenerative genes (NSLC vs. HFF)











Accession
Fold change of NSLC



GeneSymbol
Number
compared to HFF1
p-value













BMP2
NM_001200
17.49
0.000


CCND2
NM_001759
72.79
0.000


DLL1
NM_005618
40.32
0.000


EGR1
NM_001964
2.19
0.000


GAL
NM_015973
25.93
0.000


GAP43
NM_002045
1297.42
0.000


HOXB4
NM_024015
102.34
0.000


NFE2L2
AF323119
2.80
0.004


NOTCH1
NM_017617
6.75
0.000


PRPH
NM_006262
6.44
0.000


SEMA3A
NM_006080
3.03
0.004


SEMA6A
NM_020796
23.58
0.000


SOX2
NM_003106
5165.92
0.000
















TABLE X4







Down-regulated Fibroblast genes (NSLC vs. HFF)











Accession
Fold change of NSLC



GeneSymbol
Number
compared to HFF1
p-value













ACOT2
NM_006821
0.30
0.000


AEBP1
NM_001129
0.16
0.001


AGA
NM_000027
0.35
0.000


ANXA2
NM_001002857
0.26
0.029


AP4E1
NM_007347
0.30
0.008


APOE
NM_000041
0.08
0.000


ARHGDIB
NM_001175
0.24
0.009


ASAH1
NM_004315
0.31
0.000


BDKRB1
NM_000710
0.00
0.001


BDKRB2
NM_000623
0.00
0.000


BDNF
NM_170735
0.12
0.000


BMP4
NM_001202
0.28
0.001


C3
NM_000064
0.25
0.001


C5orf13
NM_004772
0.18
0.000


CACNA1C
NM_000719
0.03
0.000


CASP4
NM_033306
0.00
0.000


CASP5
NM_004347
0.00
0.001


CCL2
NM_002982
0.20
0.000


CD36
NM_001001547
0.07
0.023


CDC42EP2
NM_006779
0.06
0.000


CDC42EP3
NM_006449
0.41
0.000


CDC42EP5
NM_145057
0.41
0.040


CDH11
NM_001797
0.00
0.000


CEMP1
AL833099
0.30
0.001


CFH
NM_001014975
0.01
0.010


CITED2
NM_006079
0.14
0.000


COL12A1
NM_004370
0.00
0.001


COL1A1
NM_000088
0.01
0.000


COL1A2
NM_000089
0.00
0.001


COL3A1
NM_000090
0.00
0.001


COL5A1
NM_000093
0.00
0.000


CPT1A
NM_001876
0.16
0.002


CROT
NM_021151
0.27
0.002


CTSA
NM_000308
0.10
0.000


CTSB
NM_147780
0.11
0.001


CXCL1
NM_001511
0.01
0.003


CXCL12
NM_000609
0.00
0.001


CYP27A1
NM_000784
0.28
0.011


CYR61
NM_001554
0.10
0.000


DCHS1
NM_003737
0.29
0.000


DMPK
NM_004409
0.36
0.000


DPT
NM_001937
0.05
0.006


EFEMP1
NM_004105
0.00
0.000


ELN
NM_000501
0.13
0.001


EMX2
NM_004098
0.00
0.001


EPS8
NM_004447
0.18
0.000


ETS1
NM_005238
0.15
0.003


FAH
NM_000137
0.17
0.000


FAM14A
NM_032036
0.22
0.001


FAP
NM_004460
0.00
0.000


FBLN2
NM_001004019
0.18
0.000


FBN1
NM_000138
0.01
0.002


FGF1
NM_000800
0.20
0.004


FGF13
NM_004114
0.04
0.006


FGF2
NM_002006
0.06
0.000


FGF5
NM_004464
0.01
0.003


FGF7
NM_002009
0.04
0.001


FGF9
NM_002010
0.01
0.000


FGFR1
NM_023110
0.34
0.026


FHL2
NM_201555
0.11
0.000


FN1
NM_212482
0.00
0.001


FSTL1
NM_007085
0.09
0.000


GADD45B
NM_015675
0.09
0.001


GALNT6
NM_007210
0.13
0.001


GAS6
NM_000820
0.02
0.000


GBA
NM_001005749
0.22
0.002


GBAP
NR_002188
0.19
0.000


GCH1
NM_000161
0.22
0.001


GGTA1
NR_003191
0.28
0.013


GIT2
NM_057169
0.37
0.003


GJA1
NM_000165
0.46
0.001


GLIS1
NM_147193
0.02
0.000


GM2A
AK127910
0.25
0.010


GNS
NM_002076
0.29
0.000


GPC3
NM_004484
0.22
0.038


GREM1
NM_013372
0.00
0.011


GSTM1
NM_146421
0.27
0.001


HAAO
NM_012205
0.43
0.001


HERPUD1
NM_014685
0.19
0.000


HEXA
NM_000520
0.24
0.000


HEXB
NM_000521
0.36
0.000


HGF
NM_001010932
0.09
0.028


HGS
NM_004712
0.26
0.029


HIF1A
NM_181054
0.36
0.005


HLA-A
NM_002116
0.31
0.002


HLA-H
NR_001434
0.19
0.001


HOXB13
NM_006361
0.03
0.004


HR
NM_005144
0.18
0.002


HSPG2
NM_005529
0.19
0.004


IDUA
NM_000203
0.16
0.000


IGF1
NM_000618
0.10
0.004


IGFBP7
NM_001553
0.28
0.040


IKBKG
NM_003639
0.42
0.001


IRF1
NM_002198
0.28
0.002


ITGA1
NM_181501
0.00
0.001


ITGB3
NM_000212
0.05
0.000


KLF4
NM_004235
0.05
0.002


LEP
NM_000230
0.07
0.001


LEPRE1
NM_022356
0.24
0.000


LMNA
NM_005572
0.42
0.000


LOX
NM_002317
0.01
0.000


LOXL4
NM_032211
0.10
0.003


LRRC8C
NM_032270
0.15
0.013


MAGEL2
AJ243531
0.31
0.002


MAN2B1
NM_000528
0.45
0.006


MAP3K8
NM_005204
0.27
0.001


MEIS2
NM_170677
0.00
0.001


MKNK1
NM_003684
0.37
0.005


MMP1
NM_002421
0.00
0.000


MMP14
NM_004995
0.07
0.001


MMP2
NM_004530
0.04
0.000


MMP3
NM_002422
0.00
0.001


MOXD1
NM_015529
0.24
0.000


MRAS
NM_012219
0.15
0.001


MSX2
NM_002449
0.15
0.031


MTHFR
NM_005957
0.27
0.014


MYC
NM_002467
0.05
0.000


MYL6
NM_079423
0.33
0.001


MYL9
NM_181526
0.01
0.000


NAGLU
NM_000263
0.23
0.000


NBL1
NM_182744
0.11
0.000


NEK9
NM_033116
0.41
0.001


NF2
NM_181831
0.46
0.000


NPC1
NM_000271
0.34
0.000


OPTN
NM_001008211
0.04
0.000


P4HB
NM_000918
0.37
0.001


PALLD
NM_016081
0.29
0.001


PAPPA
NM_002581
0.05
0.000


PCDHGB4
NM_032098
0.28
0.001


PCK2
NM_004563
0.04
0.000


PCOLCE
NM_002593
0.00
0.000


PDGFRA
NM_006206
0.02
0.010


PEX14
BC017848
0.48
0.000


PFKL
NM_001002021
0.35
0.004


PPARG
NM_138711
0.01
0.000


PPFIBP2
NM_003621
0.08
0.000


PRR5
NM_015366
0.23
0.022


PSEN2
NM_012486
0.34
0.002


PTGS1
NM_000962
0.29
0.000


PXDN
AF200348
0.12
0.000


PYCARD
NM_013258
0.03
0.000


QSOX1
NM_002826
0.09
0.000


RASSF1
NM_170713
0.30
0.001


RBMS1
NM_002897
0.14
0.001


RECK
NM_021111
0.07
0.000


RET
NM_020975
0.35
0.015


RFPL1S
NR_002727
0.22
0.039


ROD1
NM_005156
0.37
0.001


RSU1
NM_012425
0.41
0.002


S100A4
NM_002961
0.03
0.000


SAMD9
NM_017654
0.07
0.007


SCARB2
NM_005506
0.42
0.001


SDC2
NM_002998
0.38
0.000


SDPR
NM_004657
0.03
0.005


SENP2
AF151697
0.44
0.006


SEPP1
NM_001085486
0.00
0.005


SFRP1
NM_003012
0.37
0.000


SHOC2
NM_007373
0.39
0.000


SIGIRR
NM_021805
0.47
0.000


SLC17A5
NM_012434
0.14
0.001


SLC22A5
NM_003060
0.21
0.001


SLC9A3R2
NM_004785
0.29
0.000


SMPD1
NM_000543
0.17
0.000


STAT1
NM_139266
0.19
0.000


STAT6
NM_003153
0.00
0.000


STS
NM_000351
0.10
0.007


STYK1
NM_018423
0.05
0.013


SUMF1
NM_182760
0.28
0.000


TAGLN
NM_001001522
0.01
0.000


TFAP2A
NM_003220
0.03
0.005


THBS2
NM_003247
0.02
0.000


THRA
NM_199334
0.31
0.000


THRB
NM_000461
0.10
0.014


TNXB
NM_019105
0.26
0.043


TPM2
NM_213674
0.12
0.000


TRIOBP
NM_007032
0.15
0.003


TRIP11
NM_004239
0.45
0.001


TSC22D3
NM_004089
0.14
0.000


TWIST1
NM_000474
0.01
0.003


VCAN
NM_004385
0.04
0.000


VOL
NM_014000
0.28
0.000


VLDLR
NM_003383
0.15
0.000


WISP1
NM_003882
0.05
0.013


WNT5A
NM_003392
0.01
0.000


YAP1
NM_006106
0.41
0.007


ZBTB7B
NM_015872
0.44
0.000
















TABLE X5







Up-regulated Neural Precursor genes (NSLC vs. hNPC)














Fold change of





Accession
NSLC compared




GeneSymbol
Number
to hNPC2
p-value
















ACTL6A
NM_178042
2.33
0.000



BCAT1
NM_005504
9.92
0.000



BDNF
NM_170735
33.90
0.000



BMP2
NM_001200
17.71
0.000



CDKN2A
NM_058197
5.57
0.000



COL18A1
NM_030582
7.22
0.001



DIAPH1
NM_005219
2.33
0.001



EDNRB
NM_003991
2.78
0.000



IDE
NM_004969
2.74
0.000



LIMK1
NM_002314
3.61
0.000



MAPK8IP1
NM_005456
2.77
0.000



MCHR1
NM_005297
4.02
0.000



MYLIP
NM_013262
4.22
0.000



NEDD4
NM_006154
2.23
0.000



NOS2A
NM_000625
267.58
0.000



PCSK9
NM_174936
9.65
0.000



PSEN2
NM_000447
2.07
0.000



SMAD1
NM_005900
3.09
0.000



TBX1
NM_080647
3.65
0.028



TGFB1
NM_000660
6.66
0.000








2Fold change represents the up-regulation of the gene in the NSLC samples as compared to the hNPC samples. (n = 3 for hNPC samples, n = 3 for NSLC samples).














TABLE X6







Up-regulated Glia genes (NSLC vs. hNPC)











Accession
Fold change of NSLC



GeneSymbol
Number
compared to hNPC1
p-value













ACSL4
NM_004458
2.10
0.000


BDNF
NM_170735
33.90
0.000


BMP4
NM_001202
20.55
0.001


CP
NM_000096
159.46
0.000


CSPG4
NM_001897
4.94
0.000


FOXC1
NM_001453
5.12
0.000


GFAP
NM_002055
13.67
0.000


GJB2
NM_004004
7.25
0.000


GLIPR1
NM_006851
5.58
0.000


ITGA3
NM_002204
24.64
0.000


LMO3
NM_018640
129.25
0.000


NEFL
NM_006158
7.14
0.000


NKX6-2
NM_177400
11.50
0.000


NRTN
NM_004558
3.39
0.001


PDCD11
NM_014976
2.48
0.000


PLP1
NM_000533
20.64
0.000


TGFB1
NM_000660
6.66
0.000


TSPAN12
NM_012338
2.58
0.006
















TABLE X7







Up-regulated Regenerative genes (NSLC vs. hNPC)












Fold change of NSLC



GeneSymbol
Accession Number
compared to hNPC1
p-value













ATR
NM_001184
2.57
0.000


BMP2
NM_001200
17.71
0.000


BMP4
NM_001202
20.55
0.001


CAV3
NM_001234
26.23
0.000


CCND1
NM_053056
10.34
0.000


CDKN2A
NM_058197
5.57
0.000


CEBPB
NM_005194
2.58
0.000


GAL
NM_015973
12.21
0.000


GAP43
NM_002045
4.27
0.000


HOXB4
NM_024015
133.37
0.000


SMAD3
NM_005902
2.27
0.000









In order to investigate the differentiation potential of NSLCs to neuronal lineages (Neurons, astrocytes, and oligodendocytes), neurospheres were dissociated and plated in laminin/poly-D-Lysine (10 μg/ml; Sigma) in differentiation medium for two weeks. The differentiation towards neuronal lineage was performed using two different mediums: NbActive medium (BrainBits™) supplemented with Brain Derived Neurotrophin Factor (BDNF, 20 ng/ml, Peprotech), all-trans-retinoic acid (ATRA, 5 μM, Spectrum), and bFGF (40 ng/ml, Peprotech) or NeuroCult™ differentiation medium (NeuroCult™ Differentiation kit, StemCell Technologies), supplemented with BDNF (20 ng/ml, Peprotech) and bFGF (40 ng/ml, Peprotech). After two weeks in culture, the cells were stained with the neuronal marker βIII-tubulin, astrocyte markers GFAP and S100p, andoligodendrocyte marker CNPase. The cells were fixed with 4% formaldehyde and the primary antibodies were added in 5% normal goat serum/PBS as follows: Mouse antibody βIII-tubulin (1:200, Abcam), rabbit antibody S100 (1:100, Abcam), and Chicken antibody CNPase (1:50, Abcam). Secondary antibodies are added in 5% normal goat serum/PBS as follows: Goat anti mouse Alexa546™ (1:200, Invitrogen), Goat anti rabbit Alexa488™ (1:200, Invitrogen), and Goat anti-chicken cy5 (1:100, Jackson ImmunoResearch Labs).


Immunohistochemistry analysis showed that NbActive medium promoted the differentiation equally to neuronal (48.66±14.07%, βIII-tubulin) and potential early oligodendrocyte lineages (50.01±4.04%, CNPase) and to a lower percentage of astrocyte cells (2.68±1.13%, S100p), while NS-A differentiation medium induced the differentiation mainly to neurons (64.89±4.11%, βIII-tubulin) and astrocytes (35.94±4.04%, S100beta), and a low percentage of potential early oligodendrocytes cells (8.68±2.71%, CNPase). The NSC-A medium was selected over NbActive for further differentiation studies. Differentiation of cells in NS-A differentiation medium promote the differentiation of hNPC and NSLC similarly as shown in Table 17 by the decrease of the percentage of sox2, musashi and nestin positive cells. NSLCs were differentiated to neuronal (74.3±0.1, GABA), astrocyte lineage (65.6±0.0, S100beta) and to a lower percentage of oligodendrocyte cells (5.2±0.6, CNPase). The same pattern of tripotent lineage differentiation was observed with hNPCs (Table 17).









TABLE 17







The percentage of cells stained positive for neural stem cell and


neuronal lineage markers in transfected and untransfected cells.


NSLCs and hNPCs were cultured in NS-A-differentiation medium


supplemented with BDNF (20 ng/ml) and FGF (40 ng/ml),


cultures were incubated at 37° C., 5% CO2, 5% O2 for three weeks.


The percentage of immunopositive cells was determined by


Cellomics ™ and represented as mean ± SD (n = 5).
















Sox2
Nestin
Musashi
S100
O4
GABA





Tripotent
hNPC
73.8 ±
46.1 ±
22.1 ±
20.8 ±
6.4 ±
68.5 ±


medium

0.5
5.2
7.0
1.3
2.9
1.6



NSLC
68.6 ±
41.0 ±
26.7 ±
65.6 ±
8.2 ±
74.3 ±




3.9
5.4
5.0
0.0
0.6
0.1









Several additional antibodies to neuronal antigens were used to characterize, in more detail, the nature of differentiated cells. Antibodies against microtubule-associated protein (MAP2b), NCAM, and synaptophysin were used as recommended by the antibody manufacturer. After three weeks in differentiation medium, there was a differentiation-induced reduction in markers of precursors cells and an increase in mature neuronal markers. The percentage of neural precursor markers such as Sox2 were decreased during differentiation, while p75NTR, βIII-tubulin and GABA were increased with lengthening differentiation time (FIG. 6); however, 04 positive cells were very low after 3 weeks of differentiation of hNPCs (6.4±2.9) and NSLCs (8.2±0.6). Synaptophysin, an antibody used to identify functional neuronal cells, was increased following 2 and 3 weeks of differentiation, indicating maturity of the neuronal cells. GABA and acetycholine markers were increased following 2 weeks of differentiation and decreased at week 3.


The morphological changes and expression of a number of neuronal antigens and genes show that the above method results in normal and viable neuronal cells. Additionally, the newly formed neuronal cells have the morphological criteria of neurons. In addition to the above markers, the differentiated cells were evaluated by characterizing morphological markers of neurite differentiation. Neuron type cells (cells strongly expressing βIII-tubulin) showed neurite formation after differentiation, including an increase in the average number of neurites per neuron (from e.g. 1.38±0.1) The same pattern was observed in plII-tubulin positive cells. Accordingly, the average neurite length (118.3±3.5 μm) and the number of branch points (3.28±0.3) per neuron also increased. The differentiated neuron-like cells developed long neurites that were greater than three cell diameters in length with a growth cone at the end, expressed neuron-specific genes, and stopped proliferating after the induction of differentiation.


Further differentiation was performed using an optimised medium that promoted the differentiation towards oligodendrocyte lineage. NSLCs and hNPCs were cultured in NS-A differentiation medium as described previously supplemented with FGF-2 (10 ng/ml, Peprotech) and sonic hedgehog (SHH, 100 ng/ml, Peprotech) for 4 days. After 4 days medium was changed to NS-A differentiation medium supplemented by T3 (60 ng/ml, Peprotech), IGF1 (10 ng/ml, Peprotech), NT-3 (10 ng/ml, Peprotech), and PDGF (10 ng/ml, Peprotech). Cells were cultured for 20 days at 37° C., 5% CO2.









TABLE 18







The percentage of cells stained positive for neural stem cell and


neuronal lineage markers in transfected and untransfected cells.


NSLCs and hNPCs were cultured in differentiation medium


supplemented with SHH (100 ng/ml, Peprotech ), T3 (60 ng/ml,


Peprotech), IGF1 (10 ng/ml, Peprotech), NT-3 (10 ng/ml, Peprotech),


and PDGF (10 ng/ml, Peprotech) to induce differentiation towards


oligodendrocytes. The percentage of immunopositive cells was


determined by Cellomics ™ and represented as mean ± SD (n = 5).













% of positive








cells
Sox2
Nestin
Musashi
S100
O4
GABA





hNPC
84.3 ±
26.9 ±
51.8 ±
33.4 ±
40.1 ±
89.6 ±



3.7
4.4
2.9
1.9
6.4
0.8


NSLC
69.3 ±
24.3 ±
45.1 ±
51.6 ±
8.5 ±
76.9 ±



4.4
2.5
11.1
9.5
0.6
1.4









Quantification of the differentiation of hNPCs and NSLCs revealed a population of cells that were positively stained for 04. As shown in Table 18, the percentage of 04 positive cells was more pronounced in differentiated hNPC (40.1±6.4%) as compared to differentiated NSLCs (8.5±0.6%) when using the above differentiation protocol.


This study showed that transfecting the cells with one or two neurogenic transcription factors in the presence of a DNA demethylator or small molecules for epigenetic modification achieves stable reprogrammed cells (NSLCs). Like a DNA demethylator, epigenetic modification (inhibition of acetylation and methylation) are sometimes useful in boosting the reprogramming process. These cells possess and retain neural stem cell properties as determined by: (1) the expression of neural stem cell genes and proteins, (2) the capacity to generate and grow as neurospheres starting from a single cell, and (3) to differentiate to neuronal lineages in differentiation conditions. When differentiated to neurons, cells display one or more neural-specific morphological, physiological and/or immunological features associated with a neuronal cell type. Useful criteria include morphological features (long processes or neurites), physiological, and/or immunological features such as expression of a set of neuronal-specific markers or antigens. Furthermore, NSLCs readily turn into a tripotent-like precursor cell with differentiation potential to a high percentage of neuronal, astrocytes and lower percentage of oligodendrocyte populations.


Example VI

Implication of BMP Signaling Pathway in the Reprogramming of HFFs


This study was designed to evaluate the role of Noggin in the process of de-differentiation of HFFs towards NSLCs. HFFs were cultured and treated with cytochalasin B as described in Example III. After two days of treatment, cells were transfected by Nucleofection as described in Example II with the constructed vector Msi1/Ngn2. Briefly, after preparing the cells, they were mixed with 2 μg of total DNA (Msi1/Ngn2) and were co-transfect with MBD2 (2 g), by the Amaxa's Nucleofector™ according to the manufacturer's protocol. The samples were then transferred into a Laminin (10 μg/ml, Sigma) coated culture plate and cultured in the presence of Neural Proliferation Medium (NeuroCult™ proliferation Kit, StemCell Technologies) with recombinant hFGF (20 ng/ml, Peprotech), recombinant hEGF (20 ng/ml, Peprotech), and with or without the presence of Noggin (20 ng/ml, Peprotech). Samples were collected at different time points (1, 3, 4, 6, and 8 days) to analyze neuronal gene expression by RT-PCR and protein expression levels by immunohistochemistry.


Fluorescent imunohistochemical staining was performed on samples after 4 days of transfection as previously described in Example I. Transfected cells were stained and analyzed for expression of Sox2, the percentage of Sox2 was 33.3±1.00% in the presence of Noggin compared to 27.5±0.50% without the presence of noggin at day 4. RT-PCR analysis of relative expression of neuronal precursor cell markers such as nestin and Sox2 after transfection of HFFs with pCMV-Msi1-2A-Ngn2 and pCMV6-XL5-MBD2 with or without the presence of Noggin (20 ng/ml) was associated with an increase in nestin and Sox2 starting at day 3 and maintained until day 8 (Table 19). No difference in the expression was noticed in the absence of Noggin. Inhibiting the BMP signaling pathway by Noggin thus enhanced reprogramming, but had no reprogramming effect on its own.









TABLE 19







RT-PCR analysis of relative expression of neuronal precursor cell markers such as nestin and


Sox2 after transfection of HFF with pCMV-Msi1-2A-Ngn2 and pCMV6-XL5-MBD2 with or


without Noggin (20 ng/ml). Relative expression of Sox2, and nestin was increased after


transfection with and without Noggin.













ACHE
GFAP
NES
SOX2
TUBB3


















Rel.
Std.
Rel.
Std.
Rel.
Std.
Rel.
Std.
Rel.
Std.



Exp.
Dev.
Exp.
Dev.
Exp.
Dev.
Exp.
Dev.
Exp.
Dev.




















 #1 Msi1/Ngn2 +
7.08
1.70
2.97
0.42
1.33
0.10
0.93
0.91
1.37
0.10


MBD2/+Noggin












Day 1












 #2 Msi1/Ngn2 +
7.34
1.03
2.01
0.08
1.28
0.18
0.60
0.10
0.98
0.05


MBD2/+Noggin












Day 2












 #3 Msi1/Ngn2 +
9.67
2.41
15.13
1.66
1.98
0.20
6333.63
277.87
0.95
0.07


MBD2/+Noggin












Day 3












 #4 Msi1/Ngn2 +
11.68
2.65
194.07
25.22
4.19
0.52
20231.33
1034.29
1.90
0.45


MBD2/+Noggin












Day 4












 #5 Msi1/Ngn2 +
3.58
0.66
227.99
16.83
1.68
0.09
6298.51
289.84
0.96
0.17


MBD2/+Noggin












Day 6












 #6 Msi1/Ngn2 +
10.89
0.57
650.34
22.92
4.42
0.03
18134.90
63.93
1.81
0.06


MBD2/+Noggin












Day 8












 #7 Ctrl
1.01
0.19
1.00
0.05
1.00
0.02
1.12
0.70
1.00
0.09


Untransfected












+Noggin Day 1












 #8 Msi1/Ngn2 +
2.79
0.83
1.62
0.19
0.99
0.08
1.28
0.25
0.75
0.01


MBD2/−Noggin












Day 1












 #9 Msi1/Ngn2 +
3.79
0.91
1.47
0.08
1.23
0.08
1.36
0.08
0.72
0.07


MBD2/−Noggin












Day 2












#10 Msi1/Ngn2 +
6.18
0.59
14.60
1.85
2.62
0.30
10949.28
448.28
0.90
0.01


MBD2/−Noggin












Day 3












#11 Msi1/Ngn2 +
5.63
0.74
74.56
16.56
2.97
0.21
19623.99
3109.69
0.75
0.11


MBD2/−Noggin












Day 4












#12 Msi1/Ngn2 +
3.21
0.96
232.42
5.47
1.47
0.07
15311.64
1909.23
0.86
0.03


MBD2/−Noggin












Day 6












#13 Msi1/Ngn2 +
3.82
0.52
496.99
75.81
3.32
0.32
26892.31
1817.05
2.05
0.10


MBD2/−Noggin












Day 8












#14 Ctrl
1.08
0.57
1.01
0.14
1.00
0.04
1.15
0.81
1.00
0.00


Untransfected












−Noggin Day 1









Example VII

NSLCs Created from HFF Cells are not Skin-Derived Precursors (SKPs)


It's known that cells termed skin-derived precursors (SKPs) may reside in adult human skin (Fernandes et al., 2004). These cells are capable of proliferating in response to EGF and bFGF and express nestin, versican and fibronectin, and can differentiate into both neuronal and mesodermal progeny. In order to verify that NSLCs are distinct from SKPs, differentiation towards adipocyte cells was performed. Adipose derived stem cells (ADSC) were maintained in StemPro™ MSC serum free medium (Invitrogen) on flasks coated with CellStart™ (Invitrogen). CellStart™ was diluted 1:100 in dPBS/Ca2+/Mg2+ and the flask incubated for 2 hours at 37° C. Cells are passaged every 3 to 4 days using Accutase™ and medium was changed every 2 days. Three to four days before initiating differentiation, ADSCs and NSLCs were seeded in 6-wellplates in CellStart™ (1:100 in dPBS/Ca2+/Mg2+/2 hours at 37° C.) coated tissue culture plates. When cells reached confluence (after 3 to 4 days), proliferation media were replaced by differentiation medium consisting in DMEM/F12 (50:50), ITS (1:100), HEPES (1:100), GlutaMAX™ (1:100), T3 (0.2 nM), Rosiglitasone (0.5 μg/ml), IBMX (100 μM) and Dexamethasone (1 μM). Three days after, IBMX and dexamethasone were withdrawn from the differentiation medium. At day 10, cells were fixed with a 4% formaldehyde solution for 10 min and stained with Oil Red O (Invitrogen) staining solution for 15 min. Staining was removed and cells washed twice with PBS. Adipose cells appeared red with lipid droplets specifically stained with Oil Red O, however NSLCs were stained negative, with no presence of lipid droplets in the cells, and the cells adopted neuronal cell morphology.


Immunohistochemistry analysis confirmed that NSLCs are distinct from SKPs (FIG. 24): NSLCs stained positive for p75NTR and negative for fibronectin and versican, while SKPs express fibronectin and versican and do not express p75NTR (Fernandes et al., 2004). This study indicates that NSLCs represent a tripotent-like precursor cell and they are not a subpopulation of SKPs.


Example VIII

BDNF Release from Neural-Like Cells (NLCs)


Neural Stem-Like Cells (NSLCs) differentiated into neuronal and glial cells were kept in culture for 55 days, and BDNF released in the conditioned medium was measured by antigen-capture ELISA at different time points and compared to the release in mature neurons (ScienCell), undifferentiated Neural Human Normal Precursor cells (NHNP, Lonza) as well as to undifferentiated NSLCs and untransfected cells (HFF). Conditioned medium from each group was collected, centrifuged, and then stored at −80° C. until assaying. BDNF concentrations were measured by ELISA kits (BDNF Emax Immunoassay System, Promega Corporation, USA), according to the manufacturer's instructions. Briefly, 96-well ELISA immunoplates were coated with Anti-BDNF (CatNb #G700B) diluted 1/1000 in carbonate buffer (pH 9.7) and incubated at 4° C. overnight. The following day, all wells were washed with TBS-Tween™ 0.5% before incubation with Block/Sample buffer 1× at room temperature for one hour without shaking. After blocking, standards and samples were added to the plates and incubated and shaken (450±100 rpm) for 2 h at room temperature. Subsequently, after washing with TBS-Tween™ wash buffer, plates were incubated for 2 h with Anti-Human BDNF pAb (1:500 dilution in Block & Sample 1× Buffer) at 4° C. After incubation, plates were washed five times with TBS-Tween™ 0.5% wash buffer and 100 μl of diluted Anti-IgYHRP Conjugate was added to each well (1:200 dilution in Block & Sample 1× Buffer) and incubated for 1 hour at room temperature with shaking (450±100 rpm). Then, plates were washed five times with TBS-Tween™ 0.5% wash buffer and 100 μl of TMB One Solution was added to each well. Following 10 minutes incubation at room temperature with shaking (450±100 rpm) for the BDNF plate, a blue color formed in the wells. After stopping the reaction by adding 100 μl of 1N hydrochloric acid, the absorbance was read at 450 nm on a microplate reader (Synergy 4™) within 30 minutes of stopping the reactions. Concentration of released BDNF in the supernatants was determined according to the standard curves.


ELISA results revealed that BDNF was released at the same concentration from differentiated Neuron-Like Cells (NLCs differentiated from NSLCs) and normal Human neuron cells starting at day 11 and remained until day 55 (Table 20), while no BDNF (except for tiny amounts in the untransfected HFF group) was released in the other groups.









TABLE 20







Quantification of BDNF release by Neural-Like Cells (NLCs) that


had been differentiated for 55 days from Neural Stem-Like Cells


(NSLCs) that had been created from transfected HFFs. BDNF


release from NLCs into the medium, at different time points,


was measured by antigen-capture ELISA and compared to


BDNF release of normal mature human neurons (ScienCell).











Control medium
Neurons
NLC













day 0





day 11
1.55
30.25
22.99


day 18
0.33
29.49
25.15


day 24
0.33
22.01
26.39


day 34
0.23
25.53
32.21


day 41
0.27
19.02
22.43


day 55
0.02
20.73
30.01









In addition to adopting neuronal morphology criteria, the NLCs were functional and possessed the capacity to release neurotrophic factor (BDNF). Generating reprogrammed neuronal-like cell lines that can locally deliver these neurotrophic factors could be used as a method to treat several neurological conditions and may offer crucial benefits in regeneration and functional recovery from brain and other injuries.


Example IX

Reprogramming of Different Cell Types Towards NSLCs:


This study was performed to investigate the capacity of keratinocytes (Invitrogen), human Adipocytes Derived Stem Cells (ADSCs, Invitrogen) and human hematopoietic stem cells (CD34+, Invitrogen) cells into neural stem-like cells.


Preparation of Human CD34+ Cells, Human ADSC and Human Keratinocytes:


Human mobilized peripheral blood CD34+ cells were purchased from StemCell Technologies and expanded as a floating culture in Petri Dishes in complete StemPro™®-34 Serum-free Medium (Invitrogen) supplemented with Stem Cell Factor (SCF, 150 g/ml, Peprotech), Granulocyte Colony-Stimulating Factor (GM-CSF, 37.5 ng/ml, Peprotech) and IL-3 (75 ng/ml, Peprotech). Medium supplemented with cytokines was changed everyday 2-3 days after centrifugation of the cell suspension at 300×g for 10 min. Every other day the cytokines were added directly to the culture without changing the media. Cells were incubated at 37° C., 5% CO2. For their passaging, cells were centrifugated, resuspended in the above medium plus cytokines and placed into the adequate number of Petri dishes.


Human Adipose-Derived Stem Cells (ADSC) were purchased from Invitrogen and expanded in complete StemPro™ MSC Serum-free medium (Invitrogen) on CellStart™™ (Invitrogen) coated flasks (diluted 1:100 in PBS containing Ca2+/Mg2+) at a cell density of 1×104 cells/cm2. Medium was replaced every two days with fresh pre-warmed complete StemPro™ MSC SFM. Cells were incubated at 37° C., 5% CO2. Cells were sub-passaged when 80% confluent by incubation for 3-5 min in pre-warmed TrypLE™™ (Invitrogen) and then collected in StemPro™ MSC medium. After centrifugation at 1500 rpm for 5 min, cells were seeded on CellStart™™ coated flasks as described above.


Primary human keratinocytes were purchased from Invitrogen and expanded in Defined Keratinocyte Serum-free medium on Coating matrix (Invitrogen) coated flasks (Invitrogen) at a cell density of 5×103 cells/cm2. The cells were incubated at 37° C., 5% CO2. Media was replaced with fresh, complete growth media every two to three days until subculture. Once the cells had reached 70-80% confluency, media was removed and the cells were incubated in Versene™ (Invitrogen) for 3-5 min at room temperature. Versene™ was removed, and pre-warmed 0.05% trypsin-EDTA (Invitrogen) was added to the flasks. After 5-10 min incubation, growth medium containing Soybean Trypsin inhibitor (Invitrogen) was added to the flasks and the cells gently triturated. After centrifugation at 100×g for 10 min, cells were resuspended in the desired volume of pre-warmed, complete growth medium on coated flasks as described above.


Prior to transfection, cells were trypsinized and transiently co-transfected with pCMV-Msi1-Ngn2 and pCMV6-XL5-MBD2 as previously described in Example IV using the Shuttle and plated into a culture plate coated with laminin (Sigma, 10 g/ml). Starting one day after transfection, cells were treated with VPA (1 mM) for 4 days and the medium was changed gradually to proliferation medium supplemented with FGF (20 ng/ml) and EGF (20 ng/ml) and were cultured for 18 days at 372C, 5% CO2 and 5% O2. Cells were then analyzed for neural stem cell markers by RT-PCR and Immunohistochemistry.


Further analysis and quantification of the reprogrammed cells revealed a population of NSLCs engendered from keratinocyte and CD34 cells. RT-PCR Analysis revealed an increase of relative expression of neural stem cell markers such as Sox2, nestin, GFAP, and βIII-tubulin after transfecting Keratinocyte and CD34+ by Msi1 and Ngn2. Relative expression of nestin and GFAP was enhanced in NSLCs created from keratinocytes and CD34+ cells as compared to NSLCs from HFFs; however, the reverse was true for Sox2 and ACHE expression. βIII-tubulin (TUBB3) and Map2b expression was highest in NSLCs created from CD34+ cells, followed by NSLCs created from HFF (Table 21). This data shows that different types of NSLCs with different gene expression profiles (and characteristics) can be created from different types of starting/source cells (and the same has been observed for creating some other types of stem-like cells discussed in this application). The data is also intriguing since it was not expected that keratinocytes (which are derived from the ectoderm just as endogenous neural stem cells) would have a lower expression than HFFs for all the genes analyzed except for Nestin (it was expected that keratinocytes would be the easiest to reprogram into NSLCs since they are derived from the ectoderm).









TABLE 21







RT-PCR analysis was performed after one month of transfection of human fibroblasts (HFF),


Keratinocytes, and CD34+ cells with Msi1/Ngn2 (MSI1/NGN2), in the presence MBD2 with


VPA treatment. Cells were cultured on coated culture plates in proliferation medium


(StemCell Technologies) supplemented with EGF (20 ng/ml) and FGF (20 ng/ml) for 18 days.


Untransfected cells were considered as negative control.














NES
MAP2
TUBB3
ACHE
GFAP
SOX2




















Rel.
Std.
Rel.
Std.
Rel.
Std.
Rel.
Std.
Rel.
Std.
Rel.
Std.



Exp.
Dev.
Exp.
Dev.
Exp.
Dev.
Exp.
Dev.
Exp.
Dev.
Exp.
Dev.






















#1 Day 12
1.00
0.07
1.00
0.05
1.00
0.01
1.01
0.15
1.00
0.02
1.08
0.59


Untransfected














HFF














#2 Day 12 HFF
2.25
0.03
21.48
2.09
3.41
0.45
12.92
1.88
558.69
80.08
71513.12
14146.80


Msi1/Ngn2 +














MBD2














#3 Day 18 HFF
2.56
0.15
17.12
0.14
2.65
0.02
4.13
0.64
75.96
8.82
84794.40
318.54


Msi1/Ngn2 +














MBD2














#4
1.07
0.54
1.00
0.07
1.00
0.02
1.01
0.19
1.06
0.48
1.00
0.01


Untransfected














Keratinocytes














#5 Day 12
11452.65
1137.13
0.96
0.11
6.78
0.28
1.09
0.05
5815.54
510.91
975.81
7.47


Keratinocytes














Msi1/Ngn2 +














MBD2














#6 Day 18
12593.79
431.06
0.93
0.04
6.41
0.27
0.48
0.03
1295.15
32.05
1047.17
139.48


Keratinocytes














Msi1/Ngn2 +














MBD2














#7
1.00
0.04
1.01
0.16
1.00
0.00
1.00
0.01
1.10
0.66
1.01
0.21


Untransfected














CD34+














#8 Day 18
839.57
134.51
346.61
33.97
33.91
4.38
0.28
0.00
2790.18
304.43
25080.35
35.93


CD34 +














Msi1/Ngn2 +














MBD2














hNPC
4.56
0.07
278.36
11.50
0.81
0.06
72.65
1.83
1285.73
5.27
565552.30
41717.72









Immunohistochemistry revealed positive staining for GFAP, Sox2, and nestin as shown in FIG. 7. NSLCs developed from HFF yield a higher percentage of positive staining for Sox2 and GFAP (55.8±3.8 and 78.1±2.4) as compared to CD34+ cells (42.8±2.7 and 24.2±4.4), and keratinocytes (47.1±2.1 and 43.4±8.9). The percentage of nestin positive cells was high in Keratinocytes (77.6±10.7) and HFF (68.45±12.9) and lower in CD34+ cells (15.5±2.7) (Table 22). Sox2 and Nestin positive staining was undetectable in ADSCs.









TABLE 22







The percentage of Sox2 and nestin positive cells for neural stem cell markers


after transfecting fibroblast, keratinocyte, and CD34+ cells with pCMV-Msi1-


Ngn2 in the presence of MBD2 and VPA. Cells were cultured on coated


culture plates in proliferation medium (StemCell Technologies) supplemented


with EGF (20 ng/ml) and FGF (20 ng/ml) for 18 days. Untransfected cells


were considered as negative control. The percentage of immunopositive cells


was determined by Cellomics ™ and represented as mean ± SD (n = 5).











% positive cells
Untransfected cells
Fibroblasts
Keratinocytes
CD34+





Sox2
1.5 ± 1.7
55.8 ± 3.8
47.1 ± 2.1
42.8 ± 2.7


GFAP
0.04 +/− 0.2
78.1 ± 2.4
43.4 ± 8.9
24.2 ± 4.4


Nestin
 0.3 +/− 0.3
68.45 ± 12.9
 77.6 ± 10.7
15.5 ± 2.7









NSLCs generated from keratinocytes and CD34+ cells were tested for tripotent capacity. Further differentiation studies were performed to induce differentiation of these NSLCs towards neuronal lineage, using NeuroCult™ differentiation medium (NeuroCult™ differentiation Kit, StemCell Technologies) supplemented with BDNF (20 ng/ml, Peprotech) and bFGF (40 ng/ml, Peprotech) as described in Example V. NSLCs generated from HFFs and hNPCs were used as controls, cultures were incubated at 37° C., 5% CO2, 5% O2 for three weeks. Samples were collected or fixed at Day 14 and 28 following differentiation for further analysis. RT-PCR analysis revealed decrease of undifferentiated genes (Nestin and Sox2) and increased of differentiated genes (Map2, βIII-tubulin, CNPase, and GFAP) as shown in Tables 23A, 23B, 23C and 23D.









TABLE 23A







RT-PCR analysis was performed on NSLCs generated from human fibroblasts (HFF),


keratinocytes, and CD34+ cells that were cultured on Poly-D-Lysin/Laminin coated


culture plates in differentiation medium for 28 days (StemCell Technologies) supplemented


with BDNF (20 ng/ml) and FGF (40 ng/ml). hNPCs (Lonza) were considered as a positive control.


hNPCs had a much lower increase in ACHE, GFAP, and MAP2b (which actually decreased in


hNPCs), but an increase in Nestin, compared to NSLCs under differentiation conditions.
















NES
MAP2
TUBB3
ACHE
GFAP
SOX2
SOX9
CNP
























Rel.
Std.
Rel.
Std.
Rel.
Std.
Rel.
Std.
Rel.
Std.
Rel.
Std.
Rel.
Std.
Rel.
Std.



Exp.
Dev.
Exp.
Dev.
Exp.
Dev.
Exp.
Dev.
Exp.
Dev.
Exp.
Dev.
Exp.
Dev.
Exp.
Dev.


























hNPC
1.00
0.08
1.00
0.10
1.00
0.08
1.01
0.16
1.00
0.09
1.01
0.16
1.00
0.12
1.00
0.09


Control


















Diff. hNPC
3.86
0.20
0.65
0.05
4.87
0.57
0.74
0.52
97.26
7.13
1.85
0.21
0.50
0.04
1.43
0.05


Day 14


















Diff. hNPC
1.86
0.06
0.68
0.02
3.67
0.13
1.33
0.09
102.74
1.89
1.29
0.01
0.73
0.05
1.37
0.02


Day 28


















NSLC
1.00
0.04
1.00
0.04
1.00
0.04
1.00
0.03
1.00
0.01
1.00
0.01
1.00
0.02
1.00
0.05


Control


















Diff. NSLC
1.38
0.01
1.00
0.09
2.06
0.02
1.57
0.24
1.79
0.12
0.73
0.01
0.56
0.01
1.31
0.05


Day 14


















Diff. NSLC
0.62
0.02
0.90
0.08
5.14
0.21
6.47
0.78
5.70
0.15
1.30
0.02
0.79
0.03
1.41
0.01


Day 28


















HFF-NS
1.00
0.00
1.00
0.05
1.00
0.01
1.00
0.07
1.00
0.00
1.00
0.07
1.00
0.01
1.00
0.02


Control


















Diff.
2.70
0.08
3.08
0.12
3.24
0.14
59.93
5.85
478.97
0.27
2.90
0.32
0.81
0.03
4.02
0.35


HFF-NS


















Day 14


















Diff.
1.27
0.05
1.48
0.11
1.59
0.03
24.62
1.00
576.80
20.98
1.52
0.00
0.86
0.08
2.74
0.23


HFF-NS


















Day 28


















Kerat-NS
1.00
0.06
1.00
0.02
1.00
0.03
1.00
0.11
1.00
0.01
1.00
0.07
1.00
0.02
1.00
0.01


Control


















Diff.
2.43
0.06
3.48
0.08
2.82
0.11
56.22
5.58
665.91
10.52
3.09
0.29
1.01
0.14
3.72
0.17


Kerat-NS


















Day 14


















Diff.
0.81
0.03
1.72
0.00
1.61
0.18
26.09
1.12
673.65
11.34
1.29
0.03
1.12
0.03
2.02
0.05


Kerat-NS


















Day 28


















CD34+-NS
1.00
0.05
1.00
0.07
1.00
0.04
1.00
0.08
1.00
0.00
1.00
0.08
1.00
0.02
1.00
0.07


Control


















Diff.
2.21
0.04
3.47
0.07
2.75
0.04
57.87
6.68
407.54
52.07
2.90
0.18
1.10
0.05
3.54
0.02


CD34+-NS


















Day 14


















Diff.
0.79
0.04
1.48
0.01
1.83
0.37
26.92
3.73
485.51
10.66
1.02
0.04
1.20
0.09
2.34
0.05


CD34+-NS


















Day 28
















TABLE 23B







RT-PCR analysis was performed on undifferentiated NSLCs generated from human fibroblasts (HFF),


keratinocytes, and CD34+ cells that were cultured on Laminin coated culture plates in Proliferation


medium for 4 days (StemCell Technologies) supplemented with EGF (20 ng/ml) and FGF (20 ng/ml).


Relative expression calibrated to undifferentiated hNPCs.
















NES
MAP2
TUBB3
ACHE
GFAP
SOX2
SOX9
CNP
























Rel.
Std.
Rel.
Std.
Rel.
Std.
Rel.
Std.
Rel.
Std.
Rel.
Std.
Rel.
Std.
Rel.
Std.



Exp.
Dev.
Exp.
Dev.
Exp.
Dev.
Exp.
Dev.
Exp.
Dev.
Exp.
Dev.
Exp.
Dev.
Exp.
Dev.


























Undifferentiated
1.00
0.08
1.00
0.10
1.00
0.08
1.01
0.16
1.00
0.09
1.01
0.16
1.00
0.12
1.00
0.09


hNPC Control


















Day 4


















Undifferentiated
1.23
0.05
0.12
0.00
1.12
0.04
0.09
0.00
21.45
0.26
0.65
0.01
0.28
0.01
0.37
0.02


NSLC Control


















Day 4


















Undifferentiated
0.94
0.00
0.12
0.01
0.92
0.01
0.03
0.00
0.38
0.00
0.37
0.02
0.32
0.00
0.31
0.00


HFF-NS


















Control Day 4


















Undifferentiated
1.00
0.06
0.09
0.00
0.97
0.03
0.03
0.00
0.23
0.00
0.38
0.03
0.26
0.00
0.30
0.00


Kerat-NS


















Control Day 4


















Undifferentiated
1.10
0.05
0.12
0.01
0.95
0.04
0.04
0.00
0.33
0.00
0.44
0.04
0.26
0.00
0.30
0.02


CD34+-NS


















Control Day 4
















TABLE 23C







RT-PCR analysis was performed on differentiated NSLCs generated from human fibroblasts (HFF),


keratinocytes, and CD34+ cells that were cultured on Poly-D-Lysin/Laminin coated culture plates in


differentiation medium for 14 days (StemCell Technologies) supplemented BDNF (20 ng/ml) and


FGF (40 ng/ml). Relative expression calibrated to Day 14 differentiated hNPCs.
















NES
MAP2
TUBB3
ACHE
GFAP
SOX2
SOX9
CNP
























Rel.
Std.
Rel.
Std.
Rel.
Std.
Rel.
Std.
Rel.
Std.
Rel.
Std.
Rel.
Std.
Rel.
Std.



Exp.
Dev.
Exp.
Dev.
Exp.
Dev.
Exp.
Dev.
Exp.
Dev.
Exp.
Dev.
Exp.
Dev.
Exp.
Dev.


























Diff. hNPC
1.00
0.05
1.00
0.07
1.00
0.12
1.15
0.80
1.00
0.07
1.00
0.11
1.00
0.08
1.00
0.03


Day 14


















Diff. NSLC
0.44
0.00
0.18
0.02
0.47
0.00
0.22
0.03
0.40
0.03
0.26
0.00
0.31
0.00
0.34
0.01


Day 14


















Diff. HFF-NS
0.66
0.02
0.56
0.02
0.62
0.03
2.96
0.29
1.86
0.00
0.58
0.06
0.52
0.02
0.87
0.08


Day 14


















Diff. Kerat-NS
0.63
0.02
0.51
0.01
0.56
0.02
2.78
0.28
1.56
0.02
0.64
0.06
0.54
0.08
0.79
0.04


Day 14


















Diff.
0.63
0.01
0.62
0.01
0.54
0.01
3.77
0.43
1.39
0.18
0.69
0.04
0.58
0.03
0.76
0.00


CD34+-NS


















Day 14
















TABLE 23D







RT-PCR analysis was performed on differentiated NSLCs generated from human fibroblasts (HFF),


keratinocytes, and CD34+ cells that were cultured on Poly D-Lysin/Laminin coated culture plates in


differentiation medium for 28 days (StemCell Technologies) supplemented with BDNF (20 ng/ml) and


FGF (40 ng/ml). Relative expression calibrated to Day 28 differentiated hNPCs.
















NES
MAP2
TUBB3
ACHE
GFAP
SOX2
SOX9
CNP
























Rel.
Std.
Rel.
Std.
Rel.
Std.
Rel.
Std.
Rel.
Std.
Rel.
Std.
Rel.
Std.
Rel.
Std.



Exp.
Dev.
Exp.
Dev.
Exp.
Dev.
Exp.
Dev.
Exp.
Dev.
Exp.
Dev.
Exp.
Dev.
Exp.
Dev.


























Diff. hNPC
1.00
0.03
1.00
0.02
1.00
0.04
1.00
0.07
1.00
0.02
1.00
0.01
1.00
0.07
1.00
0.02


Day 28


















Diff. NSLC
0.41
0.01
0.15
0.01
1.56
0.06
0.44
0.05
1.19
0.03
0.66
0.01
0.30
0.01
0.38
0.00


Day 28


















Diff. HFF-NS
0.64
0.03
0.26
0.02
0.40
0.01
0.59
0.02
2.12
0.08
0.43
0.00
0.38
0.04
0.62
0.05


Day 28


















Diff. Kerat-NS
0.44
0.02
0.24
0.00
0.42
0.05
0.62
0.03
1.50
0.03
0.38
0.01
0.40
0.01
0.44
0.01


Day 28


















Diff.
0.47
0.03
0.25
0.00
0.47
0.10
0.85
0.12
1.57
0.03
0.35
0.01
0.43
0.03
0.52
0.01


CD34+-NS


















Day 28









Fluorescent immunohistochemical staining was performed on samples after 14 days and 28 days of differentiation. The expression of Sox2 and Nestin was decreased time dependently in differentiated cells (HFF, keratinocyte, and CD34+). This decrease was associated with an increase of differentiated markers at day 28 such as GFAP (68.51±11.87 for HFF-NC, 59.55±9.12 for Keratinocyte NC, and 61.70±1.48 for CD34+-NC). A high percentage for βIII-tubulin positive cells was generated from differentiated NSLCs generated from HFF (57.83±4.49) as compared to βIII-tubulin positive cells generated from Keratinocytes (23.27±2.91) and CD34+ cells (39.15±7.99) (Table 24)









TABLE 24







The percentage of cells stained positive for neural stem cell markers and


neuronal lineage markers in hNPCs (Lonza) and transfected keratinocytes, HFF, and


CD34+ cells with pMsi1/Ngn2/MBD2. Transfected cells (NSLCs) were cultured in


Proliferation medium or differentiation medium for 28 days at 37° C., 5% CO2, 5% O2.


The percentage of immunopositive cells (Sox2, Nestin, GFAP, S100beta, and βIII-


tubulin) was determined by Cellomics ™ and represented as mean ± SD (n = 5).












% positive
Proliferation
14 days
28 days



cells
conditions
differentiation
differentiation





hNPC
Sox2
96.23 ± 0.51
59.05 ± 3.01
41.43 ± 6.05



Nestin
41.47 ± 0.23
10.77 ± 4.78
16.14 ± 7.41



S100β
37.38 ± 7.85
49.51 ± 2.39
n.d.



βIII-tubulin
 2.34 ± 0.43
11.54 ± 4.03
23.34 ± 4.77



GFAP
 1.16 ± 0.14
23.42 ± 2.51
48.04 ± 8.30


HFF-NC
Sox2
93.28 ± 0.53
79.48 ± 0.54
52.06 ± 9.07



Nestin
29.29 ± 4.72
 1.15 ± 0.46
 2.18 ± 1.96



S100β
13.51 ± 0.28
80.75 ± 3.50
 79.38 ± 10.62



βIII-tubulin
 3.91 ± 0.33
 42.16 ± 15.07
57.83 ± 4.49



GFAP
 8.41 ± 0.73
 59.66 ± 11.48
 68.51 ± 11.87


Keratinocyte-NC
Sox2
96.55 ± 1.01
76.93 ± 5.13
63.11 ± 8.54



Nestin
40.10 ± 8.41
 2.67 ± 1.61
 3.57 ± 0.48



S100β
13.58 ± 4.97
 76.6 ± 9.72
 74.75 ± 11.21



βIII-tubulin
 6.42 ± 2.94
20.58 ± 8.34
23.27 ± 2.91



GFAP
 9.36 ± 0.34
43.43 ± 2.44
59.55 ± 9.12


CD34+-NC
Sox2
95.49 ± 2.6 
81.18 ± 1.24
63.46 ± 5.14



Nestin
 51.68 ± 14.27
12.64 ± 1.27
8.46 ± 4.6



S100β
 30.1 ± 1.03
72.40 ± 4.5 
79.57 ± 8.52



βIII-tubulin
 5.82 ± 2.08
 25.04 ± 19.95
39.15 ± 7.99



GFAP
13.99 ± 5.48
 51.79 ± 13.68
61.70 ± 1.48





n.d. = not determined; ± = standard deviation


CD34+-NC: neuronal cells generated after differentiation of NSLCs generated from CD34+ cells. Each data point represents the analysis of at least 1000 cells from at least 8 images.






The % of Sox2 positive cells decreased faster, the % of Nestin positive cells generally decreased slower, and the % of cells expressing one of the differentiation markers (S100, βIII-tubulin, GFAP) generally increased slower in hNPCs than in the NSLCs during differentiation. Out of the three types of created NSLC lines, the % of cells expressing one of the differentiation markers (S100β, βIII-tubulin, GFAP) generally increased slowest in NSLCs created from keratinocytes and fastest in NSLCs created from HFFs.


This study indicates that NSLCs can be created from keratinocytes and CD34+ blood cells, and these cells share morphology and markers similarly to NSLCs generated from HFF. Similarly to hNPCs, NSLCs created from keratinocytes, CD34+ cells, and HFFs had a tendency to differentiate more towards an astrocyte lineage than a neuronal lineage (except NSLCs created from HFFs had an almost similar number of βIII-tubulin positive and GFAP positive cells) as shown by the high percentage of GFAP positive cells during differentiation, which was confirmed by S100beta staining. However, the proportion of astrocyte and neuronal cells generated from hNPCs was lower in same culture conditions, indicating that NSLCs generated from HFF, Keratinocytes, and CD34+ cells can give rise to a higher number of neuronal and astrocyte cells as compared to hNPCs. NSLCs, whether created from HFFs, Keratinocytes or CD34+ cells (or potentially even some other cell), are tripotent cells and possess the capacity to differentiate to neurons, astrocytes, and oligodendrocytes similarly to hNPCs. However, RT-PCR and immunohistochemistry analysis of transfected ADSCs did not reveal any significant expression of neural stem cell genes, indicating a need to optimize conditions for turning ADSCs to NSLCs or to investigate the effect of others neurogenic factors that could turn these into NSLCs.


Example X

Fabrication 3D Extracellular Matrix (CDM)


Fibroblast cells were cultured in DMEM medium in the presence of 10% FCS as described in Example I, followed by seeding onto 12-well plates pre-coated with laminin (10 g/ml) at a concentration of 2×106 cells/ml in defined CDM Medium consisting of a 3:1 ratio of Dulbecco's modified Eagle medium (DMEM, high glucose (4.5 g/L) with L-glutamine and sodium pyruvate) and Ham's F-12 medium supplemented with the following components: EGF (4.2×10−10M), bFGF (2.8×10−10M), ITS (8.6×10−5M), dexamethasone (1.0×10−7M), L-ascorbic acid phosphate magnesium salt n-hydrate (3.2×10−4M), L-3,3′,5-triiodothyronine (2.0×10−10M), ethanolamine (10−4M), GlutaMAX™ (4×10−3M), glutathione (3.3×10−6M), and 1% penicillin/streptomycin/amphotericin B. By culturing the fibroblast cells at hyperconfluent density in this completely chemically defined medium causes them to enter a high synthetic phase with a slow-down in proliferation, leading to the production of a living tissue equivalent (LTE) consisting of multiple layers of fibroblasts within de novo 3D extracellular matrix (CDM) that is completely synthesized by the fibroblasts themselves.


Trans-Differentiation and Reprogramming of Cells within CDM


Day 14 CDM samples were treated with cytochalsin B (10 μg/ml, Calbiochem), with the concentration of cytochalsin B reduced from 10 μg/ml to 0 μg/ml (none) over 5 days while at the same time switching the medium from CDM Medium to NbActive medium. Samples were cultured for another 12 days at 37° C., 5% CO2, and the medium was changed every day. Samples were fixed to perform immunohistochemistry as described previously to detect Neuronal markers. The following antibodies were used: mouse anti-nestin 647 (1:100, BD) and anti-βIII-tubulin (1:200, Neuromics). No clear morphology change of the cells was observed within the CDM and the immunohistochemical analysis failed to detect βIII-tubulin positive cells. Thus, inducing the trans-differentiation of cells using only cytochalasin B and chemically-defined neural medium was not sufficient to reprogram the cells.


Next, Day 6 CDM samples grown in LAS pre-coated plates at 37° C. and 5% CO2, were exposed simultaneously to cytocahlasin B (10 μg/ml) over 5 days, histone deacetylation inhibitor (VPA, 4 mM, Calbiochem) and inhibitor of DNA methylation (5-Azacytidine, 5 μM, Sigma). Four days later, the medium was changed to differentiation medium consisting of a 3:1 ratio of CDM medium without the presence of EGF and NbActive medium (BrainBits™) supplemented with NT-3 (20 ng/ml, Peprotech) and BDNF (20 ng/ml, Peprotech). The ratio of the differentiation medium was increased gradually day after day until reaching 100% of complete differentiation medium. After two weeks of treatment, cells were fixed for immunohistochemical analysis to investigate the identity of the cells. FIG. 18 shows immunostained cells with βIII-tubulin at day 7, indicating the de-differentiation of fibroblast cells to neurons. However, one week later, these trans-differentiated cells reverted back to fibroblast cells and βIII-tubulin expression was lost (FIG. 8). The loss of morphology and βIII-tubulin expression after withdrawal of the priming agents indicate that complete conversion to functional and stable reprogrammed cells did not occur.


Next CDM was treated with VPA (4 mM), 5-Aza (5 μM) and cytochalasin B (10 μg/ml) as above. After 2 days of chemical treatment, fibroblast cells within the CDM were transfected with DNA using Lipofectamine reagent (Invitrogen) as per the manufacturer's protocol. 15 μg of the eukaryotic DNA expression vectors pCMV6-XL5-Pax6, pCMV6-XL5-Msi1 and pCMV6-XL4-Ngn2 (Origene) were used to transfect the cells. 24 hours later, the media was changed to Neural Progenitor Basal Medium (Lonza) supplemented with Noggin (50 ng/ml), EGF (20 ng/ml), and bFGF (20 ng/ml), and the cells were cultured at 37° C., 5% CO2 and 5% O2, and the medium was changed every day. At day 6, differentiation was initiated by adding gradually NBActive medium (BrainBits™) supplemented with NT-3 (20 ng/ml, Peprotech), all-trans-retinoic acid (ATRA, 5 μM, Spectrum), BDNF (20 ng/ml, Peprotech), and bFGF (40 ng/ml, Peprotech). To characterize the reprogrammed cells, immunohistochemical analysis and RT-PCR was performed at various time points according to the methods described in Example II using primers for nestin, βIII-tubulin, GFAP, MAP2b, and ACHE. In agreement with previous studies, un-transfected cells and cells transfected with Pax6 did not expressed genes specific for neuronal lineages (Table 25). On the other hand, following transfection with Msi1, levels of nestin and ACHE were increased to 4-fold and 8-fold, respectively, and this expression was maintained over the 12-day period. Also levels of GFAP mRNA was enhanced time dependently by approximately 14 times. Likewise, the same pattern was observed in Ngn2 transfected cells. While expression of βIII-tubulin and MAP2b were modestly increased following transfection with one neurogenic transcription factors the regulation of gene expression after transfecting the cells with two neurogenic factors, Msi1 or Ngn2 with Pax6, did not further increase the expression of neuronal genes. FIG. 19 shows that expression of these genes was enhanced when the cells were transfected with Msi1 and Ngn2, with βIII-tubulin enhanced to almost 6-fold at day 12.









TABLE 25







RT-PCR analysis of relative expression of neuronal precursor cell markers such as nestin, βIII-tubulin,


MAP2b, ACHE, and GFAP after transfection of fibroblast cells with pCMV6-XL5-Msi1, pCMV6-XL4-Ngn2,


pCMV6-XL5-Pax6, and pCMV6-XL5-MBD2. After 24 h following transfection, CDM I Medium was changed and


cells were cultured in proliferation medium (NPBM, Lonza) supplemented with EGF (20 ng/ml. Peprotech) and


bFGF (20 ng/ml, Peprotech) for one week. Differentiation was induced by changing the medium to NbActive


(BrainBits ™) supplemented with NT-3 (20 ng/ml), bFGF (20 ng/ml), ATRA (5 μM) and Forskolin (10 μM). Cells


were incubated at 37° C., 5%CO2, 5%O2 for 12 days. Relative expression of Msi1, Ngn2, Pax6, nestin, βIII-tubulin,


ACHE, MAP2b and GFAP in NSLCs and NLCs was increased after transfection with both transcription factors Ngn2


and Msi1 with MBD2 as the DNA demethylator.
















COL5A2
FBN2
NES
MAP2
TUBB3
SOX2
ACHE
GFAP
























Rel.
Std.
Rel.
Std.
Rel.
Std.
Rel.
Std.
Rel.
Std.
Rel.
Std.
Rel.
Std.
Rel.
Std.



Exp.
Dev.
Exp.
Dev.
Exp.
Dev.
Exp.
Dev.
Exp.
Dev.
Exp.
Dev.
Exp.
Dev.
Exp.
Dev.


























#1, +CytoB,
1.00
0.07
1.00
0.01
1.00
0.04
1.00
0.05
1.00
0.05
1.00
0.05
1.00
0.10
1.00
0.11


Control


















#2, −CytoB,
1.00
0.03
1.00
0.08
1.00
0.00
1.00
0.09
1.00
0.09
1.15
0.80
1.01
0.18
1.00
0.01


Control


















#3, +CytoB,
0.85
0.04
0.75
0.02
0.60
0.01
0.29
0.01
0.44
0.00
22.39
5.26
0.81
0.19
10.14
0.15


Msi1,


















GAD45b


















#4, −CytoB,
0.87
0.03
1.81
0.09
1.84
0.04
2.31
0.00
2.09
0.03
20.28
5.33
1.99
0.74
6.03
0.05


Msi1,


















GAD45b


















#5, +CytoB,
0.84
0.04
0.77
0.03
0.44
0.00
0.24
0.00
0.36
0.01
470.84
13.43
0.63
0.05
103.22
0.80


Ngn2,


















GAD45b


















#6, −CytoB,
0.75
0.07
1.97
0.02
1.83
0.00
4.40
0.16
2.02
0.10
789.33
60.35
1.70
0.13
110.48
4.90


Ngn2,


















GAD45b


















#7, +CytoB,
0.74
0.12
1.08
0.00
0.89
0.01
0.51
0.00
0.63
0.04
1.64
0.98
0.86
0.12
2.49
0.21


Pax6,


















GAD45b


















#8, −CytoB,
0.66
0.04
2.41
0.09
2.70
0.03
4.96
0.30
3.48
0.07
0.46
0.33
2.97
1.04
0.43
0.09


Pax6,


















GAD45b


















#9, +CytoB,
0.14
0.01
0.28
0.01
1.30
0.03
4.07
0.11
0.84
0.00
54768.27
6709.56
0.81
0.24
3391.96
64.63


Msi1, Ngn2,


















GAD45b


















#10, −CytoB,
0.12
0.00
0.73
0.03
5.28
0.21
50.84
1.23
4.93
0.28
17400.66
822.88
3.58
0.10
1255.76
5.27


Msi1, Ngn2


















GAD45b


















#11, +CytoB,
0.10
0.00
0.26
0.01
1.11
0.01
3.69
0.09
0.76
0.00
55588.41
1331.20
0.55
0.14
2849.96
261.51


Msi1, Ngn2


















MBD2


















#12, −CytoB,
0.44
0.01
1.47
0.06
5.49
0.14
47.30
0.11
5.50
0.31
14587.46
789.19
3.90
0.13
1424.04
39.29


Msi1, Ngn2


















MBD2


















#13, +CytoB,
1.11
0.04
1.09
0.06
0.92
0.08
0.68
0.01
0.82
0.03
63.93
2.81
1.19
0.17
17.43
1.86


GAD45b


















#14, −CytoB,
0.94
0.01
2.22
0.00
2.82
0.02
6.49
0.30
4.01
0.05
6.12
0.61
2.34
0.17
1.42
0.10


GAD45b


















#15, +CytoB,
0.83
0.00
0.83
0.05
0.36
0.01
0.16
0.01
0.36
0.00
3.42
3.74
0.63
0.37
2.18
0.12


MBD2


















#16, −CytoB,
0.68
0.02
1.55
0.04
1.57
0.05
1.47
0.01
2.00
0.00
0.52
0.29
1.45
0.15
0.55
0.04


MBD2


















#17, +CytoB,
1.10
0.01
1.16
0.03
1.37
0.01
1.12
0.06
0.86
0.06
5.59
1.48
1.07
0.27
1.70
0.46


Msi1, Ngn2


















#18, −CytoB,
0.93
0.04
2.52
0.10
3.48
0.01
9.01
0.02
4.55
0.18
1.78
1.46
3.83
0.42
0.59
0.01


Msi1, Ngn2


















#19, +CytoB,
0.20
0.03
0.36
0.01
1.25
0.05
6.68
0.31
0.72
0.02
66592.29
3481.89
2.57
0.03
4450.08
131.85


Msi1, MBD2


















#20, −CytoB,
0.12
0.00
0.64
0.03
4.70
0.22
77.51
0.11
4.12
0.11
19128.03
1542.00
8.14
0.13
999.22
24.75


Msi1, MBD2


















#21, +CytoB,
0.17
0.01
0.28
0.00
1.16
0.04
5.73
0.06
0.62
0.00
67945.51
3000.74
2.15
0.04
4736.83
11.92


Ngn2, MBD2


















#22, −CytoB,
0.17
0.00
0.78
0.03
4.32
0.08
68.89
5.26
4.01
0.04
16570.91
92.96
7.04
0.53
1427.13
13.19


Ngn2, MBD2


















#23, +CytoB,
0.71
0.05
0.79
0.06
0.87
0.01
0.63
0.06
0.67
0.04
2.86
0.70
1.08
0.08
2.08
0.11


Msi1


















#24, −CytoB,
0.66
0.04
1.92
0.17
2.03
0.02
2.77
0.02
2.68
0.02
0.32
0.12
1.85
0.65
0.58
0.04


Msi1









Same pattern of gene expression was observed when transfecting the cells with three transcription factors (Msi1, Ngn2, and Pax6), but the expression was less pronounced than in cells transfecting with just Msi1 and Ngn2. In terms of immunohistochemical analysis after the 12 days of the transfection, cells displayed neuronal markers after transfection with Msi1 or Ngn2, as indicated by the expression of nestin and MAP2b (FIG. 9). Cells transfected with pCMV-XL-PAx6 did not stain for Nestin and MAP2b.


This study shows that transfecting cells within CDM with only one neurogenic factor (Msi1 or Ngn2) induces morphological changes and expression of one or more markers of neural stem cells and neuronal cells. Since the reprogrammed cells expressed a key neurogenic factor, a neuronal precursor marker, and a mature neuronal marker at low percentage (10%), this suggests that cells within the CDM were transformed to NSLCs and then started to differentiated through the various phases of the neuronal determination and differentiation program induced in neural stem cells.


Example XI

Gene Expression Analysis of Reprogrammed Cells within CDM


This study was designed to test the effect of transfecting cells with Msi1 and Ngn2 in the presence of MBD2 in the reprogramming process. Cells were transfected after two days of pre-treatment with cytocahlasin B with the DNA expression vectors using Lipofectamine reagent as described in Example X. 15 μg of eukaryotic DNA expression vectors pCMV6-XL5-Musashi or pCMV6-XL4-Ngn2, and pCMV6-XL5-MBD2 (Origene), were used to co-transfect cells. After 24 hours, the media was changed to CDM:Neural Progenitor Maintenance Medium (1:1) supplemented with Noggin (50 ng/ml), EGF (20 ng/ml), and bFGF (20 ng/ml). Medium was changed every day by increasing the percentage of NPBM and decreasing CDM medium. Cells were cultured for 6 days at 37° C., 5% CO2 and 5% O2. After one week, differentiation was initiated by gradually supplementing the NPBM Medium with NT-3 (20 ng/ml, Peprotech), all-trans-retinoic acid (ATRA, 5 μM, Spectrum), BDNF (20 ng/ml, Peprotech), and bFGF (40 ng/ml, Peprotech). Samples were collected at the end of the study (day 14) and data were analyzed by gene array to identify genes that were reproducibly found to be specific for neuronal lineages.


Gene Expression Analysis


Gene expression analysis on 8 samples was performed as previously described in Example I with the customized Neuronal Markers 2 TLDA In order to identify the expression of genes related to neural stem cells, neuronal cells and glial cells, and growth factors expressed by the cells after transfection. The expression of oligodendrocyte genes, such as NKx2.2, olig2, and MAG was increased by Msi1 and Ngn2; however, the increased was more pronounced by Msi1 as compared to Ngn2 (Table 26). Two markers for astrocytes (GFAP and AQP4) were highly expressed after transfection with Msi1 and Ngn2 in the presence of the DNA demethylator MBD2. Interestingly, several markers of early neuronal cells were enhanced; 12 days after transfection, TDLA data revealed increases in specific markers for interneurons, such as somatostatin and calbindin1. Doublecortin (DCX), which is expressed by migrating immature cells during development, and acetylcholine (ACHE), an early marker of neuronal cells, were highly expressed in reprogrammed cells (Table 26). Transfection with Msi1 or Ngn2 increased the expression of dihydropyrimidinase-like 3 (DPYSL3), an early marker of newborn neurons to five-fold with Msi1 and seven-fold with Ngn2. Expression of microtubule-associated protein 2 (MAP2), an essential marker for development and maintenance of early neuronal morphology, and neuronal cell adhesion molecule (NCAM) were highly expressed with Msi1 and Ngn2. The expression of enolase-2, a marker of mature neurons, was 20-fold enhanced by Msi1 and Ngn2. Member of the NeuroD family NeuroD1 was highly expressed after transfection with Msi1 to 84.22 fold and to 34.27 by Ngn2. Gene expression of growth factors such as IGF-1, IGF-2, NPY and CSF-3 was enhanced following transfection with Msi1 or Ngn2. The expression of VEGF and GDNF genes were increased to almost five-fold and seven-fold by Msi1 and Ngn2, respectively. However in transfected cells, the expression of BDNF, EGF, and bFGF were not activated and even down-regulated as compared to untransfected cells. The expression of growth associated protein (GAP-43), a growth- and regeneration-associated marker of neurite extension, and expression of netrin, implicated in neuronal development and guidance, were highly expressed in transfected cells (Table 26). Expression of receptors for growth and neurotrophic factors was increased, such as type III receptor tyrosine kinase, Neurotrophic tyrosine kinase receptor, and neurotrophic tyrosine kinase. The fibroblast-specific markers vimentin and fibronectin were down-regulated in the reprogrammed cells.


Transfection of HFF with only Msi1 and Ngn2 in the presence of MBD2 increased the expression of glial cells and neuronal cells markers.









TABLE 26







Gene array of CDM transfected with pMsi1 and pNgn2 following the pre-treatment


with cytochalasin B (10 μg/ml), VPA (4 mM) and 5-Azacytidine (5 μM). Transfected


cells were cultured in differentiation medium (NbActive, BrainBits ™) supplemented


by ATRA (5 μM), bFGF (40 ng/ml) and BDNF (20 ng/ml).














Relative
Relative



Common name and
Company
expression
expression


Symbol
description
Gene ID
Msi1
Ngn2














Astrocytes and






oligodendrocytes






markers






NKx2-2
Markers for oligodendrocyte progenitors
NM_002509.2
1.72
10.19


OLIG2
Oligodendrocyte lineage transcription factor 2
NM_005806.2
1.72
1.52


MBP
Myclin-basic protein
NM-001025090.1
1.72
1.52


GFAP
Glial fibriliary acidic protein
NM_002055.4
6.04
2.41


AQP4
Aquaporin 4
NM_001650.4
1.72
1.52


DIO2
Deiodinase iodothyronine type II
NM_013989.3
8.29
10.61


NC markers






SST
Somatostatin, specific marker for interneurons
NM_001048.3
very high
very high


CALB1
Calbindin 1, interneuron marker
NM_004929.2
1.72
1.52


Tubulin1A
Are necessary for axonal growth
NM_006009.2
0.63
0.76


NES
Precursor neurons (nestin)
NM_006617.1
2.42
2.86


DCX
An early neuronal marker (Doublecortin)
NM_178151.1
1.72
1.52


ACHE
Acetylcholinesterase, marker of early neuronal
NM_015831.2
10.68
20.37



development





ENO2
A marker for neurons cells, enolase
NM_001975.2
0.55
0.54


NEUROD1
Neural marker; expression gradually increased
NM_002500.2
1.72
1.50



from neural precursor to fully differentiated






neuron





DPYSL3
Dihydropyrimidinase-like 3, marker of
NM_001387.2
0.62
0.71



immature neurons





MAP2
Microtubule-associated protein 2, essential for
NM_002374.3
1.99
1.70



development of early neuronal morphology and






maintenance of adult neuronal morphology





NCAM
Neural cell adhesion molecule 1
NM_18135.3
3.11
5.72


CEND1
Cell cycle exit & neuronal differentiation, early
NM_016564.3
6.68
8.28



marker of proliferating precursor cells that will






differentiate to neurons





Neuroregeneration






and survival genes






FGF2
Fibroblast growth factor
NM_002006.4
1.19
1.26


EGF
Epidermal growth factor,
Hs00153181_m1
28.37
52.13


IGF-1
Insulin growth factor-1
NM_000618.2
0.82
1.03


IGF-2
Insulin growth factor-2
NM_0000612.3
0.99
1.21


CSF3
Granulocyte colony-stimulating factor
NM_2219.1
very high
very high


BDNF
Brain derived growth factor, neurogenesis
NM-199231.1
8.54
7.84


GDNF
Glial derived neurotrophic factor
NM-000614.2
0.63
0.91


CNTF
Ciliary neurotrophic factor
NM_001025366.1
3.80
14.92


VEGF
Vascular endothelial growth factor
NM_130850.1
6.28
7.22


BMP-4
Bone morphogenetic protein 4
NM_002253.1
1.17
1.34


KDR
Typo III receptor tyrosine kinase)
NM_006180.3
113.85
43.87


NTRK2
Neurotrophic tyrosine kinase recepto (TrkB)
NM_000905.2
0.02
0.02


NPY
Neuropeptide Y
NM_009905.2
33.39
1.52


NTF-5
Neurotrophin 5
NM_006179.3
4.43
5.93


PIK3CG
phosphoinositide-3-kinase,
NW_002649.2
1.70
1.50


STAT3
Signal transduction transcription 3
NM_213662.1
3.15
2.24


Gap43
Growth associated protein 43
NM_002045.2
1.82
2.98


NTN1
Netrin 1, implicated in neuronal development
NM_004822.2
0.50
0.29



and guidance





NTRk2
Neurotrophic tyrosine kinase, receptor, type 2
NM_006180.3
0.02
0.02


L1CAM
L1 cell adhesion molecule, associated with
NM_024003.1
0.08
0.11



regenerating axons





LIMK1
LIM domain kinase 1
NM_002314.2
2.88
2.96


Vimentin
Radial glia and fibroblast marker
NM-003380.2
0.21
0.20


Fibronectin
fibronectin is a marker for fibroblasts
NM_212474.1
0.15
0.14









Example XII

Reprogramming of Cells within CDM by Lipofectamine and Nucleofection


This study was designed to improve transfection of CDM by combining lipofectamine and nucleofection and using two vectors pCMV6-XL5-Msi1 and pCMV6-XL4-Ngn2 individually or in combination together with pCMV-XL5-MBD2. Cells within Day 4 CDM were lipotransfected for 6 hours with Msi1/MBD2, Ngn2/MBD2 or Msi/Ngn2/MBD2 after 2 days of pre-treatment with or without cytochalasin B. In parallel, transfection was performed on fresh HFFs after the 6 hours using Nucleofection as described in Example II, and transferred on top of the CDM when the lipofectamine media was changed to fresh CDM medium. After 24 hours, the medium was changed to Neural Progenitor Basal Medium (NPBM, Lonza) with the presence of Noggin (50 ng/ml, Peprotech), recombinant hFGF (20 ng/ml, Peprotech), and recombinant hEGF (20 ng/ml, Peprotech). Differentiation was induced at day 7, by adding NSA-A differentiation medium (StemCell Technologies) for 21 days.


Gene Expression Analysis


Samples were collected at 8, 15, and 21 days to evaluate the nature of newly formed cells by analyzing the expression of several neuronal marker genes using RT-PCR according to the methods previously described in Example I. As shown in Table 27, cells transfected with one neurogenic transcription factor (Msi1 or Ngn2) express high levels of nestin and βIII-tubulin at day 8. The same pattern of expression was observed at day 15 and 21, while the expression was slightly decreased in the absence of cytochalasin B in cells transfected with Ngn2. The expression of all genes, except the mature neuronal marker MAP2b, were remarkably increased in cells transfected with both neurogenic transcription factors. The upregulation of these genes was slightly reduced in the absence of cytochalasin B, indicating its role in enhancing reprogramming.









TABLE 27







RT-PCR analysis of relative expression of neuronal stem cell markers such as nestin, Sox2, and


GFAP after transfection of fibroblast cells within the CDM with different combinations with or


without the co-treatment with cytochalasin B. Relative expression of Sox2, nestin, and GFAP in


NSLCs was increased after transfection with both transcription factors Ngn2 and Msi1 with


MBD2 as the DNA demethylator.














MSI1
NGN2
TUBB3
GFAP
NES
MAP2




















Rel.
Std.
Rel.
Std.
Rel.
Std.
Rel.
Std.
Rel.
Std.
Rel.
Std.



Exp.
Dev.
Exp.
Dev.
Exp.
Dev.
Exp.
Dev.
Exp.
Dev.
Exp.
Dev.






















#1 Day 8 CDM − CytoB
1.11
0.21
1.33
0.20
1.10
0.02
0.91
0.02
1.18
0.09
0.91
0.02


Control














#2 Day 8 CDM − CytoB
1.11
0.17
0.65
0.08
0.92
0.06
0.91
0.11
0.82
0.01
0.91
0.11


Control














#3 Day 8 CDM − CytoB
0.83
0.01
0.71
0.86
0.99
0.04
1.21
0.00
1.03
0.00
1.21
0.00


Control














#4 Day 8 CDM + CytoB
7.42
0.35
1.52
0.53
1.32
0.16
0.44
0.06
1.04
0.02
0.44
0.06


Control














#5 Day 8 CDM + CytoB
7.01
0.42
2.14
0.58
1.23
0.07
0.62
0.05
1.02
0.06
0.62
0.05


Control














#6 Day 8 CDM + CytoB
9.15
0.48
0.76
0.08
0.40
0.05
0.59
0.14
0.34
0.16
0.59
0.14


Control














#7 Day 15 CDM − CytoB
1.45
0.07
1.53
0.33
1.32
0.01
0.90
0.07
1.31
0.03
0.90
0.07


Control














#8 Day 15 CDM − CytoB
0.79
0.02
2.01
1.49
0.91
0.03
1.14
0.16
0.91
0.01
1.14
0.16


Control














#9 Day 15 CDM − CytoB
0.87
0.04
0.64
0.72
0.84
0.08
0.98
0.15
0.84
0.01
0.98
0.15


Control














#10 Day 15 CDM + CytoB
1.27
0.14
0.99
0.66
1.70
0.21
0.36
0.02
1.08
0.08
0.36
0.02


Control














#11 Day 15 CDM + CytoB
1.39
0.04
0.97
0.65
2.65
0.38
0.44
0.06
1.97
0.30
0.44
0.06


Control














#12 Day 15 CDM + CytoB
1.09
0.21
0.49
0.46
1.32
0.14
0.47
0.15
2.45
0.15
0.47
0.15


Control














#13 Day 21 CDM − CytoB
1.21
0.00
1.06
0.06
1.10
0.01
0.86
0.16
1.07
0.01
0.86
0.16


Control














#14 Day 21 CDM − CytoB
0.97
0.09
2.16
0.77
0.96
0.01
1.11
0.10
0.94
0.01
1.11
0.10


Control














#15 Day 21 CDM − CytoB
0.86
0.02
1.01
1.27
0.94
0.00
1.08
0.26
0.99
0.04
1.08
0.26


Control














#16 Day 21 CDM + CytoB
1.41
0.21
1.29
1.64
2.46
0.07
0.88
0.22
1.58
0.05
0.88
0.22


Control














#17 Day 21 CDM + CytoB
2.24
0.00
0.35
0.01
2.23
0.03
0.55
0.16
1.57
0.02
0.55
0.16


Control














#18 Day 21 CDM + CytoB
2.18
0.14
0.77
0.06
2.29
0.12
0.54
0.04
1.47
0.04
0.54
0.04


Control














#19 Day 8 CDM − CytoB
694.16
18.10
0.51
0.05
1.46
0.04
2.18
0.13
1.02
0.03
2.18
0.13


Msi1/MBD2














#20 Day 8 CDM − CytoB
2.38
0.29
4106.88
48.57
0.46
0.02
1.88
0.14
0.99
0.02
1.88
0.14


Ngn2/MBD2














#21 Day 8 CDM − CytoB
365.04
6.71
2702.81
55.69
4.44
0.02
2.95
0.38
5.11
0.05
2.95
0.38


Msi1/Ngn2/MBD2














#22 Day 8 CDM + CytoB
1262.00
63.21
0.75
0.91
0.54
0.03
2.48
0.11
1.16
0.05
2.48
0.11


Msi1/MBD2














#23 Day 8 CDM + CytoB
2.34
0.20
10963.51
19.89
0.53
0.00
2.27
0.26
1.00
0.06
2.27
0.26


Ngn2/MBD2














#24 Day 8 CDM + CytoB
869.15
65.33
6401.28
87.12
4.58
0.01
3.65
0.13
3.15
0.00
3.65
0.13


Msi1/Ngn2/MBD2














#25 Day 15 CDM − CytoB
41.07
1.74
2.58
0.36
1.43
0.05
0.58
0.06
1.34
0.07
0.58
0.06


Msi1/MBD2














#26 Day 15 CDM − CytoB
0.73
0.02
2192.64
15.74
0.95
0.08
1.01
0.09
0.99
0.03
1.01
0.09


Ngn2/MBD2














#27 Day 15 CDM − CytoB
45.59
2.33
3318.42
51.51
5.32
0.08
3.80
0.01
4.32
0.01
4.80
0.01


Msi1/Ngn2/MBD2














#28 Day 15 CDM + CytoB
106.34
4.43
4.90
1.70
1.47
0.01
0.57
0.10
1.19
0.03
0.57
0.10


Msi1/MBD2














#29 Day 15 CDM + CytoB
1.09
0.11
6715.95
505.86
1.30
0.05
0.70
0.17
1.18
0.07
0.70
0.17


Ngn2/MBD2














#30 Day 15 CDM + CytoB
46.77
0.76
2816.33
90.83
5.76
0.02
4.52
0.09
3.60
0.03
5.52
0.09


Msi1/Ngn2/MBD2














#31 Day 21 CDM − CytoB
22.94
1.09
10.09
2.72
1.08
0.07
0.58
0.08
1.17
0.02
0.58
0.08


Msi1/MBD2














#32 Day 21 CDM − CytoB
0.78
0.02
4450.56
255.75
1.00
0.03
0.75
0.21
1.09
0.03
0.75
0.21


Ngn2/MBD2














#33 Day 21 CDM − CytoB
24.02
0.86
2509.95
64.00
5.18
0.05
4.74
0.16
4.37
0.06
3.74
0.16


Msi1/Ngn2/MBD2














#34 Day 21 CDM + CytoB
54.17
1.41
8.31
3.32
1.42
0.05
0.70
0.22
1.71
0.02
0.70
0.22


Msi1/MBD2














#35 Day 21 CDM + CytoB
1.19
0.15
1180.19
27.29
1.21
0.06
1.03
0.34
1.31
0.04
1.03
0.34


Ngn2/MBD2














#36 Day 21 CDM + CytoB
81.66
1.34
7789.96
345.72
5.24
0.05
5.84
0.10
4.37
0.05
5.84
0.10


Msi1/Ngn2/MBD2










Imunohistochemical Analysis


Samples were collected at 4, 8, 14, and 21 days to evaluate the nature of any reprogrammed cells by analyzing the expression of several neuronal markers using immunohistochemical analysis according to the methods previously described in Example I. The immunhistochemical analysis at various time points revealed that within the first 8 days the expression of nestin was induced in a large proportion of cells and decreased time-dependently after inducing the differentiation (FIG. 10).


This study indicates that upon transfecting the cells with one or two neurogenic genes in the presence of cytochalasin B and MBD2, reprogrammed cells were stable in culture, responded to environmental changes (proliferation vs differentiation), and expressed neuronal markers for at least 24 days in culture.


Example XIII

Telomerase Activity of NSLCs


Telomerase is active in neural precursor cells and suggest that its regulation is an important parameter for cellular proliferation to occur in the mammalian brain (Caporaso G L et, 2003). This study was performed to evaluate telomerase activity in cell extracts of adherent NSLCs (NSLCs cultured on laminin-coated plates) as well as NSLCs in floating neurospheres (NSLCs cultured in plates with a low-bind surface) at early (P7) and late passage (P27). The telomerase activity of the 4 samples was measured by the PCR-based telomere repeat amplification protocol (TRAP) using the TRAPeze® Telomerase Detection Kit (Chemicon). Briefly, the cells were grown in 24-well plates, washed in PBS, and homogenized for 30 min on ice in buffer containing 10 mM Tris-HCl, pH 7.5, 1 mM MgCl2, 1 mM EGTA, 0.1 mM Benzamidine, 5 mM β-mercaptoethanol, 0.5% CHAPS and 10% Glycerol (1×CHAPS Lysis Buffer, provided in kit) and RNase Inhibitor. The samples were spun down and the protein concentration of the supernatant was determined using the BCA Assay. 900 ng of protein from each cell extract was added directly to the TRAP reaction mixture containing TRAP reaction buffer, dNTPs, template substrate (TS) primer, TRAP primer mix and Taq polymerase. The reaction mixtures were incubated at 30° C. for 30 minutes for template synthesis, followed by a PCR procedure (95° C./15 min for initial denaturation, 94° C./30 sec, 59° C./30 sec, 72° C./1 min for 32 cycles) for amplification of the extended telomerase products. To detect telomerase activity, polyacrylamide gel electrophoresis (PAGE) was performed for the reaction products on a 10% non-denaturing TBE gel. After electrophoresis, the gel was stained with SYBR® Green I Nucleic Acid Gel Stain for 30 minutes, followed by image capture using a Gel-Documentation System (Alpha Innotech).


All 4 samples were telomerase positive (as indicated by the TRAP product ladder) as shown in FIG. 11. As expected, the Heat-treated control (ΔH) showed no Telomerase activity (Negative Control). A 36 bp internal control band (S-IC) is used to monitor PCR amplification (to distinguish false-negative results). This S-IC band was observed for all samples except for the test samples. This may have been due to the excessively high telomerase activity in the test samples; amplification of the TRAP products and the S-IC control band are semi-competitive. All controls gave expected results (No TRAP products for CHAPS ctrl, and TRAP ladder of products for the positive control cells and the TSR8 control).


Example XIV

Tumor Formation Assay


Malignantly transformed cells show reduced requirements for extracellular growth promoting factors, are not restricted by cell-cell contact, and are often immortal. Anchorage-independent growth and proliferation is one of the hallmarks of malignant transformation, which is considered the most accurate and stringent in vitro assay for detecting malignant transformation of cells.


Adherent and neurosphere NSLCs at early and late passage (P7 and P25), as well as normal human neuroprogenitor cells (hNPCs), were investigated for the anchorage-independent growth. HFFs were used as a negative control and cervical carcinoma HeLa cells were used as a positive control. Cells were sedimented by centrifugation at 150×g for 3 min at room temperature (RT). The assay was performed using the CytoSelect™ 96-well cell transformation assay (CellBiolabs). The base agar layer (1.2%) was dissolved in 2×DMEM/20% PBS solution and 50 μl of the agar solution was added to the plate and incubated for 30 min at 4° C. to solidify. Prior to adding the cell agar layer, the plate was allowed to warm up for 15 minutes at 37° C. The cells were resuspended at different density (20.000 and 5000 cells/well), except the hNPCs were resuspended only at 5000 cells/well due to a lack of enough cells. The cells were mixed with the 1.2% agar solution, 2×DMEM/20% PBS, and cell suspension (1:1:1), and 75 μl of the mixture was transferred to wells already containing the solidified base agar layer, and was then placed in 4° C. for 15 minutes to allow the cell agar layer to solidify. 100 μl of proliferation medium (StemCell Technologies) was added and the plate was incubated for 8 days at 37° C. and 5% CO2 before being solubilized, lysed and detected by the CyQuant™ GR dye in a fluorescence plate reader. The fluorescence measurement was performed using the Flexstation™ (Molecular Devices) with a 485/538 nm filter.









TABLE 28







Fluorescence measurement (Relative Fluorescence Unit, RFU) indicate


that under the same conditions only carcinoma HeLa cells grow as an anchorage-


independent colony, while both hNPCs and NSLCs (adherent and floating neuro-


spheres) were negative for tumor growth in the standard agar plate tumor formation


assay (CytoSelect ™ cell transformation kit, Cell Biolabs Inc.).











Cell density/






Cell types
Hela
HFF
NSLCs
HNPCs














20.000
60.05 ± 8.70
14.82 ± 1.57
19.22 ± 1.85
19.00 ± 2.71


10.000
39.03 ± 3.97
13.73 ± 1.05
14.99 ± 1.12
21.61 ± 9.95


5000
24.70 ± 3.89
11.65 ± 0.57
12.29 ± 0.79
12.45 ± 0.73









As shown in Table 28, fluorescence measurement indicated that under the same conditions only carcinoma HeLa cells significantly grew and proliferated as anchorage-independent colonies, while both hNPCs and NSLCs (adherent and floating neurospheres) were negative for tumor growth (same value as HFFs (negative control) for 5,000 and 10,000 cells) in the standard agar plate tumor formation assay by visual observation of cells by light microscopic observation using bright field at 10× confirm Fluorescence measurement. Thus the transient transfection method and genes used allows the reprogramming of cells without the neoplastic transformation that generally occurs with stable transfection or certain genes via a series of genetic and epigenetic alterations that yield a cell population that is capable of proliferating independently of both external and internal signals that normally restrain growth.


Example XVI

No Genomic Integration of Plasmid DNA in NSLCs from Transient Transfection


The DNA plasmid Msi1/Ngn2 (designed and constructed in house) was used in transient transfection for generation of NSLCs along with MBD2 (for sample 1), or 5-Aza and VPA (for sample 2). Two weeks after transfection, Southern blot was performed to test for possible genomic integration of the plasmid DNA. 3 μg of genomic DNA extracted from the NSLC samples, as well as from HFF (a human fibroblast cell line) used as a negative control, was digested with several restriction enzymes including BgIII, PstI and StuI, subjected to electrophoresis on a 1% agarose gel and transferred to a positively charged nylon membrane (Roche). The membrane was hybridized in the DIG Easy Hyb™ buffer (Roche) at 42° C. overnight with a 1.2 kb Dig-labeled PCR probe amplified from the plasmid DNA using a set of primers. The membrane was washed twice at room temperature with 2×SSC, 0.1% SDS for 5 min per wash, twice with 0.5×SSC, 0.1% SDS at 65° C. for 15 min per wash. Hybridization signals of the membrane were detected using the CDP-Star™ substrate (Roche). The membrane was exposed to an X-ray film for analysis. The signals were stripped from the membrane using stripping buffer (0.2 M NaOH, 0.1% SDS). The membrane was re-hybridized with a 0.9 kb Dig-labeled PCR probe amplified from the plasmid DNA using a set of primers.


The Southern blot analysis (FIG. 12) with the 1.2 kb Dig-labeled PCR probe revealed distinct signals in the positive control samples where the Msi1/Ngn2 plasmid DNA was spiked into HFF genomic DNA for the equivalence of 1, 10 or 100 integrations per genome. There were a few weak and identical bands that appeared in the restriction enzyme digested genomic DNA from HFF, NSLC samples #1 and #2, suggesting that there is no plasmid DNA integration in the genomic DNA of NSLCs. These bands may represent the endogenous Ngn2 gene since the 1.2 kb Dig-labeled PCR probe contains a small part of the Ngn2 gene. This data shows that no, or only a tiny number of, NSLCs had plasmid integration into the host genome after transient transfection, and that the transfected genes are only present in the cell for a short period of time (less than two weeks).


Example XVII

Neuroprotective Effect of Transplanted hNSLCs in:


1) Animal Model of Multiple Sclerosis.


Multiple Sclerosis (MS) is an incurable inflammatory demyelinating disease of the central nervous system (CNS) (Frohman E M et al 2006). Therapies for MS rely on manipulation of the immune system, but with often modest effectiveness on reducing clinical episodes or permanent neurological disability, requiring frequent injections, and with sometimes-significant side effects (Langer-Gould A et al 2004). Experimental Allergic Encephalomyelitis (EAE) is an animal model of MS commonly used for studying disease mechanisms and testing potential therapies. EAE can be induced in a variety of species and strains of animals [mice, Rat, marmoset monkey, rhesus macaques] using various CNS antigens [Myelin Oligodendrocyte Glycoprotein (MOG), proteolipid protein (PLP) and myelin basic protein (MBP)].


After obtaining all appropriate animal approvals for the experiments, Female 7 to 8 weeks old C57BL/6 mice were purchased from Charles Rivers, and housed at MISPRO animal facility for one week before experimentation for adaption to the new environment. C57BL/6 mice were injected s.c. with 100 μg MOG 35-55 in CFA (Sheldon Biotechnology, McGill University) containing 5 mg/ml Mycobacterium tuberculosis H37Ra (Difco, inc), at 2 sites on the back. All mice received 200 ng pertussis toxin (List Biological Laboratories, Inc) i.p. on day 0 and 2, while clinical scores were calculated blindly daily during a 43 day period, according to the 0-5 scale as follows: 1, limp tail or waddling gait with tail tonicity; 2, waddling gait with limp tail (ataxia); 2.5, ataxia with partial limb paralysis; 3, full paralysis of 1 limb; 3.5, full paralysis of 1 limb with partial paralysis of second limb; 4, full paralysis of 2 limbs; 4.5, moribund; and 5, death.


Treatment of EAE Animal Model with and without the Cells:


hNSLC and hNPCs (1.5×106 cells in 200 μl PBS/each mouse) were given by single injection i.v. via the tail vein when the animals started to show symptoms of EAE (day 13 i.v). Both animals groups received cyclosporine (10 mg/kg/day) one day before the injection of cells and daily from the day of transplantation to avoid any rejection of the human cells. Sham-treated age-, sex-, and strain-matched mice, injected i.p. with PBS alone, were used as controls. All groups of animals were observed for 43 days. Animals were sacrificed at 43 days p.t., brains and spinal cord were harvested in 30% sucrose in PBS. Statistical analysis of the clinical scores revealed that the clinical signs of EAE were significantly attenuated in NSLC-injected animals as compared to control and hNPCs-injected animals. Cumulative scores was significantly reduced in the NSLC transplanted animals (FIG. 13) and the treatment has no effect on body weight.


2) Hemiplegic Animal Model (Unilateral Ablation of the Left Sensorimotor Cortex in Adult Rats).


After obtaining all appropriate animal approvals for the experiments, 8 rats per group (Sprague-Dawley, 250-300 g, Charles River) were anaesthetized using ketamine (Bimeda-MTC)/xylazine (50/10 mg/kg, Novopharm) and placed onto a stereotaxic frame. A midline cranial incision was performed with a sterile surgical scalpel blade, the cranial vault exposed and the bregma identified. The skull above the sensorimotor cortex was opened and the sensorimotor cortex area [0.5-4.0 mm caudal to bregma and 1.8-3.8 mm lateral to the midline (Paxinos and Watson 1986)] was carefully aspirated. After ablation, the treatments (Alginate, Alginate+hNPC, Alginate+NSLCs, RMx+NSLCs, RMx Only, Fibrin Gel, or Saline) were applied directly on the brain after ablation. The opening in the skull was then filled with Bone Wax. In case of a bleeding, small pieces of sterile homeostatic tissue were inserted into the lesion in order to stop the bleeding. The sutures were performed using Ethicon™ monofilament suture ½ circle needle shape. Surgeries were performed in sterile clean rooms, and topical antibiotics (Cicatrin®, GlaxoSmithKline) were applied to the exposed skull and scalp to limit local infection. Rats were immunosuppressed by daily injection i.p. of cyclosporine A (10 mg/kg/day) starting the day before the surgery until the end of the study period. The purpose of the cyclosporine A injection was to reduce the rat's immune reaction to the treatment. The immune-suppression was sustained until the end of the study to ensure that any potential failure of regeneration (if taken place) was not due to the immune reaction against the treatment. Functional scores were performed weekly, in all groups, sensorimotor impairment was evaluated based on the behavioural tests as described below.


Rotarod Test:


The rotarod speed was manually calibrated for the 10 and 20 RPM speed before all procedures. Animals were required to perch on the stationary rod for 30 sec to acclimate themselves to the environment. During this time, if any animal fell, it was placed back on the rod until it had achieved stationary capabilities for a period of 30 seconds. The animals were allowed 3 trials. The animals that were comfortable staying on the stationary rod for 30 sec were allowed to run with a constant speed of 10 and 20 RPM for 60 sec, and the number of falls were electronically recorded.


Beam Walking:


Beam walking measures hindlimb coordination by means of distance traveled across 100 cm beam (2.3 cm in diameter, 48 cm off the floor). Rats were systematically trained to walk along the elevated beam from start to finish with the aim of completing the task. A safe location, i.e, a flat box, is placed at the end of the beam so that the rat is motivated to complete the task.


Scale Used for Evaluation of Beam-Walking Performance













Scale
Performance characteristic







1
Animals fail to traverse the beam and do not place the hindlimb on the horizontal surface of the



beam


2
Animals fail to traverse the beam, but place the hindlimb on the horizontal surface of the beam



and maintain balance


3
Animals traverse the beam while dragging the hindlimb


4
Animals traverse the beam and place the hindlimb at least once during the traverse


5
Animals traverse the beam using the hindlimb to aid less than 50% of its steps on the beam


6
Animals traverse the beam using the left hindlimb to aid more than 50% of its steps on the beam


7
Animals traverse the beam with no more than two foot slips


8
Normal animals









Before the surgery, all the animals fell at least once from the rotarod, not because they had a walking or coordination problem, but because the speed was high. After the surgery (2 days), all the animals showed signs of significant walking and coordination problems leading to an increase in the number of falls from the rotarod. Three weeks after the surgery, the number of falls was clearly reduced for the animals receiving NSLCs as treatment compared to controls (FIG. 14).


Animals passed the beam-walking test before the surgery without any difficulty. The rats crossed the 100 cm beam and got to the safe spot without falling off the beam. Two days after surgery, all groups completely failed to pass the test, and the animals were not able to stay in balance on the beam. One week after the surgery, all the animals showed some improvement in their walking capacity, but no significant difference was noticeable between the different treated groups. From week 4 until week 26, the animals treated with NSLCs as well as RMx showed significant improvements in their walking capacity compared to the controls (FIG. 15).


Example XVIII

Transfection of HFF by Various Combinations of Genes Using the Shuttle® Device and Treatment with Different Small Molecules for Reprogramming to Mesendoderm-Like Cells


HFF cells were cultured as described in CDM II medium as described in Example I with only modifying EGF (5 ng/ml) and FGF (10 ng/ml), and transfecting using the Nucleofector™® 96-well Shuttle® Device (Lonza) following the procedure described in Example IV. The cells were transfected with various combinations of cDNA clones as described in Table 29. After transfection, the cells were plated on 0.1% Gelatin-coated plates and incubated at 37° C., 5% CO2, 5% O2. Medium was changed every other day according to Table 30. Cells were analyzed at Day 4 by Quantitative Real-time PCR.









TABLE 29







Various combinations of plasmids with potential to transfect


the cells towards mesendoderm lineage.












Day −2 to Day 0
Plasmids transfected at Day 01







 1
Untreated
Oct4, FoxD3, MBD2



 2

Oct4, T, MBD2



 3

Oct4, Mixl1, MBD2



 4

Oct4, Sox17, MBD2



 5

FoxD3, T, MBD2



 6

FoxD3, Mixl1, MBD2



 7

FoxD3, Sox17, MBD2



 8

T, Mixl1, MBD2



 9

T, Sox17, MBD2



10

Mixl1, Sox17, MBD2



13
Pre-treated with
Oct4, FoxD3



14
VPA & 5-Aza
FoxD3, T



15

FoxD3, Mix1l



16

FoxD3, Sox17



17

Oct4, FoxD3, T



18

Mixl1, Sox17, FoxA2



19

Oct4, FoxD3, T



20

Mixl1, Sox17, FoxA2








1where Oct4 = pCMV6-XL4-Oct4, FoxD3 = pCMV6-XL5-FoxD3, MBD2 = pCMV6-AC-MBD2, T = pCMV6-XL5-T, Mixl1 = pCMV6-XL5-MIXL1, Sox17 = pCMV6-XL4-SOX17, FoxA2 = pCMV6-XL5-FOXA2. All clones were purchased from Origene and prepared using the EndoFree Plasmid Maxi Kit (Qiagen).














TABLE 30







Medium composition from Day −2 to Day 10


Media Composition2











Day 0
Day 1
Day 2 to Day 3
Day 4 to Day 7
Day 8 to Day 10





CDM II (3:1 of
CDM II (50%) +
IMDM/F12 +
IMDM/F12 +
IMDM/F12 +


DMEM:F12;
IMDM/F12
NEAA + ITS +
NEAA + ITS +
NEAA + ITS +


GlutaMAX ™ 100x,
(50%) +
HSA + bFGF +
HSA + bFGF +
HSA + bFGF +


Dexthamesone,
NEAA + ITS +
EGF + VPA +
EGF + Activin
EGF + BMP4


19.7 μg/ml,
HSA + bFGF +
Activin A +
A + CHIR99021 +



Glutathione (500
EGF + VPA +
CHIR99021 +
BMP4



μg/ml, L-Ascorbic
Activin A +
BMP4




75 mg/ml, Selenious
CHIR99021





acid 2.5 μg/ml,






Insulin solution






10 mg/ml, T3






675 ng/ml,






ethanolamine






500X, bFGF






2.5 ug/ml, and Egf






(1.25 ug/ml) +






Activin A +HSA






2Supplements added to media at the following concentrations: Activin A (Peprotech, 30 ng/ml), HSA (Baxter, 0.5%), NEAA (Gibco, 1X), ITS (Gibco, 1X), EGF (Peprotech, 5 ng/ml), bFGF (Peprotech, 10 ng/ml), CHIR99021 (Stemgent, 2 uM), VPA (Stemgent, 1 mM), 5-Aza (Sigma, 0.5 uM), BMP4 (Peprotech, 10 ng/ml)







Cells were collected on Day 4 by detaching with TrypLE™, followed by centrifugation at 80×g for 5 minutes. Supernatant was aspirated and the cell pellet was frozen at −86° C. until ready for RNA Isolation. RNA isolation and quantification was performed as previously described in Example I. cDNA was prepared and quantitative real-time PCR was performed as previously described in Example II, except the following Taqman™® Gene Expression Assays (Applied Biosystems) were used:
















Gene
Taqman ™ ® Assay ID









GAPDH (housekeeper)
Hs99999905_m1



PPIA (housekeeper)
Hs99999904_m1



FOXA2
Hs00232764_m1



SOX17
Hs00751752_s1



Endogenous T
Hs00610073_g1



GSC
Hs00418279_m1



CXCR4
Hs00607978_s1



GATA4
Hs00171403_m1



CER1
Hs00193796_m1



CDH1 (E-cadherin)
Hs01023894_m1



p63
Hs00978340_m1



SOX2
SOX2_1078-ANY

















TABLE 31







Relative Expression FoxA2, Sox17, and Cxcr4 after transfecting HFFs once with


various gene combinations with potential to reprogram cells into mesoendoderm-


like cells. The exact values are not significantly accurate due to low RNA yield,


however a trend of increasing gene expression was detected for FoxA2, Sox17,


and CXCR4.











FOXA2
SOX17
CXCR4














Rel.
Std.
Rel.
Std.
Rel.
Std.



Exp.
Dev.
Exp.
Dev.
Exp.
Dev.
















Untreated HFF
1.00
0.04
1.00
0.04
1.00
0.04


Day 4 HFF untransf.
1.01
0.06
1.01
0.06
4.77
2.51


(+G.F),








Day 4 HFF untransf.
1.38
0.11
1.38
0.11
1.38
0.11


(−G.F),








Day 4 HFF Untransf.
0.98
0.02
0.98
0.02
3.32
3.31


(+G.F.),








Day 4 HFF Untransf.
4.12
4.07
1.28
0.06
1.28
0.06


(−G.F.),








Day 4 Oct4/FoxD3/MBD2
4.67
4.60
3.19
2.78
76.43
7.91


Day 4 Oct4/T/MBD2
3.91
3.55
4.33
2.36
15.18
2.52


Day 4 Oct4/Mixl1/MBD2
2.66
1.77
10.33
0.43
7.31
3.21


Day 4 Oct4/Sox17/MBD2
14.19
4.85
413533.31
127089.61
56.04
0.71


Day 4 FoxD3/T/MBD2
38.62
38.00
3.12
1.32
42.41
5.23


Day 4 FoxD3/Mixl1/MBD2
7.76
5.29
2.41
0.30
137.17
27.74


Day 4 FoxD3/Sox17/MBD2
26.02
1.95
50904.45
1523.33
131.03
17.53


Day 4 T/Mixl1/MBD2
3.67
3.26
5.64
4.15
14.04
2.89


Day 4 T/Sox17/MBD2
9.76
9.70
209797.21
24533.81
111.35
16.40


Day 4 Mixl1/Sox17/MBD2
3.60
3.10
237310.10
57448.60
36.76
1.07


Day 4 Oct4/FoxD3
13.87
0.16
13.87
0.16
35.44
14.57


Day 4 FoxD3/T
60.93
60.18
19.45
1.51
19.45
1.51


Day 4 FoxD3/Mixl1
21.20
2.31
28.96
8.66
62.31
55.82


Day 4 FoxD3/Sox17
96.88
3.60
54177.20
3313.15
44.57
41.51


Day 4 Oct4/FoxD3/T
25.99
18.15
12.27
1.26
21.17
11.33


Day 4 Mixl1/Sox17/FoxA2
1850864.68
98259.84
112641.65
15923.21
23.18
23.10


Day 4 Oct4/FoxD3/T
9.52
5.61
1.52
0.02
35.74
4.36


(IMDM/F12)








Day 4 Mixl1/Sox17/FoxA2
486705.82
19101.53
57060.09
1262.81
13.44
2.36


(IMDM/F12)
















TABLE 32







Expression of GATA4, CDH1 (E-cadherin), p63, and SOX2 relative to


untreated HFF control 4 days after transfecting HFF cells with various gene


combinations with potential to reprogram cells into mesoendoderm-like cells.













CDH1 (E-





GATA4
cadherin)
p63
SOX2
















Rel.
Std.
Rel.
Std.
Rel.
Std.
Rel.
Std.



Exp.
Dev.
Exp.
Dev.
Exp.
Dev.
Exp.
Dev.


















Untreated HFF
1.00
0.04
1.00
0.04
1.00
0.04
1.00
0.04


Day 4 HFF untransf.
12.13
0.70
1.01
0.06
3.09
1.45
1.11
0.21


(+G.F),










Day 4 HFF untransf.
4.48
0.85
1.38
0.11
3.11
2.54
1.38
0.11


(−G.F),










Day 4 HFF Untransf.
2.37
2.00
0.98
0.02
4.41
4.40
1.94
1.34


(+G.F.),










Day4 HFF Untransf.
6.12
3.33
1.28
0.06
13.23
7.43
1.28
0.06


(−G.F.),










Day 4 Oct4/FoxD3/MBD2
95.23
27.44
98.90
21.58
1.81
0.86
12.72
1.53


Day 4 Oct4/T/MBD2
33.66
10.30
1.42
0.02
2.05
0.87
2.62
1.67


Day 4 Oct4/Mixl1/MBD2
106.33
5.70
1.43
0.03
8.68
0.99
25.68
2.18


Day 4 Oct4/Sox17/MBD2
23.50
5.39
4.65
4.43
95.23
13.86
18.77
6.94


Day 4 FoxD3/T/MBD2
121.36
11.68
26.85
0.02
2.22
0.04
16.99
4.74


Day 4 FoxD3/Mixl1/MBD2
130.21
21.04
69.19
22.84
4.05
3.56
1.52
0.01


Day 4 FoxD3/Sox17/MBD2
99.49
30.30
6.89
3.69
1.78
0.01
15.19
9.08


Day 4 T/Mixl1/MBD2
110.30
3.55
1.36
0.00
1.36
0.00
6.64
2.25


Day 4 T/Sox17/MBD2
53.19
4.02
2.69
1.86
18.01
0.54
14.21
5.21


Day 4 Mixl1/Sox17/MBD2
16.53
16.50
2.91
2.13
13.44
6.68
10.55
3.27


Day 4 Oct4/FoxD3
66.45
26.34
47.31
47.30
13.87
0.16
23.87
14.31


Day 4 FoxD3/T
68.25
68.00
39.08
29.27
19.45
1.51
19.45
1.51


Day 4 FoxD3/Mixl1
78.18
78.00
21.20
2.31
21.20
2.31
25.10
3.20


Day 4 FoxD3/Sox17
176.45
93.54
15.64
0.60
15.64
0.60
26.78
16.35


Day 4 Oct4/FoxD3/T
12.27
1.26
12.27
1.26
12.27
1.26
12.27
1.26


Day 4 Mixl1/Sox17/FoxA2
85.89
64.52
20.06
20.00
3.67
0.13
13.66
0.66


Day 4 Oct4/FoxD3/T
89.05
50.00
10.40
8.14
1.52
0.02
1.52
0.02


Day 4 Mixl1/Sox17/FoxA2
6.16
6.10
1.23
0.04
1.23
0.04
1.23
0.04









Identification of gene combinations that may induce the formation of Mesendoderm-like cells was investigated by transfection with combinations of Oct4, Sox17, FoxD3, T, MixI1, FoxA2, and MBD2. As shown in Table 25 and 26, the Relative Expression of CXCR4 and GATA4, both Mesendoderm/Endoderm/Mesoderm markers, appear to be up-regulated in various combinations, most noticeably in FoxD3/MixI11/MBD2 and FoxD3/Sox17/MBD2. Similarly, FOXA2, a marker for Endoderm and Mesoderm, was up-regulated FoxD3/Sox17-transfected sample, although the expression is still very low. Four days following transfection, SOX17 is still highly expressed in the SOX17-transfected samples (50,000 to 400,000-fold as compared to the untreated HFF sample). The SOX17 gene expression represents leftover plasmid DNA (exogenous SOX17) that still remains 4 days post-transfection, and any endogenous SOX17 expression that may have been induced. Ectoderm markers CDH1, p63 and Sox2 were also up-regulated in some samples (e.g. Oct4/FoxD3/MBD2, Oct4/Sox17/MBD2).


Reprogramming HFFs into Pancreatic Progenitor-Like Cells:


HFF cells were cultured as described in Example I, and transfected using the Nucleofector™® 96-well Shuttle® Device (Lonza) following the procedure described in Example IV. The cells were transfected with various combinations of cDNA clones as described in Table 27. After transfection, the cells were plated on Fibronectin-coated collagen gels and incubated at 37° C., 5% CO2, 5% O2. Fibronectin-coated Collagen gel plates were prepared prior to transfection. Rat Tail Collagen I (Gibco) was diluted to 1.125 mg/ml using 10×PBS and distilled water, where 125 μl was added to each well of a 24-well plate and incubated in 37° C. for 40 minutes. After rinsing with 1×PBS, Fibronectin (BD Biosciences) was added on top of the gel at a concentration of 1.9 μg/well. Media was changed every other day according to Table 33. Cells were analyzed at Day 7 by Quantitative Real-time PCR.









TABLE 33







Plasmids and media composition from Day 0 to Day 14











Media Composition2












Plasmids transfected






at Day 01
Day 0
Day 1 to Day 3
Day 4 to Day 14





 1
FoxD3, Sox17, Pdx1,
CDM II +
DMEM/F12 +
DMEM/F12 + NEAA + ITS +



MBD2
Activin A + HSA
NEAA + ITS +
HSA + B27 + EGF + bFGF +


 2
FoxD3, Sox17, Ngn3,

HSA + B27 + EGF +
Retinoic Acid + FGF10 +



MBD2

bFGF + Activin A +
Cyclopamine + Noggin


 3
FoxD3, Mixl1, Pdx1,

CHIR99021 + Na




MBD2

Butyrate



 4
FoxD3, Mixl1, Ngn3,






MBD2





 5
Sox17, Mixl1, Pdx1,






MBD2





 6
Sox17, Mixl1, Ngn3,






MBD2





 7
FoxD3, Sox17, Mixl1,

DMEM/F12 +




Pdx1

NEAA + ITS +



 8
FoxD3, Sox17, Mixl1,

HSA + B27 + EGF +




Ngn3

bFGF + Activin A +



 9
FoxD3, Sox17, Pdx1,

CHIR99021 + Na




Ngn3

Butyrate + VPA +



10
FoxD3, Mixl1, Pdx1,

5-Aza




Ngn3





11
Sox17, Mixl1, Pdx1,






Ngn3






1where FoxD3 = pCMV6-XL5-FoxD3, Sox17 = pCMV6-XL4-SOX17, Mixl1 = pCMV6-XL5-MIXL1, Pdx1 = pCMV6-XL5-Pdx1, and Ngn3 = pCMV6-XL5-Ngn3. All clones were purchased from Origene and prepared using the EndoFree Plasmid Maxi Kit (Qiagen).




2Supplements added to media at the following concentrations: Activin A (Peprotech, 30 ng/ml), HSA (Baxter, 0.5%), NEAA (Gibco, 1X), ITS (Gibco, 1X), B27 (Gibco, 1%), EGF (Peprotech, 5 ng/ml), bFGF (Peprotech, 10 ng/ml), CHIR99021 (Stemgent, 2 uM), Na Butyrate (Stemgent, 1 mM), VPA (Stemgent, 1 mM), 5-Aza (Sigma, 0.5 uM), Retinoic Acid (Sigma, 2 uM), FGF10 (Peprotech, 50 ng/ml), Cyclopamine (Stemgent, 2.5 uM), Noggin (Peprotech, 50 ng/ml)







Cells were collected on Day 7 and RNA isolation and quantification was performed as previously described in Example I. cDNA was prepared and quantitative real-time PCR was performed as previously described in Example II, except the following Taqman™® Gene Expression Assays (Applied Biosystems) were used:
















Gene
Taqman ™ ® Assay ID









GAPDH (housekeeper)
Hs99999905_m1



PPIA (housekeeper)
Hs99999904_m1



FOXA2
Hs00232764_m1



SOX17
Hs00751752_s1



GATA4
Hs00171403_m1



Endo PDX1
PDX1_1201



SOX9
Hs00165814_m1



NGN3
Hs01875204_s1



NKX2-2
Hs00159616_m1



PAX4
Hs00173014_m1



INS
Hs02741908_m1



CXCR4
Hs00607978_s1










Identification of gene combinations that may induce the formation of Pancreatic Progenitor-like cells was investigated by transfection with combinations of FoxD3, Sox17, Pdx1, Ngn3, MixI1, and MBD2. FoxA2, a marker for Endoderm and Mesoderm, was slightly up-regulated for the FoxD3/Sox17/Ngn3/MBD2-transfected sample as compared to the GFP mock-transfected control sample. Similarly, CXCR4, also a marker for both endoderm and mesoderm, was slightly up-regulated (3-fold compared to GFP-ctrl) for the FoxD3/MixI1/Ngn3/MBD2-transfected sample. 7 days following transfection, SOX17 can still be detected for the samples transfected with SOX17 at varying levels (4 to 570-fold up-regulation as compared to the GFP-ctrl). The highest SOX17 expression up-regulation is detected for the sample transfected with Sox17/MixI1/Pdx1/Ngn3 (570-fold as compared to GFP-ctrl), which may suggest that this gene combination may increase the amount of SOX17 RNA in cells.


Example XIX

Reprogramming Human Adipocytes Derived Stem Cells (ADSC) to Pluripotent-Like Stem Cells (PLSC):


ADSCs (Invitrogen Corporation) were cultured in cell culture flasks with complete StemPro™-43 medium (Invitrogen) at 37° C., 5% CO2 and the medium was changed 3 times per week. After 3 days in culture cells (passage 5) were trypsinized and counted to be transfected. Cells were transiently transfected with one plasmid: pCMV6-Oct4-2A-Klf4-2A-Nanog, pCMV-Sall4-2A-Oct4-2A-Klf4-2A-Nanog, pCMV-Dax1-2A-Oct4-2A-klf4, pCMV-FoxD3-2A-Oct4-2A-klf4, pCMV-Oct4-2A-Klf4-2A-Sall4, pCMV-MBD2-2A-Oct4-2A-Klf4-2A, pCMV-AGR2-2A-Oct4-2A-Klf4-2A, or Rex1-EF-Oct4-2A-Klf4 (2 μg); or by two plasmids: pEF-Oct4nuc-IRES2-MBD2 with pCMV-Sox2nuc-IREC-Lin28 or pCMV-Klf4nuc-IRES2-Tpt1nuc or pEF-Stella-IRES2-NPM2, using Nucleofector™ as described in Example II. Following the transfection cells were cultured in 6-well plates in suspension with 50:50 ratio of adipocytes complete medium (StemPro™-43) and embryonic stem cells medium (mTeSR1). After two days in culture, cells were re-transfected with the same plasmids listed above and cells were plated in 96 well-plates coated with Matrigel™ (BD Biosciences) in the presence of mTesR complete medium supplemented with thiazovivin (0.5 μM), an ALK-5 inhibitor (SB 341542, Stemgent, 2 μM), and inhibitor of MEK (PD0325901, Stemgent, 0.5 μM). Medium was changed every day and cells were cultured for 22 days at 37° C., 5% CO2, 5% O2. Alkaline Phosphatase Detection Kit (AP, Millipore) and immunohistochemistry were performed to analyse the expression of pluripotency markers. ALP staining was performed using AP detection kit (Millipore) according to manufacturer's instructions.


Visual observation of reprogrammed cells was performed by Cellomics™ using a live staining for SSEA-4647 (BD Biosciences) and TRA-1-81555 (BD Biosciences) starting on Day 6 after transfection and every 5 days thereafter. Reprogrammed colonies of PLSCs, positively stained with SSEA-4 and TRA1-81, was observed only with Plasmid pCMV-Sall4-2A-Oct4-2A-Klf4-2A-Nanog, pEF-Rex1-EF-Oct4-2A-Klf4-2A-RFP, pEF-Oct4nuc-IRES1-MBD2 with pCMV-Sox2nuc-IRES1-Lin28, and pEF-Oct4nuc-IRES1-MBD2 with pCMV-Klf4nuc-IRES2-Tpt1nuc. These colonies emerged around Day 6 and maintained in culture up to the end of the study period (Day 22) with a stable morphology. Among the plasmids cited above, pCMV-Sall4-2A-Oct4-2A-Klf4-Nanog and pEF-Rex1-EF-Oct4-2A-Klf4-2A-RFP gave the highest number of colonies. Live staining showed that these colonies express typical pluripotency markers, including SSEA-4 and TRA1-81, and further analysis of these colonies showed that the colonies also expressed other ESC markers such as alkaline phosphatase and Oct4 (FIG. 16). When the cultures were treated with PD0325901 and SB431542 for up to 22 days, a 4-fold improvement in efficiency over the conventional method was obtained following the transfection of ADSCs with pCMV-Sall4-2A-Oct4-2A-Klf4-Nanog and pEF-Rex1-EF-Oct4-2A-Klf4-2A-RFP.


Based on the previous study, the highest reprogramming efficiency was observed using pEF-Rex1-EF-Oct4-2A-Klf4-2A-RFP and pCMV-Sall4-2A-Oct4-2A-Klf4-2A-Nanog. Another study was designed to ascertain the effect of pEF-Rex1-EF-Oct4-2A-Klf4-2A-RFP on the reprogramming efficiency and to investigate the effect of individual pluripotent genes Rex1, Oct4, and Klf4 in different combinations. ADSCs were transfected as above with pEF-Rex1-EF-Oct4-2A-Klf4-2A-RFP, pCMV6-XL5-Rex1, pCMV6-XL4-Oct4/pCMV6-XL5-Klf4, pCMV6-XL5-Rex1/pCMV6-XL4-Oct4, or pCMV6-XL5-Rex/pCMV6-XL5-Klf4. After the second transfection, ADSC were cultured in 96-well plates coated with Matrigel™ for 24 days in the presence of mTeSR1 medium supplemented with SB341542 and PD 0.325901 at 37° C., 5% CO2, 5% O2. In order to characterize subpopulations of cells after transfection, live staining, immunohistochemistry, and AP staining was used to follow the change in pluripotent markers. 1-5% of total cells transfected with Rex1/Oct4 or Rex1/Klf4 showed a SSEA4+ and TRA-1-81+ phenotype, and this pattern was stable until the end of the study period (Day 22). The observation over time showed that the phenotype of these colonies moved from an early SSEA-4+ phenotype to a late Oct4+/Sox2/Nanog+ phenotype by Day 22, which is closer to the final reprogrammed state of a pluripotent-like stem cell (FIG. 17).


Various genes were tested for their effect on reprogramming efficiency towards pluripotent-like cells. ADSC cells were cultured as described in Example IX with 2 days VPA and 5-AZA pre-treatment (1 mM and 0.5 μM respectively) in StemPro™ MSC SFM medium. Cells were transfected using the Nucleofector™® 96-well Shuttle® Device (Lonza) following procedure described in Example IV and using the transfection program EW-104 with the DNA mixes described in Table 34. Following transfection the cells were plated in StemPro™ MSC SFM medium described in example A on Matrigel™ (BD Biosciences) coated 24 well plates and incubated at 37° C., 5% CO2, 5% O2. On Day 1, media was changed to a mix of 75% StemPro™ MSC and 25% hES cell medium; the percentage of StemPro™ MSC was decreased every day over four days to have 100% hES cell medium by Day 4. From then onwards the medium was changed every two days. The hES cell medium consisted in Dulbecco's Modified Eagle's Medium (DMEM, Invitrogen) supplemented with 20% Knockout™ Serum Replacement (KSR, Invitrogen), 1 mM GlutaMAX™, 100 μM Non-essential Amino acids, 100 μM β-mercaptoethanol and 10 ng/ml Fgf-2. Different inhibitors and growth factors were added through the course of the experiment; these are listed in Table 34. Cells were analysed at Day 7 and Day 14 by immunohistochemistry analysis and at Day 7 by RT-PCR.









TABLE 34







Plasmids and media composition from Day 1 to Day 14.













From day
Plasmids






−2
transfected at


From



to day 0
day 0
From day 1 to day 3
From day 3 to day 7
day 7 to day 14





 1
VPA + 5-
pCMV6-XL4-
StemPro ™/hES
StemPro ™/hES
hES medium



Aza pre-
Oct4 +
medium + ActivinA
medium + ActivinA




treated
pCMV6-XL5-
(30 ng/ml) +
(30 ng/ml) +





Sox2 +
CHIR99021 (3 μM)
CHIR99021 (3 μM)





pCMV6-XL5-







MBD2





 2
VPA + 5-
pCMV6-XL4-
StemPro ™/hES
StemPro ™/hES
hES medium



Aza pre-
Oct4 +
medium + ActivinA
medium + ActivinA




treated
pCMV6-XL5-
(30 ng/ml) +
(30 ng/ml) +





FoxD3 +
CHIR99021 (3 μM)
CHIR99021 (3 μM)





pCMV6-XL5-







MBD2





 3
VPA + 5-
pCMV6-XL4-
StemPro ™/hES
StemPro ™/hES
hES medium



Az apre-
Oct4 +
medium + ActivinA
medium + ActivinA




treated
pCMV6-XL5-
(30 ng/ml) +
(30 ng/ml) +





UTF1 +
CHIR99021 (3 μM)
CHIR99021 (3 μM)





pCMV6-XL5-







MBD2





 4
VPA + 5-
pCMV6-XL4-
StemPro ™/hES
StemPro ™/hES
hES medium



Aza pre-
Oct4 +
medium + ActivinA
medium + ActivinA




treated
pCMV6-XL4-
(30 ng/ml) +
(30 ng/ml) +





DPPA4 +
CHIR99021 (3 μM)
CHIR99021 (3 μM)





pCMV6-XL5-







MBD2





 5
VPA + 5-
pCMV6-XL5-
StemPro ™/hES
StemPro ™/hES
hES medium



Aza pre-
Sox2 +
medium + ActivinA
medium + ActivinA




treated
pCMV6-XL5-
(30 ng/ml) +
(30 ng/ml) +





FoxD3 +
CHIR99021 (3 μM)
CHIR99021 (3 μM)





pCMV6-XL5-







MBD2





 6
VPA + 5-
pCMV6-XL5-
StemPro ™/hES
StemPro ™/hES
hES medium



Aza pre-
Sox2 +
medium + ActivinA
medium + ActivinA




treated
pCMV6-XL5-
(30 ng/ml) +
(30 ng/ml) +





UTF1 +
CHIR99021 (3 μM)
CHIR99021 (3 μM)





pCMV6-XL5-







MBD2





 7
VPA + 5-
pCMV6-XL5-
StemPro ™/hES
StemPro ™/hES
hES medium



Aza pre-
Sox2 +
medium + ActivinA
medium + ActivinA




treated
pCMV6-XL4-
(30 ng/ml) +
(30 ng/ml) +





DPPA4 +
CHIR99021 (3 μM)
CHIR99021 (3 μM)





pCMV6-XL5-







MBD2





 8
VPA + 5-
pCMV6-XL5-
StemPro ™/hES
StemPro ™/hES
hES medium



Aza pre-
FoxD3 +
medium + ActivinA
medium + ActivinA




treated
pCMV6-XL5-
(30 ng/ml) +
(30 ng/ml) +





UTF1 +
CHIR99021 (3 μM)
CHIR99021 (3 μM)





pCMV6-XL5-







MBD2





 9
VPA + 5-
pCMV6-XL5-
StemPro ™/hES
StemPro ™/hES
hES medium



Aza pre-
FoxD3 +
medium + ActivinA
medium + ActivinA




treated
pCMV6-XL4-
(30 ng/ml) +
(30 ng/ml) +





DPPA4 +
CHIR99021 (3 μM)
CHIR99021 (3 μM)





pCMV6-XL5-







MBD2





10
VPA + 5-
pCMV6-XL5-
StemPro ™/hES
StemPro ™/hES
hES medium



Aza pre-
UTF1 +
medium + ActivinA
medium + ActivinA




treated
pCMV6-XL4-
(30 ng/ml) +
(30 ng/ml) +





DPPA4 +
CHIR99021 (3 μM)
CHIR99021 (3 μM)





pCMV6-XL5-







MBD2





11
VPA + 5-
pCMV6-XL4-
StemPro ™/hES
StemPro ™/hES
hES medium



Aza pre-
Oct4 +
medium + ActivinA
medium + ActivinA




treated
pCMV6-XL5-
(30 ng/ml) +
(30 ng/ml) +





Sox2 +
CHIR99021 (3 μM) +
CHIR99021 (3 μM)





pCMV6-XL5-
VPA + 5-Aza






FoxD3





12
VPA + 5-
pCMV6-XL4-
StemPro ™/hES
StemPro ™/hES
hES medium



Aza pre-
Oct4 +
medium + ActivinA
medium + ActivinA




treated
pCMV6-XL5-
(30 ng/ml) +
(30 ng/ml) +





Sox2 +
CHIR99021 (3 μM) +
CHIR99021 (3 μM)





pCMV6-XL5-
VPA + 5-Aza






UTF1





13
VPA + 5-
pCMV6-XL4-
StemPro ™/hES
StemPro ™/hES
hES medium



Aza pre-
Oct4 +
medium + ActivinA
medium + ActivinA




treated
pCMV6-XL5-
(30 ng/ml) +
(30 ng/ml) +





Sox2 +
CHIR99021 (3 μM) +
CHIR99021 (3 μM)





pCMV6-XL4-
VPA + 5-Aza






DPPA4





14
VPA + 5-
pCMV6-XL4-
StemPro ™/hES
StemPro ™/hES
hES medium



Aza pre-
Oct4 +
medium + ActivinA
medium + ActivinA




treated
pCMV6-XL5-
(30 ng/ml) +
(30 ng/ml) +





FoxD3 +
CHIR99021 (3 μM) +
CHIR99021 (3 μM)





pCMV6-XL5-
VPA + 5-Aza






UTF1





15
VPA + 5-
pCMV6-XL4-
StemPro ™/hES
StemPro ™/hES
hES medium



Aza pre-
Oct4 +
medium + ActivinA
medium + ActivinA




treated
pCMV6-XL5-
(30 ng/ml) +
(30 ng/ml) +





FoxD3 +
CHIR99021 (3 μM) +
CHIR99021 (3 μM)





pCMV6-XL4-
VPA + 5-Aza






DPPA4





16
VPA + 5-
pCMV6-XL4-
StemPro ™/hES
StemPro ™/hES
hES medium



Aza pre-
Oct4 +
medium + ActivinA
medium + ActivinA




treated
pCMV6-XL5-
(30 ng/ml) +
(30 ng/ml) +





UTF1 +
CHIR99021 (3 μM) +
CHIR99021 (3 μM)





pCMV6-XL4-
VPA + 5-Aza






DPPA4





17
VPA + 5-
pCMV6-XL5-
StemPro ™/hES
StemPro ™/hES
hES medium



Aza pre-
Sox2 +
medium + ActivinA
medium + ActivinA




treated
pCMV6-XL5-
(30 ng/ml) +
(30 ng/ml) +





FoxD3 +
CHIR99021 (3 μM) +
CHIR99021 (3 μM)





pCMV6-XL5-
VPA + 5-Aza






UTF1





18
VPA + 5-
pCMV6-XL5-
StemPro ™/hES
StemPro ™/hES
hES medium



Aza pre-
Sox2 +
medium + ActivinA
medium + ActivinA




treated
pCMV6-XL5-
(30 ng/ml) +
(30 ng/ml) +





FoxD3 +
CHIR99021 (3 μM) +
CHIR99021 (3 μM)





pCMV6-XL4-
VPA + 5-Aza






DPPA4





19
VPA + 5-
pCMV6-XL5-
StemPro ™/hES
StemPro ™/hES
hES medium



Aza pre-
Sox2 +
medium + ActivinA
medium + ActivinA




treated
pCMV6-XL5-
(30 ng/ml) +
(30 ng/ml) +





UTF1 +
CHIR99021 (3 μM) +
CHIR99021 (3 μM)





pCMV6-XL4-
VPA + 5-Aza






DPPA4





20
VPA + 5-
pCMV6-XL5-
StemPro ™/hES
StemPro ™/hES
hES medium



Aza pre-
FoxD3 +
medium + ActivinA
medium + ActivinA




treated
pCMV6-XL5-
(30 ng/ml) +
(30 ng/ml) +





UTF1 +
CHIR99021 (3 μM) +
CHIR99021 (3 μM)





pCMV6-XL4-
VPA + 5-Aza






DPPA4





21
VPA + 5-
GFP
StemPro ™/hES
StemPro ™/hES
hES medium



Aza pre-

medium + ActivinA
medium + ActivinA




treated

(30 ng/ml) +
(30 ng/ml) +






CHIR99021 (3 μM)
CHIR99021 (3 μM)






+/or− VPA + 5-Aza









In order to characterize subpopulations of cells after transfection, live staining, immunohistochemistry, and AP staining was performed to follow the change in pluripotent markers. Cells transfected with either Oct4/UTF1/MBD2, Oct4/Dppa4/MBD2, FoxD3/Dppa4/MBD2, Oct4/FoxD3/Dppa4, or Sox2/FoxD3/UTF1 showed positive colonies for TRA1-60, TRA1-81, and SSEA4. This observation indicated that MBD2 generally had no effect by itself on reprogramming towards pluripotent-like cells, except in the case of Oct4/FoxD3/MBD2 transfection. Colonies started to form on Day 7 and continued to form until Day 14 (FIG. 18) (the end of the study period). These colonies were positive for AP as well.


These results were confirmed by RT-PCR analysis showing up-regulation of Oct4 expression as shown in Table 35. Relative expression for SOX2 was also slightly up-regulation in Day 7 after transfecting cells with Oct4/Foxd3/MBD2. There is also a trend of Sox2 up-regulation following transfection with Oct4/Sox2/Foxd3 and Oct4/Foxd3/Utf1.









TABLE 35







Relative expression of Pluripotent genes after


transfecting ADSCs with various combinations


of vectors as described in Table 34.











Endogenous



OCT4
SOX2












Rel.
Std.
Rel.
Std.



Exp.
Dev.
Exp.
Dev.














 #1 Day 7, Oct4/Sox2/MBD2
25.20
1.89
3.89
2.06


 #2 Day 7, Oct4/Foxd3/MBD2
11.28
0.13
18.79
0.03


 #3 Day 7, Oct4/Utf1/MBD2
2.01
0.20
2.93
1.73


 #4 Day 7, Oct4/Dppa4/MBD2
9.68
1.36
1.18
0.15


 #5 Day 7, Sox2/Foxd3/MBD2
1.06
0.55
2.68
2.90


 #6 Day 7, Sox2/Utf1/MBD2
0.66
0.10
3.36
0.68


 #7 Day 7, Sox2/Dppa4/MBD2
0.74
0.00
5.03
4.73


 #8 Day 7, Foxd3/Utf1/MBD2
1.31
0.61
4.15
2.92


 #9 Day 7, Foxd3/Dppa4/MBD2
0.63
0.02
3.90
2.17


#10 Day 7, Utf1/Dppa4/MBD2
0.96
0.04
4.97
1.92


#11 Day 7, Oct4/Sox2/Foxd3
48.17
1.89
7.68
1.79


#12 Day 7, Oct4/Sox2/Utf1
48.97
6.93
3.71
0.39


#13 Day 7, Oct4/Sox2/Dppa4
32.40
2.74
4.61
2.37


#14 Day 7, Oct4/Foxd3/Utf1
4.30
0.91
9.83
3.03


#15 Day 7, Oct4/Foxd3/Dppa4
4.21
0.11
4.57
0.85


#16 Day 7, Oct4/Utf1/Dppa4
10.29
3.70
3.53
1.63


#17 Day 7, Sox2/Foxd3/Utf1
1.42
0.83
3.32
2.12


#18 Day 7, Sox2/Foxd3/Dppa4
1.19
0.14
3.37
1.23


#19 Day 7, Sox2/Utf1/Dppa4
1.34
0.09
2.33
2.91


#20 Day 7, Foxd3/Utf1/Dppa4
0.72
0.07
2.45
0.27


#21 Day 7, GFP (−VPA/−5aza)
1.02
0.29
1.01
0.17


#22 Day 7, Untransf. ADSC
1.25
N/A
0.30
N/A


(−VPA/−5aza)






#23 Day 7, GFP (+VPA/+5aza)
1.01
0.20
1.87
2.23


#24 Day 7, Untransf. ADSC
1.45
N/A
0.27
N/A


(+VPA/+5aza)














Reprogramming Efficiency of Defined Pluripotency Factors on HFF after Triple Transfection (One Transfection Every 3 Days)


HFF cells were cultured as described in Example I with the exception of the concentrations of VPA and 5-AZA that were respectively 2 mM and 2.5 μM. Cells were transfected using the Nucleofector™® II Device (Lonza) following procedure described in Example II with the exception of the DNA quantity: 1 μg of each of the 3 plasmids DNA was used. The cells that had been pre-treated with VPA and 5-Aza and the untreated cells were both transfected with a mix of pCMV-Oct4nuc-IRES2-Sox2nuc, pCMV-Klf4nuc-IRES2-Cmycnuc or pCMV-Nanognuc-IRES2-Lin28. Following transfection the cells were plated in the fibroblasts medium described in Example I, supplemented with or without VPA and 5-AZA on Matrigel™ (BDBiosciences) coated on 6-well plates and incubated at 37° C., 5% CO2. On Day 1 and 2, media was changed to 100% mTeSR1 medium (StemCell Technologies) supplemented with or without VPA and 5-AZA. On Day 3 and Day 6, cells from each condition were detached by incubation in TrypLE™ for 5 min, counted and centrifuged. Cells were retransfected as above and plated on Matrigel™ coated plates in mTeSR1 medium supplemented with or without VPA and 5-AZA. Media was changed daily as described for day 1 and 2. Medium was supplemented in Y27632 (Stemgent, 10 μM) from day 7 to day 14 to promote viability and clonal expansion of potential reprogrammed cells. Cells were analysed at Day 20 using the Alkaline Phosphatase Detection Kit (Millipore) and by immunohistochemistry analysis.


This analysis revealed that after three transfections, three clones were found to be positive for alkaline phosphatase activity and showed a rounded cell/colony morphology. Staining with antibodies against the embryonic stem (ES) cell markers SSEA-4 and TRA-1-81 confirmed that these clones were pluripotent-like (FIG. 19). Surrounding HFF cells were negative for these markers. These clones were obtained only in the condition that did not contain inhibitors (i.e.: VPA and 5-AZA). Unexpectedly, no clones were observed for the condition treated with these inhibitors.


Reprogramming of NSLCs into Pluripotency


NSLC and neuronal stem cells derived from BG-01, a human ES cell line that expresses markers that are characteristic of ES cells including SSEA-3, SSEA-4, TRA-1-60, TRA-1-81, and OCT-3/4, were reprogrammed into pluripotency. BG-01 cells had previously been cultured in conditions to induce the differentiation towards neural stem cells as described by Chambers S M et al., 2009. NSLCs and BG-01-NSC were cultured in proliferation medium supplemented with FGF (20 ng/ml) and EGF (20 ng/ml). NSLCs and BG-01-NSCs were transfected as previously described in Example II by two episomal vectors, pEF-Oct4nuc-IRES2-MBD2 (NC1) or pCMV-FoxD3-2A-Oct4-2A-Klf4 (F72). Following transfection cells were collected and plated onto uncoated petri-dishes in the presence of Proliferation medium and mTeSR1 medium (50:50) in proliferation conditions at 37° C., 5% CO2. After 48 hours, cells were re-transfected by the same plasmid and plated in 96-well plates coated with Matrigel™ and cultured in the presence of mTeSR1 medium supplemented by the small molecules BIX01294 (Stemgent, 2 μM) and BayK8644 (Stemgent, 2 μM) at 37° C., 5% O2 for 22 days. Live staining and immunohistochemistry were performed to characterize subpopulations of cells for pluripotency markers.


NSLCs and BG-01-NSCs were positively stained with SSEA-4 starting on Day 7 and maintained until 22 days in culture (the end of the study) (FIG. 20). Within ten days, cells that were morphologically similar to ESCs were observed and they expressed a wide panel of pluripotency markers, including SSEA-4, TRA1-81, Nanog and Oct4 (FIG. 20). This study identified another way to get pluripotent-like cells from somatic cells via Neural Stem-Like Cells (NSLCs). The utility of NSLCs could offer multiple advantages for reprogramming towards pluripotent-like cells. For example, obviating the requirement for tumorigenic genes like c-Myc reduces the risk of inducing cancerous cells. For neuroregenerative and neurodegenerative applications these cells could represent an invaluable source of cells to investigate furthermore human pluripotent cell induction, and also represent a potential source of cells for deriving patient-specific multipotent and pluripotent stem cells for modeling human disease.


Example XX

Teratoma Formation Assay in SCID Mice


Transplantation of human pluripotent stem cells (SC) into “severely compromised immuno-deficient” (SCID) mice leads to the formation of differentiated tumors comprising all three germ layers for pluripotent stem cells, resembling spontaneous human teratomas, and specialized tissue for multipotent stem cells. These assays are considered the standards in the literature for demonstrating differentiation potential of pluripotent stem cells and hold promise as a standard for assessing safety among SC-derived cell populations intended for therapeutic applications


After all appropriate animal approvals for the experiment has been obtained, 24 mice were purchased from Charles Rivers, and housed at MISPRO animal facility for one week without any experimentation for adaption to the new environment. One million human NSLCs, normal human neuroprogenitor cells (hNPCs), or human embryonic stem (ES) cells in 100 μl Phosphate buffered saline, calcium- and magnesium-free (CMF-PBS) were injected with a 21-G needle intramuscularly into the right hind limb of the 4-week-old male SCID-beige mice under anesthesia with Ketamine/xylazine (8 mice per group). Following injection, the syringe was aspirated up and down a couple of times in a culture dish containing medium to verify that the cells were injected and not stuck inside the syringe.


The mice were maintained for 12 weeks and monitored for clinical signs and any tumor growth regularly. Any specialized tissue or teratoma growth was monitored by external examination and an increase in the size of the muscle relative to the same muscle on the left hind limb. When a specialized tissue or teratoma was identified, the location and size of the growth was measured (using measuring calipers) and recorded. The specialized tissue or teratoma is usually first identified as a small growth of the muscle size compared to the left control muscle. Animals were monitored weekly until onset of any tumor growth, and daily after tumors appeared. After 12 weeks, the mice were sacrificed by CO2 euthanasia. Each entire animal was observed for any tumor growth anywhere on the animal, and the injected muscle and the comparable left muscle control were measured (with measuring calipers)(see results table below) and then removed and stored in 4% paraformaldehyde solution for histological analysis. The sizes of the muscles were as follows:















Left leg (control)
Right leg (treated)












Dorso-

Dorso-




ventral
Lateral
ventral
Lateral


Treatment
width
width
width
width





Human Embryonic
6.44 ± 0.11
5.03 ± 0.17
6.91 ± 0.15
 5.3 ± 0.14


Stem Cells






Human
6.60 ± 0.17
5.43 ± 0.15
7.01 ± 0.23
5.58 ± 0.13


Neuroprogenitor






Cells






Human NSLC
6.85 ± 0.2 
5.32 ± 0.14
6.86 ± 0.21
5.33 ± 0.11









Values represent the Average of 8 mice±the standard error


Measurement of the size of the muscles revealed that all the human embryonic stem cell injected muscles were bigger than the comparable left muscle controls, indicating teratoma growth in the ES cell injected muscles. About half of all the human neuroprogenitor cell injected muscles were bigger than the comparable left muscle controls, while the mice injected with NSLC did not show any difference between the muscles (treated with the cells or not). The mice injected with NSLC did not show any evidence of tumor or teratoma growth.


Example XXI: Transfection of ADSCs by Various Combinations of Genes and Treatment with Small Molecules for Reprogramming to Mesendoderm-Like Cells

Human Adipose-Derived Stem Cells (ADSCs) were purchased from Invitrogen and expanded in complete StemPro™ MSC serum-free medium (Invitrogen on CellStar™ coated flasks (diluted 1:100 in PBS containing Ca2+/Mg2+) at a cell density of 1×104 cells/cm2. ADSCs were cultured as described in complete StemPro medium (Invitrogen) and were pretreated for 2 days prior to transfection with VPA (1 mM) and 5-Aza (0.5 μM). Cells were transfected using the Nucleofector® 96-well Shuffle Device (Lonza) following the procedure described previously. The cells were transfected with various combinations of cDNA clones summarized in Table 36. After transfection, the cells were plated on 0.1% Gelatin-coated plates and incubated at 37° C., 5% CO2, 5% O2. Medium was changed every other day according to Table 37. Cells were analyzed at Day 10 by Quantitative Realtime PCR.









TABLE 36







Various combinations of plasmids with potential to transfect


the cells towards mesendoderm lineage.












Day −2 to





Day 0
Plasmids transfected at Day 01







 1
Untreated
FoxD3, Ngn3, Sox17, MBD2



 2

FoxD3, Eomes, Gata6, Mixl1, MBD2



 3
Pre-treated
Eomes, FoxD3, Gata6, Mixl1



 4
with
Eomes, FoxD3, Gata6, Sox17



 5
VPA & 5-Aza
Eomes, Gata6, Mixl1, T



 6

FoxD3, Gata4, Mixl1, Sox.17



 7

Gata4. Gata6, Mixl1 Oct4



 8

Gata4, Mixl1, Sox17, T



 9

Eomes, Mixl1, Sox17



10

Eomes, Gata6, Sox17



11

Gata4, Mixl1, Sox17



12

Gata6, Mixl1, Sox17



13

GFP








1where FoxD3 = pCMV6-XL5-FoxD3, MBD2 = pCMV6-AC-MBD2, T = pCMV6-XL5-T, Mixl1 = pCMV6-XL5-Mixl1, Sox17 = pCMV6-XL4-SOX17, Gata4 = pCMV6-XL5-Gata4, Eomes = pCMV6-XL4-Eomes, and Gata6 = pCMV6-XL6-Gata6, All clones were purchased from Grigene and prepared using the Endorree Plasmid Maxi Kit (Qiagen.).














TABLE 37







Medium composition from Day −2 to Day 20












Day -2 to 0
Day 1
Day 2 to day 3
Day 4 to day 7
Day 8 to day 10
Day 11 to day 20





StemPro
CDM II (50%) +
IMDM/F12 +
IMDM/F12 +
IMDM/F12 +
IMDM/F12 +


medium +
IMDM/F12
NEAA +
NEAA +
NEAA +
NEAA +


VPA + 5Aza
(50%) + NEAA +
ITS + HSA +
ITS + HSA +
ITS + HAS +
ITS + HSA +



ITS + HSA +
bFGF + EGF +
bFGF + EGF +
bFGF + EGF +
Exendin-4,



bFGF + EGF +
VPA + Activin
Activin A +
BMP4
nicotinamide



VPA + Activin
A + 5-Aza +
5-Aza + BMP4





A + 5-Aza
BMP4






2Supplements added to media at the following concentrations: Activin A (Peprotech, 30 ng/ml), HSA (Baxter, 0.5%), NEAA (Gibco, 1X), ITS (Gibco, 1X), EGF (Peprotech, 5 ng/ml), bFGF (Peprotech, 10 ng/ml), VPA (Stemgent, 1 mM), 5-Aza (Sigma, 0.5 uM), BMP4 (Peprotech, 10 ng/ml), Exendin-4 (50 ng/ml, Cat Nb: 24463, Anaspec), and Nicotinamide (10 mM final., Cat Nb: N5535, Sigma).







Cells were collected on Day 10 by detaching with TrypLE, followed by centrifugation at 80.times.g for 5 minutes. Supernatant was aspirated and the cell pellet was frozen at −86° C. until ready for RNA Isolation. RNA isolation and quantification was performed as previously described: RNA isolation and cDNA was prepared using the High Capacity cDNA RT kit (Applied Biosystems) as per the manufacturer's instructions with a final cDNA concentration of 2 ng/l. Real-time PCR was then performed for each gene of interest using the FAST PCR master mix (Applied Biosystems) and the Taqman® Gene Expression Assays (Applied Biosystems) listed below:

















Taqman ®
Forward
Reverse




Assay
Primer
Primer
Probe


Gene
ID
Sequence
Sequence
Sequence







GAPDH
Hs99999905_m1





(house-






keeper)









PPIA 
Hs99999904 m1





(house-






keeper)









FOXA2
Hs00232764_m1








SOX17
Hs00751752_s1








Endogen- 
Hs00610073_g1





ous T









GSC
Hs00418279_m1








CXCR4
Hs00607978_s1








GATA4
Hs00171403_m1








CER1
Hs00193796_m1








CDH1 
Hs01023894_m1





(E-






cadherin)









p63
Hs00978340_m1








SOX2
SOX2_1078-ANY
CCACCTACA
GACCACC
CTGGCAT




GCATGTCCT
GAACCCA
GGCTCTTG 




ACTC
TGGA
(SEQ ID




(SEQ ID 
(SEQ ID 
NO: 3)




NO: 1)
(NO: 2)









Ten days following the transfection with various combinations of Oct4, Sox17, FoxD3, T, MixI1, FoxA2, and MBD2 cDNA, analysis by RT-PCR to investigate the expression of Mesoendoderm and mesendoderm differentiation genes was performed. As summarized in Table 38, the Relative Expression of Sox17 and FoxA2, the markers for Endoderm and Mesoderm, was up-regulated noticeably in the FoxD3/MixI1/Ngn3/MBD2-transfected sample, while the expression of mesendoderm differentiation (ex. pancreatic lineage) genes are low indicating an early mesendoderm phenotype. Ten days following transfection, SOX17 is still highly expressed in the SOX17-transfected samples as compared to the untreated ADCs sample that represents leftover plasmid DNA (exogenous SOX17) that still remains 10 days post-transfection, and any endogenous SOX17 expression that may have been induced.









TABLE 38





Relative Expression of the mesendoderm genes FoxA2 and Sox17 after


transfecting ADSCs once with various gene combinations with potential to


reprogram cells into mesoendoderm-like cells.




















FoxA2
Sox17
Gata4
EndoPdX1
















Rel.
Std.
Rel.
Std.
Rel.
Std.
Rel.
Std.


Day 10
Exp.
Dev.
Exp.
Dev.
Exp.
Dev.
Exp.
Dev.





Foxd3/Sox17/Ngn3/
1.13
0.06
44.41
0.51
0.16
0.11
1.13
0.06


MBD2










Foxd3/Mixl1/Ngn3/
48.03
2.28
11.17
0.58
0.08
0.00
1.71
0.79


MBD2










Foxd3/Mixl1/Pdx1/
2.36
1.68
0.33
0.07
0.08
0.00
1.19
0.03


MBD2










Eomes/FoxD3/Gata6/
3.12
0.06
0.05
0.00
0.22
0.00
3.12
0.06


Mixl1










Eomes/FoxD3/Gata6/
2.64
0.01
137.24
12.01
0.71
0.70
2.64
0.01


Sox17










Eomes/Gata6/Mixl1/T
1.51
0.02
1.37
0.05
0.11
0.00
1.51
0.02


FoxD3/Gata4/Mixl1/
0.78
0.00
84.72
3.22
0.21
0.00
0.78
0.00


Sox17










Gata4/Gata6/Mixl1/
1.35
0.04
3.20
1.20
0.36
0.01
1.35
0.04


oct4










Gata/Mixl1/Sox17/T
1.20
0.21
228.37
5.78
1.48
0.93
0.41
0.00


Eomes/Mixl1/Sox17
1.72
1.41
139.00
7.36
0.20
0.01
0.76
0.05


Eomes/Gata6/Sox17
0.57
0.03
573.14
65.66
0.15
0.01
0.93
0.49


Gata4/Mixl1/Sox17
1.23
0.02
253.21
45.51
123.32
62.32
1.52
0.21


Gata6/Mixl1/Sox17
1.20
0.32
354.21
7.65
0.32
0.01
1.52
0.52


ADCs untransf.
0.49
0.02
0.93
0.19
0.49
0.48
0.49
0.02















Sox9
Ngn3
Nkx2.2
Insulin
















Rel.
Std.
Rel.
Std.
Rel.
Std.
Rel.
Std.


Day 10
Exp.
Dev.
Exp.
Dev.
Exp.
Dev.
Exp.
Dev.





Foxd3/Sox17/Ngn3/
0.98
0.05
1.13
0.06
1.13
0.06
1.13
0.06


MBD2










Foxd3/Mixl1/Ngn3/
0.97
0.08
1.13
0.04
1.51
0.51
1.13
0.04


MBD2










Foxd3/Mixl1/Pdx1/
1.24
0.08
1.19
0.03
1.19
0.03
1.66
0.63


MBD2










Eomes/FoxD3/Gata6/
1.87
0.02
3.12
0.06
3.12
0.06
3.12
0.06


Mix11










Eomes/FoxD3/Gata6/
1.73
0.02
2.64
0.01
2.64
0.01
2.64
0.01


Sox17










Eomees/Gata6/Mixl1/T
1.46
0.03
1.51
0.02
1.51
0.02
1.51
0.02


FoxD3/Gata4/Mixl1/
1.19
0.12
0.55
0.00
0.78
0.00
0.26
0.00


Sox17










Gata4/Gata6/Mixl1/
1.33
0.10
0.95
0.03
1.35
0.04
0.45
0.01


oct4










Gata/Mixl1/Sox17/T
0.62
0.02
0.29
0.00
0.41
0.00
0.14
0.00


Eomes/Mixl1/Sox17
1.21
0.03
0.54
0.04
0.76
0.05
0.44
0.28


Eomes/Gata6/Sox17
0.97
0.05
0.40
0.02
1.55
1.38
0.19
0.01


Gata4/Mixl1/Sox17
2.32
0.04
0.58
0.01
2.25
0.65
0.98
0.01


Gata6/Mixl1/Sox17
1.23
0.05
0.98
0.12
2.32
0.54
0.78
0.08


ADCs untransf.
1.16
0.08
0.34
0.01
0.73
0.33
0.16
0.01









Example XXII

Transfection of ADSCs by Various Combinations of Genes and Treatment with Small Molecules for Reprogramming to Pancreatic Progenitor-Like Cells


Reprogramming ADSCs into Pancreatic Progenitor-Like Cells: Human ADSCs were purchased from Invitrogen and expanded in complete StemPro™ MSC serum-free medium (Invitrogen on CellStart™ coated flasks (diluted 1:100 in PBS containing Ca2+/Mg2+) at a cell density of 1×104 cells/cm2. Cells were transfected using the Nucleofector® 96-well Shuttle® Device (Lonza) following the procedure described previously. The cells were transfected with various combinations of cDNA clones as described in Table 4. After transfection, the cells were plated on Fibronectin-coated collagen gels and incubated at 37° C., 5% CO2, 5% O2. Plates were coated with fibronectin (BD Biosciences) at a concentration of 1.9 .mu.g/well. Media was changed every other day according to Table 39. Following transfection cells will be cultured using high concentration of activin A (50 ng/ml) and BMP (30 ng/ml) to push the reprogramming towards the mesendoderm. To initiate the transition of definitive endoderm to Primitive Gut tube, the medium was supplemented by FGF 10 (Peprotech) and cyclopamine (Stemgent). Thereafter, the gut tube was exposed to Retinoic acid, cyclopamine, and FGF10. In order to stimulate the expression of NGN3 and NKx2.2, the medium was supplemented by exendin 4 and hepatocyte growth factor.









TABLE 39







Plasmids and media composition from Day 0 to Day 20


Media Composition1













Plasmids transfected at
Day −2 to




Day 15 and


Day 0
Day 0
Day 1 to 3
Day 3 to 6
Day 7 to 9
Day 10 to 14
over





1-MBD2/Oct4/Sox17
StemPro
StemPro/
RPMI, B27
DMEM free
DMEM free
DMEM/F12


2-MBD2/Oct4/Pdx1
medium
RPMI with
1%. With
of
of
nicotinamide


3-Sox17/PDX1/Ngn3

17.5 mM
1 mM
glucose/F12
glucose/F12
(10 nM), +


4-MBD2/Ngn3/Sox17

glucose.
glutamine,
(1:1) + B27
(1:1) + B27
Hepatocyte


5-MBD2/FoxA2/NGN3

ITS, 1 mM
1% HSA +
1%, 1%
1%, 1%
growth factor


6-MBD2/PDX1/Ngn3

glutamine,
FGF10
HSA,
HSA,
(20 ng/ml &


7-Oct4/Sox17/Pdx1/Ngn3

1% HSA +
(10 ng/m/)
Retinoic acid
Exendin4
exendin4


8-Oct4/Pdx1/Ngn3

Activin A

(2 nM)
(10 nM) &
(10 nM)


9-FoxA2/Pdx1/Sox17

(50 ng/ml) +

Cyclopanine
Cyclopamine



10-GFP

BMP4

(10 mM),
(10 mM),





(30 ng/ml)

FGF10








(10 ng/ml)






1Supplements added to media at the following concentrations: ActivinA (Peprotech, 30 ng/ml), HSA (Baxter. 0.5%). NEAA (Gibco, 1X), ITS (Gibco, 1X), B27 (Gibco, 1%), bFGF (Peprotech, 10 ng/ml),,, VPA (Stemgent, 1 mM), 5-Aza (Sigma, 0.5 uM), Retinoic Acid (Sigma, 2 uM), FGF10 (Peprotech, 50 ng/ml), Cyclopamine (Stemgent, 2.5 uM), Exendin-4 (50 ng/ml, Cat Nb: 24463, Anaspec).







Cells were collected on Day 20 to be analyzed by Quatitative Real-time PCR and RNA isolation and quantification was performed as described previously. cDNA was prepared and quantitative real-time PCR was performed with the following Taqman® Gene Expression Assays (Applied Biosystems) used:

















Taqman ®
Forward
Reverse
Probe



Assay
Primer
Primer
Se-


Gene
ID
Sequence
Sequence
quence







GAPDH
Hs99999905_m1





(house-






keeper)









PPIA
Hs99999904_m1





(house-






keeper)









FOXA2
Hs00232764_m1








SOX17
Hs00751752_s1








GATA4
Hs00171403_m1








Endogen-
PDX1_1201
GGCCCTCTTTT
GTAGGAGGGC
ACAGCCA


ous

AGTGATACTG
AGGGATGTG
CAAACAA


PDX1

GATT
SEQ ID
CG




(SEQ ID
(NO: 5)
(SEQ ID




NO: 4)

NO: 6)





SOX9
Hs00165814_m1








NGN3
Hs01875204_s1








NKK2-2
Hs00159616_m1








PAX4
Hs00173014_m1








INS
Hs02741908_m1








CXCR4
Hs00607978_s1









RT-PCR was used to evaluate the expression of pancreatic-related genes from cells after 20 days post-transfection with different combinations of genes (Table 40). This study revealed that FoxA2 (13.12±0.06), Nkx2.2 (23.12±0.06) and Gata4 (5.21±2.36), markers for Endoderm and Mesoderm, were up-regulated for the Sox17/Ngn3/Pdx1-transfected sample as compared to the GFP mock-transfected control sample. Addition of Oct4 to this gene combination increased Gata4 expression as well as Sox17 and endogenous Pdx1 expression, while both gene combinations expressed insulin indicative of islet .beta.-like cells.









TABLE 40





Relative Expression of mesendoderm, pancreatic progenitor, and islet


β-cell genes after transfecting ADSCs once with various gene combinations with


potential to reprogram cells into pancreatic progenitor like cells (including


mesoendoderm-like cells and islet β-like cells).




















FoxA2
Sox17
Gata4
EndoPdx1
















Rel.
Std.
Rel.
Std.
Rel.
Std.
Rel.
Std.


Day 20
Exp.
Dev.
Exp.
Dev.
Exp.
Dev.
Exp.
Dev.





MBD2/Oct4/Sox17
1.13
0.06
62.97
27.53
0.16
0.11
1.13
0.06


MBD2/Oct4/Pdx1
4.72
3.36
10.59
0.29
0.08
0.00
15.91
0.03


MBD2/FoxA2/PDX1
43.16
6.34
0.33
0.07
0.08
0.00
14.69
2.26


Sox17/PDX1/Ngn3
13.12
0.06
72.37
27.67
5.21
2.38
32.08
14.55


MBD2/Ngn3/Sox17
2.64
0.01
137.24
12.01
0.71
0.07
2.64
0.01


MBD2/FoxA2/NGN3
72.59
0.02
10.22
0.14
0.11
0.00
1.51
0.02


MBD2/PDX1/Ngn3
0.78
0.00
42.36
1.61
0.21
0.00
20.07
6.32


Oct4/Sox17/Pdx1/Ngn3
11.35
0.04
139.00
7.36
34.53
17.18
116.68
41.77


Oct4/Pdx1/Ngn3
1.72
1.41
1.33
0.36
9.85
5.24
67.42
34.45


FoxA2/Pdx1/Sox17
100.57
0.03
143.29
16.42
0.15
0.01
13.19
4.24


ADSC untransf.
0.04
0.01
0.21
0.08
0.08
0.00
1.16
0.01















Sox9
Ngn3
Nkx2.2
Insulin
















Rel.
Std.
Rel.
Std.
Rel.
Std.
Rel.
Std.


Day 20
Exp.
Dev.
Exp.
Dev.
Exp.
Dev.
Exp.
Dev.





MBD2/Oct4/Sox17
0.98
0.05
1.13
0.06
1.13
0.06
1.13
0.06


MBD2/Oct4/Pdx1
0.97
0.08
1.13
0.04
1.51
0.51
1.13
0.04


MBD2/FoxA2/PDX1
1.24
0.08
1.19
0.03
1.19
0.03
1.66
0.63


Sox17/PDX1/Ngn3
1.87
0.02
97.04
18.77
23.12
0.06
6.12
0.06


MBD2/Ngn3/Sox17
1.73
0.02
12.58
3.08
2.64
0.01
2.64
0.01


MBD2/FoxA2/NGN3
1.46
0.03
20.33
5.17
1.51
0.02
1.51
0.02


MBD2/PDX1/Ngn3
1.19
0.12
59.24
14.07
0.78
0.00
0.26
0.00


Oct4/Sox17/Pdx1/Ngn3
1.33
0.10
121.30
4.76
5.35
0.04
0.07
0.06


Oct4/Pdx1/Ngn3
1.21
0.03
34.50
2.38
0.76
0.05
1.44
0.28


FoxA2/Pdx1/Sox17
0.97
0.05
0.40
0.02
1.55
1.38
0.19
0.01


ADSC untransf.
1.02
0.13
1.16
0.01
1.71
0.77
1.16
0.01









Next immunohistochemical analysis was performed to evaluate protein expression in cells transfecting with the different gene combinations. On day 20, the cells were immunostained and examined using the Cellomics™ ArrayScan VTI. Cells were fixed with a 4% formaldehyde/PBS solution for 10 min at room temperature and subsequently permeabilized for 5 min with 0.1% Triton X-100 in 4% formaldehyde/PBS. After two brief washes with PBS, unspecific antibody binding was blocked by a 30 min incubation with 5% normal goat serum in PBS. Then primary antibodies were added in 5% normal goat serum/PBS as follows: Mouse anti-Gata4 (1:100, BD) and mouse anti-FoxA2 (1:200, BD), a marker for endoderm layer. After a 2 h incubation the cells were washed 4 times for 5 min each with 0.1% Tween/PBS. Appropriate fluorescence-tagged secondary antibody was used for visualization; Goat anti-mouse 546 (1:200, invitrogen) prepared in 5% normal goat serum/PBS was used. After incubation for one hour, cells were washed in 0.1% Tween/PBS three times for 5 min each. The DNA stain Hoechst33342 (Invitrogen) was used as a marker of nuclei (dilution 1:5000 in PBS, 10 min incubation). Fluorescence images were taken with the Cellomics™ ArrayScan HCS Reader microscopy system. To determine an estimate of the percentage of cells adopting endodermal phenotypes, random fields were selected and for each field the total number of cells (as determined by counting Hoechst stained nuclei) and the total number of cells positive were determined. The cells in the Sox17/Pdx1/Ngn3 and Oct4/Sox17/Pdx1/Ngn3 transfected groups significantly expressed both Gata4 and FoxA2: the number of positive cells was enhanced in the presence of Oct4 as shown in Table 41.









TABLE 41







Percentage of positive cells for Gata4 and FoxA2 at day 20


after transfection of ADSCs with different expression vectors.









Day 20











cell count
Gata4%
FoxA2%













GFP
4825
0.21
0.23


untransfected
18646
0.01
0.03


MBD2/Oct4/Sox17
12563
0.17
0.35


MBD2/Oct4/Pdx1
13481
2.61
0.2


MBD2/FoxA2/Pdx1
13418
0.17
0.16


Sox17/Pdx1/Ngn3
15306
11.15
16.9


MBD2/Ngn3/Sox17
19867
1.32
0.31


MBD2/Ngn3/FoxA2
14984
0.33
0.42


MBD2/Ngn3/Pdx1
18613
0.31
10.29


Oct4/Sox17/Pdx1/Ngn3
19515
21.07
30.02


Oct4/Pdx1/Ngn3
6103
0.28
0.49


FoxA2/Pdx1/Sox17
10001
0.65
8.82





After transfection cells were cultured in IMDM/F12 supplemented with different type of growth factors and small molecules (as described in Table 4). The percentage of immunopositive cells was determined by the Cellomics ™ ArrayScan HCS Reader.






Transfection with Pdx1 Ngn3, Sox17 (and Oct4) increased significantly selected pancreas-related genes (Pdx1, Ngn3, NKX2.2, Gata4, FoxA2) indicating a transition from endoderm to a pancreatic fate during this time period. To evaluate pancreatic hormone secretion, pancreatic-like cells were cultured for 50 days, during which time the medium was replaced with fresh medium every two days. We then assayed the supernatant for insulin in the conditioned medium by antigen-capture ELISA kit (Abnova, KA0921) at different time points (day 25 and day 55) and compared to the release of insulin in control cultures. According to the manufacturer's instructions, briefly, 96-well ELISA immunoplates were coated with Anti-insulin (CatNb #) diluted 1/1000 in carbonate buffer (pH 9.7) and incubated at 4° C. overnight. The following day, all wells were washed with TBS-Tween 0.5% before incubation with Block/Sample buffer 1× at room temperature for one hour without shaking. After blocking, standards and samples were added to the plates and incubated and shaken (450±100 rpm) for 2 h at room temperature. Subsequently, after washing with TBS-Tween wash buffer, plates were incubated for 2 h with Anti-Human insulin pAb (1:500 dilution in Block & Sample 1× Buffer) at 4° C. After incubation, plates were washed five times with TBS-Tween 0.5% wash buffer and 100 μl of standard and sample was added in the plates precoated with anti-insulin and incubated for 1 hour at room temperature with shaking (450±100 rpm). Then, plates were washed five times with TBS-Tween 0.5% wash buffer and 100 μl of TMB One Solution was added to each well. Following 10 minutes incubation at room temperature with shaking (450±100 rpm) for the insulin plate, a blue color formed in the wells. After stopping the reaction by adding 100 μl of 1N hydrochloric acid, the absorbance was read at 450 nm on a microplate reader (Synergy 4) within 30 minutes of stopping the reactions. Concentration of released insulin in the supernatants was determined according to the standard curves. ELISA results revealed that insulin was released from cells transfected with Oct4/Sox17/Pdx1/Ngn3, and to a lesser extent in the other groups as summarized in Table 42.









TABLE 42







Quantification of insulin release by pancreatic


progenitor/β-like cells that had been cultured for


25 and 55 days after transfecting ADSCs.










Concentration
Concentration



of insulin
of insulin



(μlU/ml)
(μlU/ml)


Insulin release
at day 25
at day 50












MBD2/Oct4/Sox17
0.05
0.07


MBD2/Oct4//Pdx1
1.02
1.87


MBD2/FoxA2/PDX1
1.13
1.11


Sox17/PDX1/Ngn3
2.3
4.32


MBD2/Npt3/Sox17
0.07
1.03


MBD2/FoxA2/NGN3
0.09
1.30


MBD2/PDX1/Ngn3
1.23
1.03


Oct4/Sox17/Pdx1/Ngn3
4.32
7.85


Oct4/Pdx1/Ngn3
2.45
3.21


FoxA2/Pdx1/Sox17
1.02
1.23


ADSC untransfected
0.01
0.03





Insulin release into the medium, at days 25 and 55, was measured by antigen-capture ELISA (Abnova).






In addition to increase of gene expression, the transfecting cells were able to release insulin even in low concentrations. Generating reprogrammed .beta.-like cell lines that can locally deliver insulin could be used as a method to treat and allow functional recovery from diabetes.


Example XXIII: Transfection of ADSCs by Various Combinations of Genes and Treatment with Different Small Molecules for Reprogramming to Cardiac Progenitor Like Cells

ADSCs were cultured in StemPro™ MSC serum-free medium (Invitrogen) as previously described, and then transfected with different combinations of cDNA clones as described in Table 43 using the Nucleofectora 96-well Shuttle® Device (Lonza) as described previously. After transfection, the cells were plated on Matrigel-coated plates and incubated at 37° C., 5% CO2, 5% O2. Medium was changed every other day according to Table 44. Cells were analyzed at Day 20 by Quantitative Real-time PCR and by Immunohistochemistry.









TABLE 43







Various combinations of plasmids with potential to transfect


the cells towards Cardiac Progenitor-like cells lineage.












Day −3 to





Day 0
Plasmids transfected at Day 01







 1
Pre-treated
T, FoxD3, Sox17, Mesp1



 2
with
Foxd3, Sox17, Mesp1, Nkx2.5



 3
VPA & 5-Aza
Foxd3, Tbx5, Baf, Nkx2.5



 4

Foxd3, T, Mesp1, Gata6



 5

Sox17, Tbx5, Baf, Nkx2.5



 6

Foxd3, T, Mesp1, Gata4



 7

T, Tbx5, Baf, Nkx2.5



 8

Foxd3, T, Mesp1, Tbx5



 9

Mesp1, Tbx5, Baf, Nkx2.5



10

Foxd3, T, Mesp1, Tbx5



11

Foxd3, Sox17, Mesp1, Gata6



12

Foxd3, Sox17, Mesp1, Gata4



13

FoxD3, Sox17, Mesp1, Tbx5





GFP

















TABLE 44







media composition from day −3 to day 20.


Media Composition1











Day −3 to 0
Day 1
Day 2 to Day 3
Day 4 to Day 6
Day 7 to Day 20





StemPRo
IMDM/F12
IMDM/F12 +
IMDM/F12 +
IMDM/F12 +


medium +
(50%) + NEAA
NEAA + ITS +
NEAA + ITS +
NEAA + ITS +


VPA + 5-Aza
(1X) + ITS +
HSA + FGF8
HSA + FGF8
HSA + FGF



HAS (5 mg/ml) +
(10 ng/ml) +
(10 ng/ml) +
(50 ng/ml) +



VPA + Activin
5Aza + VPA +
FGF (50 ng/ml) +
BMP4 (50 ng/ml)



A (30 ng/ml) +
CHIR99021
5Aza + CHIR99021




CHIR99021
(2 nM) + Wnt11
(2 nM + Wnt11




(2 nM)
(50 ng/ml) +
(50 ng/ml) +





BMP4 (50 ng/ml)
BMP4 (50 ng/ml)






1Supplements added to media at the following concentration: Activin A (Peprotech, 30 ng/ml), HSA (Baxter, 0.5%), NEAA (Gibco, 1X), ITS (Gibco IX) FGF8 (Peprotech, 10 ng/ml), bFGP (Peprotech, 50 ng/ml), CHIR99021 (Stemgent, 2 uM), VPA (Stemgent, 1 mM), 5-Aza (Sigma, 0.5 uM), BMP4 (Peprotech, 50 ng/ml)







Cells were collected on Day 20 by detaching with TrypLE, followed by centrifugation at 80.times.g for 5 minutes. Supernatant was aspirated and the cell pellet was frozen at −86° C. until ready for RNA Isolation. RNA isolation, cDNA preparation and quantitative real-time PCR was performed as previously described.


Gene expression analysis on day 20 following transfection revealed an increased of several Mesoderm markers. Interestingly, three markers for cardiac progenitors cells (Nkx2.5, MyoCD, Tbx5) and three markers of early mesoderm lineage (Gata4, Mesp1, and Meox1) were highly increased after transfecting cells with brachyury (T)/Tbx5/Nkx2.2/Mesp1. Similarly, expression of Mesp1, Tbx5, Nkx2.5, MyoCd, and Meox1 was increased after transfecting the cells with Foxd3/Sox17/Mesp1/Tbx5; while expression of Mesp1, Gata4, Nkx2.5, MyoCd, and Meox1 was increased after transfecting with Foxd3/Sox17/Mesp1/Nkx2.5 and Foxd3/T/Mesp1/Gata4. As shown in Table 45, the Relative Expression of mesoderm and cardiac progenitor cells markers are especially up-regulated in the combination of Mesp1/Tbx5/Nkx2.5/T, Foxd3/Sox17/Mesp1/Nkx2.5 or/Tbx5, and Foxd3/T/Mesp1/Gata4. In general, transfection of ADSCs with a combination Mesp1, FoxD3, Tbx5, Brachyury, Nkx2.5, Sox17 and/or Gata4 increased the expression of mesoderm and cardiac progenitors markers.


In addition, samples were collected at day 20 to evaluate the nature of reprogrammed cells by analyzing the expression of mesoderm markers using Immunohistochemistry analysis for Brachyury and Nkx2.5 according to the methods previously described. Brachyury was significantly expressed in the Mesp1/Tbbx5/Nkx2.5/T, Foxd3/Sox17/Mesp1/Nkx2.5 or/Tbx5, and Foxd3/T/Mesp1/Gata4 groups, while all groups expressed Nkx2.5. These cells could represent and invaluable source of cells to investigate furthermore human cardiac cells and also represent a potential source of cells for deriving patient-specific multipotent stem cells for modeling, regenerating, or treating human cardiovascular diseases.









TABLE 45







Relative Expression Gata4, Mesp1, Tbx5, Nkx 2.5, MyoCD, and Meox1,


after transfecting ADCs once with various gene combinations with potential to


reprogram ADSCs cells into Cardiac Progenitor-like cells.














Gata4
MESP1
TBX5
NKX2.5
MYOCD
MEOX1




















Rel.
Std.
Rel.
Std.
Rel.
Std.
Rel.
Std.
Rel.
Std.
Rel.
Std.


Day 20
Exp.
Dev.
Exp.
Dev.
Exp.
Dev.
Exp.
Dev.
Exp.
Dev.
Exp.
Dev.






















GFP-transfected ADCs
1.01
0.16
1.01
0.19
1.00
0.05
1.02
0.27
1.00
0.07
1.02
0.27


Foxd3/Sox17/T/Mesp1
0.02
0.01
6.70
1.18
1.52
0.12
0.94
0.07
1.98
0.13
1.37
0.10


Foxd3/Tbx5/Baf60c/Nkx2.5
0.08
0.03
1.86
0.20
1.59
0.08
24.94
2.97
2.11
0.11
1.17
0.15


Sox17/Tbx5/Baf60c/Nkx2.5
0.62
0.09
3.89
0.10
1.45
0.00
41.72
2.35
0.90
0.02
1.19
0.02


T/Tbx5/Baf6oc/Nkx2.5
0.93
0.12
7.59
0.61
1.76
0.12
27.54
2.36
0.94
0.05
1.23
0.19


Mesp1/Tbx5/Baf60c/Nkx2.5
1.12
0.13
31.55
0.91
4.26
0.07
154.76
3.50
1.54
0.06
5.96
1.60


Foxd3/Sox17/Mesp1/Gata6
0.71
0.02
19.59
0.76
1.77
0.15
31.38
1.23
1.44
0.09
3.30
0.68


Foxd3/Sox17/Mesp1/Gata4
0.02
0.00
2.66
0.44
1.45
0.24
0.81
0.24
0.75
0.03
0.65
0.17


Foxd3/Sox17/Mesp1/Tbx5
1.75
3.18
138.28
2.20
15.61
0.87
45.20
14.88
11.06
1.02
4.36
3.45


Foxd3/Sox17/Mesp1/Nkx2.5
17.76
4.34
22.03
0.08
3.03
0.01
95.73
11.68
10.25
2.32
10.54
2.39


Foxd3/T/Mesp1/Gata6
27.10
6.47
3.59
0.42
2.58
0.29
5.11
0.85
21.03
1.02
12.99
1.31


Foxd3/T/Mesp1/Gata4
15.87
6.13
50.20
6.41
2.37
0.18
55.43
6.95
19.86
2.10
4.32
0.15


Foxd3/T/Mesp1/Tbx5
0.83
0.02
8.20
0.48
2.40
0.37
29.25
1.28
0.76
0.08
1.40
0.46


Foxd3/T/Mesp1/Nkx2.5
19.79
2.03
111.35
15.32
2.56
0.35
32.52
12.53
0.29
0.03
24.70
3.12


T/Tbx5/Nkx2.5/Mesp1
18.46
1.06
103.09
22.28
102.53
15.02
96.32
16.60
15.32
5.01
15.73
2.83









Example XXIV: Reprogramming Human ADSCs to Pluripotent-Like Stem Cells (PLSC)

Based on previous results in Example XIX, the highest reprogramming efficiency was observed using pEF-Rex1-EF-Oct4-2A-Klf4-2A-RFP (NF10) and pCMV-Sall4-2A-Oct4-2A-Klf4-2A-Nanog (S71). ADSCs (Invitrogen Corporation) were cultured in cell culture flasks with complete StemPro-43 medium (Invitrogen) at 37° C., 5% CO2 and the medium was changed 3 times per week. After 3 days in culture cells (passage 5) were trypsinized and counted to be transfected. Cells were transiently transfected with one plasmid: pCMV-Sall4-2A-Oct4-2A-Klf4-2A-Nanog or Rex1-EF-Oct4-2A-Klf4-2A-RFP (2 .mu.g) using nucleofector as described in Example II. Following the transfection cells were cultured in 6-well plates in suspension with 50:50 ratio of adipocyte complete medium (StemPro-43) and embryonic stem cell medium (mTesR1). After two days in culture, cells were re-transfected with the same plasmids listed above and cells were plated in 96 well-plates coated with Matrigel (BD Biosciences) in the presence of mTesR complete medium supplemented with thiazovivin (0.5 μM), an ALK-5 inhibitor (SB 341542, Stemgent, 2 μM), and inhibitor of MEK (PD0325901, Stemgent, 0.5 μM). Medium was changed every day and cells were cultured for 22 days at 37° C., 5% CO2, 5% O2. Alkaline Phosphatase Detection Kit (AP, Millipore) and immunohistochemistry were performed to analyze the expression of pluripotency markers. ALP staining was performed using AP detection kit (Millipore) according to manufacturer's instructions. Colonies emerged around Day 15 and maintained in culture up to the end of the study period (up to 2 months) with a stable morphology. Live staining showed that these colonies express typical pluripotency markers, including SSEA-4 and TRA1-81, TRA1-60, Oct4, Nanog, Sox2, and E-cadherin (FIG. 25). Further analysis of these colonies showed that the colonies present as well other ESC markers such as alkaline phosphatase (ALP, not shown). Supplementing the small molecules PD0325901 and SB431542 with the KSOR medium treated cultures, a 6 fold improvement in efficiency over the conventional method was obtained following the transfection of ADSCs with pCMV-pEF-Rex1-EF-Oct4-2A-Klf4-2A-RFP. This pattern did not change up to the 2 month culture period and same positive colonies were observed after transfecting ADSCs with Sall14-2A-Oct4-2A-Klf4-Nanog (not shown). The observation over time showed that the phenotype of these colonies moves from an early SSEA+ phenotype to a late Oct4+/Sox2+/Nanog+ phenotype, which is closer to the final reprogrammed state. Following the reprogramming of ADSCs cells to Pluripotent-like cells using the two vectors pEF-Rex 1-EF-Oct4-2A-Klf4-2A-RFP or pCMV-Sall4-2A-Oct4-2A-Klf4-2A-Nanog, a study was performed to examine the differentiation capacity of these cells. Beginning on Day 30, reprogrammed cells were cultured in conditions in order to stimulate the differentiation to embryonic bodies (EBs). These differentiation conditions consisted of placing the cells on uncoated petri-dish in EB Differentiation medium consisting of KSOR medium with FBS 5% without FGF for 45 days. To further differentiate the embryonic bodies, Day 30 EBs were cultured in three different media in order to drive cell differentiation to either ectoderm, mesoderm or endoderm lineages in CDM medium consisting of DMEMIF12 supplemented with 2 mM Glutamine, 0.11 mM 2-mercaptoethanol, 1 mM nonessential amino acids, and 0.5 mg/ml HAS (Yao S et al., PNAS 2006). This CDM medium was further supplemented with different concentrations of cytokines to induce the differentiation towards the three lineages. Ectoderm media consisted of N2/B27-CDM in the presence of Noggin (100 ng/ml) and after 10 days NGF (20 ng/ml) was added to the medium and the cells were grown on laminin-coated plates. Endoderm media consisted of N211327 CDM with the presence of Activin A (100 ng/ml) and the cells were grown on Gelatin-coated plates. Mesoderm media consisted of N2/B28-CDM in the presence of Activin A (50 ng/ml) and BMP-4 (50 ng/ml) and the cells were grown on Gelatin-coated plates. Cells that had originally been reprogrammed with pCMV-Sall4-2A-Oct4-2A-Klf4-2A-Nanog differentiated better than cells originally reprogrammed with pEF-Rex1-EF-Oct4-2A-Klf4-2A-RFP which detached from the coated plates. Visual observation of the differentiation of the cells into the three lineages was performed by RT-PCR and Cellomics using the Ectoderm markers (Nestin, GFAP, Beta-tubulin, Sox2), Endoderm markers (GATA4, Brachyury) and Mesoderm markers (GATA4, Brachyury, and Nkx2.5). RT-PCR analysis revealed the Identification of endoderm, mesoderm and ectoderm markers for all 3 embryonic germ layers (Table 46).









TABLE 46





RT-PCR analysis of three embryonic germ layers following the


differentiation of PLSCs.




















CDH1
ASCL1





(E-cadherin)
(MASH1)
TP63
NOTCH1
















Rel.
Std.
Rel.
Std.
Rel.
Std.
Rel.
Std.


A-Ectoderm
Exp.
Dev.
Exp.
Dev.
Exp.
Dev.
Exp.
Dev.





S71-transf. ADSC
59180.71
2036.56
70.28
12.35
123114.61
17452.81
5.47
0.19


NF10-transf. ADSC
77944.51
1747.33
48.20
3.27
166929.89
6007.52
6.47
0.12


Mel-2 Undifferentiated P17
84021.24
178.93
9.80
4.30
10.72
2.96
5.90
0.48


ADSC Untreated Ctrl
1.20
0.93
1.00
0.00
1.00
0.00
1.00
0.03













SOX17
CDX2











B-Endoderm
Rel. Exp.
Std. Dev.
Rel. Exp.
Std. Dev.





S71-transf. ADSC
9.39
2.21
37.77
5.11


NF10-transf. ADSC
2.33
0.94
21.05
18.59


Mel-2 Undifferentiated P17
0.77
0.00
4.34
5.05


ADSC Untreated Ctrl
1.00
0.00
1.00
0.00














FOXA2
GSC
CXCR4













C-Mesoderm
Rel. Exp.
Std. Dev.
Rel. Exp.
Std. Dev.
Rel. Exp.
Std. Dev.





S71-transf. ADSC
1193.70
51.50
0.85
0.11
512.33
45.62


NF10-transf. ADSC
1662.76
160.28
0.57
0.01
352.65
39.26


Mel-2 Undifferentiated P17
191.53
18.61
0.01
0.01
105.05
10.62


ADSC Untreated Ctrl
1.00
0.00
1.00
0.06
1.19
0.91









The Expression of TP63 (Ectoderm/Surface Ectoderm) was up-regulated in both the Day 71 S71-transfected sample (123.000-fold) and the Day 75 NF10-transfected sample (167.000-fold) as compared to the ADSC untreated control. The expression of NOTCH1, a marker for the neuroectoderm, was slightly up-regulated in the Day 71 S71-transfected sample (5-fold) and the Day 75 NF10-transfected sample (6-fold) as compared to the ADSC untreated control. Expression of CDH1/E-cadherin, surface ectoderm marker, was up-regulated in both the Day 71 S71-transfected sample (59.000-fold) and the Day 75 NF10-transfected sample (78.000-fold) as compared to the ADSC untreated control. Furthermore, the expression of ASCL1/MASH1, a neural crest marker, was up-regulated in both the Day 71 S71-transfected sample (70-fold) and the Day 75 NF10-transfected sample (48-fold) as compared to the ADSC untreated control.


The expression of definitive endodeiin markers were analyzed, such as SOX17, CDX2, and NKX2.1. The expression of SOX17 was up-regulated in the Day 71 S71-transfected sample (9-fold) as compared to the ADSC untreated control. CDX2 gene expression is up-regulated in both the Day 71 S71-transfected sample (38-fold) and the Day 75 NFIO-transfected sample (27-fold) as compared to the ADSC untreated control. However, NKX2-1 (Endoderm-Primitive Gut Derivative) was not expressed in any of the 4 test samples at the time points tested.


Markers of Mesoderm were analyzed as well: the expression of T (Mesoderm) was up-regulated in the Day 71 S71-transfected sample (16-fold) as compared to the ADSC untreated control. However the expression of MEOX1 OSR1 (intermediate mesoderm), and FOXF1 (lateral mesoderm) was decreased in both the S71-transfected and the NF10-transfected samples as compared to the ADSC Untreated Ctrl. The expression of GATA4 is up-regulated in both the Day 71 S71-transfected sample (150-fold) and the Day 75 NF10-transfected sample (110-fold) as compared to the ADSC untreated control.


The expression of FOXA2 (Mesendoderm, Mesoderm, Endoderm) was up-regulated in both the Day 71 S71-transfected sample (1.100-fold) and the Day 75 NF10-transfected sample (1.600-fold) as compared to the ADSC untreated control. No significant change in GSC (mesoendoderm, Endoderm) expression for both the S71-transfected and the NFIO-transfected samples as compared to the ADSC Untreated Ctrl. The expression of CXCR4, a marker for Mesoderm/Endoderm, was up-regulated in both the Day 71 S71-transfected sample (500-fold) and the Day 75 NF10-transfected sample (350-fold) as compared to the ADSC untreated control.


Using immunohistochemistry, the potential of PLSCs to express and differentiate into the three embryonic layers markers was analyzed. Consistent with their hES-like morphology, PLSCs were able to differentiation towards the three lineages:


Ectodermal differentiation: Nestin is strongly and widely expressed throughout the experiment. The neural markers GFAP and BIII-tubulin are also expressed, while Sox2 as a neural precursor marker was not detectable at the time points tested, indicating that cells had already differentiated into the neural pathway. At day 12 and 18, many cells co-expressed GFAP and 131H-tubulin. At day 28, however, GFAP expressing cells have almost completely disappeared, while BIII-tubulin positive cells with well formed neurites become detectable (FIG. 26). The medium supported neural differentiation, but less astrocyte differentiation, since, especially at day 12 and 18, there are high amounts of GFAP-positive collapsed cells detectable, indicating massive cell death of this type of cells. The omnipresence of the neural markers suggest that differentiation occurs along the neural pathway in this media; although different differentiation lineages could be achieved with different media compositions and growth substrates/growth conditions known in the art. By Day 12 of differentiation, the expression of the stem cell markers Oct4 and Nanog are completely suppressed.


Endodermal differentiation: Large fields of cells that are positive for FoxA2 are detectable indicating endodermal differentiation.


Mesodermal differentiation: Cells treated for mesodermal differentiation cease to express Oct4 and nanog (not shown), indicating that they indeed started to differentiate.


The strong and ubiquitous expression of GATA4 and the later appearance of nuclear Nkx2.5 stain indicate that many cells might develop a cardiac profile in the conditions tested.


There are some cell clusters that express brachyury staining together with GATA4.


As for the Ectoderm and Endoderm differentiation, different media compositions and growth substrates/growth conditions known in the art would promote the acquisition of other differentiation lineages.


Example XXV: Cytotoxic Effects of Human PLSCs

Human Hepatocellular Carcinoma cell line (HepG2, CRL-10741, ATCC) were seeded in a 24 well plate (1×104 cells/cm2) in DMEM with 10% Fetal bovine serum (FBS). PLSCs were harvested using dispase and seeded (1×104 cells/cm2) in transwells (Corning, 0.4 μm) above the HepG2 containing wells. The HepG2 cells were analyzed for cytotoxicity after 24 hours by high-content analysis staining by Propodium Iodide, Yoprol, and Ethidium Heterodimer (cell-impeimeable DNA stains that stain only cells with a compromised plasma membrane as occurs in unhealthy cells) along with Calcein (a live stain, marking enzymatic activity in the cytoplasm), Mitotracker Red (marks active, healthy mitochondria) and Cleaved Caspase 3 (a marker for apoptotic or stressed cells). HepG2 cells in the presence of PLSCs had significantly less Propodium Iodide, Yoprol and Cleaved Caspase 3 staining indicating that the PLSCs had a protective and/or therapeutic/regenerative effect on these cells (Table 47).









TABLE 47







The percentage of Yopro1, Propidium, ethidium Heterodimer, Calcein


AM, Mitotracker Red, and Cleaved Caspase-3 positive cells for untreated and


PLSCs treated hepatocytes.













Average




% positive cells
intensity















Number of

Propidium
Ethidium

Cleaved
Mitotracker



cells
Yopro1
Iodide
Heterodimer
CalceinAM
Caspase3
Red





HepG2
14539 ± 1359
5.15 ± 0.47
4.57 ± 0.21
5.66 ± 0.02
69.41 ± 1.44
0.95 ± 0.05
26.88 ± 3.52


untreated









HepG2,
12498 ± 1454
3.41 ± 0.37
3.85 ± 0.36
5.81 ± 0.33
71.67 ± 1.55
0.63 ± 0.03
27.99 ± 3.74


PSLC









treated









Example XXVI: Teratoma Formation Assay in Immunodeficient NOD-SCID Mice

Fifty million human NSLCs derived from fibroblast cells (HFF) and fifty millions NSLCs derived from blood cells (CD34+) (according to Examples IV and IX), as well as twenty-five million Human embryonic stem cells (Mel2) were prepared as units of five million cells each embedded with 30% Matrigel (Invitrogen) in 200 μl phosphate buffered saline which were then injected subcutaneously into the back of individual NOD/SCID mice using a 21-G needle within 20 minutes of preparation. Each cell line had been in culture for approximately 1 month prior to the study. Thus 10 mice received an injection of 5 million NSLCs derived from HFF, 10 mice received an injection of 5 million NSLCs derived from CD34+ cells, and 5 mice received an injection of 5 million Mel2 cells. All standard and appropriate animal approval and ethical committee approvals were obtained prior to the commencement of the study.


Although the study was for 3 months, within 1 month of injection one of the mice injected with the Mel2 cells had to be sacrificed due to the size of the teratoma that had formed according to the animal approval protocol. Less than 3 weeks later, the rest of the Mel2 injected mice had to also be sacrificed due to the size of the teratoma that had formed. None of the 10 mice injected with NSLCs derived from fibroblast cells or the 10 mice injected with NSLCs derived from CD34+ blood cells formed any tumors or teratomas, indicating that these NSLCs are safe multipotent stem cells.


Example XXVII: Differentiation of NSLC Derived from Mia, Cd34+ Cells, and Keratinocytes to Different Neuronal Lineages

To investigate the differentiation potential of NSLCs to different types of neurons, NSLC Neurospheres were dissociated and plated onto laminin/poly-D-Lysine (10 μg/ml; Sigma) coated plates in NeuroCult differentiation medium (NeuroCult Differentiation basal, StemCell Technologies) supplemented with NeuroCult® SM1 Neuronal Supplement and BDNF (20 ng/ml, Peprotech), bFGF (40 ng/ml, Peprotech), FGF 8 (20 ng/ml, Peprotech) and SHH (20 ng/ml, Peprotech) for 30 days. NeuroCult® SM1 (StemCell Technologies) is a standardized serum-free supplement containing antioxidants and retinoic acid; its formulation was developed based on the published supplement formulation identified as B27 but has been optimized to give reproducibly high numbers of functional neurons with minimal glial cell contamination (<1% GFAP+). After one month in culture, the cells were stained with the neuronal marker tyrosine hydroxylase, acetycholine, GABA, and Dopamine. All cell lines stained positive for each of these markers. Immunohistochemistry analysis showed that differentiation medium supplemented with Neurocult SM1 and neurotrophic factors promoted the differentiation of NSLCs derived from HFF, CD34+ cells, and Keratinocytes to dopaminergic, adrenergic, and Gabanergic neurons.


This study showed that NSLCs derived from fibroblast, keratinocyte, or blood cells are capable to differentiate towards different types of neurons in the appropriate differentiation conditions. The specific neurons could be used to treat diseases where such neurons are affected or have been lost, for example, dopaminergic neruons in Parkinson's disease. Other types of multipotent stem cells and progenitor cells prepared according to the methods in the previous examples would be expected to reprogram or differentiate more along certain pathways with the appropriate media and supplements, growth substrate and growth conditions known in the art.


Example XXVIII: Comparison of Expression of Specific Genes in Human HFFs, NSLCs Created from the HFFs, and Human Primary Neuroprogenitor Cells

The expression of selected genes and proteins in NSLCs created from HFFs according to Example IV were determined. Total RNA was extracted from cells, using Trizol following manufacturer's recommendation. Briefly, cells were examined for the expression of different genes associated with pluripotency: SOX2 (460 bp), OCT4a (172 bp), OCT4b (169 bp) and NANOG (276 bp); early neural markers: SOX2 (460 bp), NES (327 bp), CD133 (200 bp), PAX6 (431 bp) and ASCL (220 bp); Notch signaling: NOTCH1 (126 bp), NOTCH2 (475 bp), HES1 (314 bp) and HESS (265 bp); neurotrophic factors: GDNF (389 bp) and BDNF (743 bp) and astrocyte marker GFAP (650 bp) and markers of neurons: NFL (284 bp), NFM (333 bp), NFH (316 bp), SYN (289 bp), NSE (292 bp) and MAP2 (321 bp), Positive controls used were NT2 cells (human NT2/D1 teratomacarcinoma cell line (ATCC), NT2-derived neurons (NT2-N) and astrocytes (NT2-A) and SH-SY5Y cells were appropriate. No template control was used as negative control for every gene examined.


As seen in FIG. 27, NSLCs had the same expression profile as human primary neuroprogenitor cells (NT; Lonza), namely in terms of expression of Sox2, Oct4b, Notch1, Notch2, Hes1, Hes5, Nestin, CD133, Pax6, Ascl1 (Mash1), NFL, NFM, NSE and Map2, but additionally the of the neurotrophic factors GDNF and BDNF. The HFF cells that the NSLCs were created from also expressed Oct4b (Oct4b is a spicing variant of Oct4a (a stem cell marker) which expression is not limited to stem cells), Nestin, some of the notch signaling genes and neuronal markers, making it significantly easier to reprogram than adult dermal fibroblasts that do not express this genes (thus, for example, Nestin is important to add to the reprogramming cocktail when reprogramming adult fibroblasts to NSLCs).


Next immunohistochemistry was used to confirm that the encoded protein of some of the above expressed genes was present in the cells. Cells were fixed with 65% Ethanol and 0.15M NaCl for 20 minutes and stained for the following proteins: SOX2, NES, GFAP, MAP2, NCAM and RBPJ (green). Phase and Hoechst (blue) counter stain was also preformed. NT2 and NP cells were used as comparison and positive controls. NSLCs expressed intense staining for Sox2, Nestin, GFAP, Map2, and RBPJ with only a small amount of NCAM detected. NP cells had an identical staining pattern to the NSLCs except slightly less intense staining especially for Sox2 and Nestin. The NT2 cells only expressed Sox2, Nestin and RBPJ at appreciable levels. The HFFs from which the NSLCs were created from only expressed Nestin and RBPJ at appreciable levels.


Example XXIX: Potential of NSLCs for CNS Therapeutic Applications

NSLCs prepared from HFFs according to Example IV were tested for their ability to form functional cell-cell communication through gap junctions, which is an important characteristic for therapeutic potential when implanted into the CNS. Dye coupling experiments showed functional cell-cell communication in NSLC cells through gap junctions. A single NSCL cell was pre-loaded with two dyes [Dil (red) and calcein AM (green)] and plated on an unlabeled layer of NSCL cells. Calcein readily transferred from the donor cells to a large population of receiving cells after 3 hours in vitro, confirming gap junctional intercellular communication among NSCL cells (FIG. 28).


Next NSLCs were tested for the expression of synaptotagmin (a synaptic vesicle protein) and MAP2 (a neural differentiation associated marker) since NSLCs have some unique characteristics over native neural stem/progenitor cells including expression of some growth factors as well as a more significant expression of some neural differentiation markers while maintaining the ability to remain in a stem cell like state. NSLCs were found to readily express synaptotagmin and Map2 (FIG. 29).


Next NSLCs were tested for their tolerance to glutamate and NMDA to determine their potential robustness in areas of significant ischemia, trauma, and neurodegeneration in the CNS. NSLCs maintained a healthy neuronal state in the presence of glutamate (200 mM), a characteristic not observed in the presence of NMDA (25 mM), indicating an ability to form synapses upon neuronal differentiation.


Next the NSLCs were tested for their ability to attach and survive on 3D scaffolds; in this case PGA scaffolds (FIG. 30). NSLCs were found to readily attach to the PGA scaffolds designed for implantation into the brain. Carboxyfluourodiacetate (CFDA) confirms optimal cell survival (>93%) 24 hours after cells were plated on the scaffold. NSLCs showed robust attachment and survival to the surface for at least one week in vitro. Furthermore, Neurites grew along the length of each PGA fiber, following the pattern of the scaffold, indicating that NSLCs can be grown on 3D scaffold and structures useful for implantation into humans or animals, or for in vitro modeling.


PATENT LITERATURE CITED

Bhasin, Vishal. 2010. Method of Producing Progenitor Cells from Differentiated Cells. PCT/AU2010000120.


Cifarelli R A, Cellini F, Liddo R D, Parnigotto et al., 2010. Method For the Production of Multicomponent Stem Cells, Relative kits and Used in the Medical Field. US 2010/0003223A1.


Kremer B K F, Fandrich F, 2006. Dedifferentiated, Programmable Stem Cells of Monocytic Origin, and Their Production and Use. U.S. Pat. No. 7,138,257 B2.


Winner G F. Newson B S, Rill D R, and Williams Jim C. 2010. Monocyte-Derived Stem Cells. US2010/0047908A1.


Bennett D et al. 2009 Stem-Like Cells and Method for Reprogramming Adult Mammalian Somatic Cells. Wo/2009/079007.


Akamatsu W, Okano H. Method for Producing Neural Stem Cells. Wo/2010/052904.


You, S, Moon J H, Yoon B S, Kim K D, Park G, Jun E K, Kim B, Yoo, S J, Kwak S S, Maeng I. 2007. Dedifferentiation d'Astrocytes en Cellules Souches Neuronales au Moyen du Gene BMI-1. (WO/2007/097494).


You, S, Moon J H, Yoon B S, Kim D, Park G, Jun E K, Kim B, Yoo, S J, Kwak S S, Maeng I. 2009. De-Differentiation of Astrocytes into Neural Stem Cell using Nanog. US2009/0246870A1.


Chen S, Ding S, Schltz P G. Compositions and Methods for Inducing Cell Dedifferentiation. US2005/0176707A1.


You S, et al., 2009. De-Differentiation of Astrocytes into Neural Stem Cells using SHH. Us 2009/0227023.


Oliveri, R and Andersen, C Y. 2009. Method for Increasing the Plasticity Level of a Cell. WO/2009/018832.


Imai T, Tokunaga A, Yoshida T, Mikoshiba K, Nakafuku M, Okana H. Num Protein. 2004. Expression Inhibitor making Use Musashi. Us 2004/0054140A1.


Okano, H. 2003. RNA-Binding Protein Musahi 2. WO03/016531.


SCIENTIFIC PUBLICATION LITERATURE CITED

Takahashi K, Tanabe K, Ohnuki M, Narita M, Ichisaka T, Tomoka K, Yamanaka S. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell. 2007 Nov. 30; 131(5):861-72.


Okita K, Hong H, Takahashi K, Yamanaka S. Generation of mouse-induced pluripotent stem cells with plasmid vectors. Nat. Protoc. 2010; 5(3):418-28.


Yu J, Hu K, Smuga-Otto, Tian S, Stewart R, Slukvin I I, Thomson J A. Human induced pluripotent stem cells free of vector and transgene sequences. Science 2009 8; 324(5928):797-801.


Zeitlow R, Lane E L, Dunnet S B, Rosser A E. Human stem cells for CNSrepair. Cell Tissue Res. 2008; 331(1):301-22.


Singec I, Jandial R, Crain A, Nikkhah G, and Snyde E. Y. The Leading Edge of Stem Cell Therapeutics. Annual Review of Medicine 2007:58; 313-328.


Mimeault, M., Hauke, R. & Batra, S. K. 2007. Stem cells: a revolution in therapeutics-recent advances in stem cell biology and their therapeutic applications in regenerative medicine and cancer therapies. Clin Pharmacol Ther, 82, 252-64.


Levesque, M F and Neuman T. Transdiffentiation of transfected epidermal basal cells into neural progenitor cells, neuronal cells and/or glial cells. Patent, filling date 2000.


Shea T B. Neuritogenesis in mouse NB2a/dl neuroblastoma cells: triggering by calcium influx and involvement of actin and tubulin dynamics. Cell Biol Int Rep. 1990; 14(11):967-79.


Yeomans N D, Trier J S, Moxey P C, and Markezin E T. Maturation and differentiation of cultured fetal stomach. Effects of corticosteroids, pentagastrin, and cytochalasin B. Gasteroenterology 1976; 71(5):770-7.


Paterson F C, Rudland P S. Microtubule-disrupting drugs increase the frequency of conversion of a rat mammary epithelial stem cell line to elongated, myoepithelial-like cells in culture. J Cell Phsiol. 1985; 125(1):135-50.


Bouwens L. Transdifferentiation versus stem cell hypothesis for the regeneration of islet beta-cells in the pancreas. Micro Res Tech. 1998; 43(4):332-6.


Bouwens L. Cytokeratins and cell differentiation in the pancreas. J Pathol. 1998b; 184(3):234-9.


Theise N D, Nimmakayalu M, Gardner R, Illei P B, Morgan G, Teperman L, Henegariu O, Krause D S. Liver from bone marrow in humans. Hepatology 2000; 32(1):11-6.


Woodbury D, Schwarz E J, Prockop D J, Black I B. Adult rat and human bone marrow stromal cells differentiate into neurons. J Neurosci Res. 2000; 61(4):364-70.


Brunet, J F; Ghysen, A. Deconstructing cell determination: proneural genes and neuronal identity. Bioessays. 1999; 21:313-318.


Bertrand N, Castro D S, and Guillemot F. Proneural genes and the specification of neural cell types. Nat Rev Neurosci. 2002; 3(7):517-30.


McCormick M B, Tamimi R M, Snider L, Asakura A, Bergstrom D, Tapscott S J. NeuroD2 and neuroD3: distinct expression patterns and transcriptional activation potentials within the neuroD gene family. Mol Cell Biol. 1996; 16(10):5792-800.


Guillemot F, Lo L C, Johnson J E, Auerbach A, Anderson D J, Joyner A I. Mammalian achaete-scute homolog 1 is required for the early development of olfactory and autonomic neurons. Cell 1993; 75(3):463-76.


Fode C, Gradwohl G, Morin X, Dierich A, LeMeur M, Goridis C, Guillemot F. The bHLH protein NEUROGENIN 2 is a determination factor for epibranchial placode-derived sensory neurons. Neuron 1998; 20(3):483-94.


Fernandes K J L, McKenzie I A, Mill P, Smith K M, Akhavan M, Barnabe-Heider F, Biernaskie J, Junek A, et al. A dermal niche for multipotent adult skin-derived precursor cells. Nature Cell Biology 2004; 6:1082-1093.


Jacobsen F, Hirsch T, Mittler D, Schulte M, Lehnhardt M, Druecke D, Homann H H, Steinau H U, Steinstraesser L. Polybrene improves transfection efficacy of recombinant replication-deficient adenovirus in cutaneous cells and burned skin. J Gene Med. 2006; 8(2):138-46.


Kearns C M, Gash D M. GDNF protects nigral dopamine neurons against 6-hydroxydopamine in vivo. Brain Res. 1995; 672(1-2):104-11.


Gash D M, Zhang Z, Ovadia A, Cass W A, Yi A, Simmerman L, Russel D, Martin D, Lapchak P A, Collins F, Hoffer B J, Gerhardt G A. Functional recovery in parkinsonian monkeys treated with GDNF. Nature 1996; 380(6571):252-5.


Lindner M D, Winn S R, Baetge E E, Hammang J P, Gentile F T, Doherty E, McDermott P E, Frydel B, Ullman M D, Schallert T et al. Implantation of encapsulated catecholamine and GDNF-producing cells in rats with unilateral dopamine depletions and parkinsonian symptoms. Exp Neurol. 1995; 132(1):62-76.


Kordower J H, Emborg M E, Bloch J, Ma S Y, Chu Y, Leventhal L, McBride J, Chen E Y, Palfi S, Roitberg B Z, Brown W D, Holden J E, et al. Neurodegeneration prevented by lentiviral vector delivery of GDNF in primate models of Parkinson's disease. Science 2000; 290(5492):767-73.


Martinez-Serrano A, Bjorklund A. Immortalized neural progenitor cells for CNS gene transfer and repair. Trends Neurosci. 1997; 20(11):530-8.


Chambers S M, Fasano C A, Papapetrou E P, Tomishima M, Sadelain M, Studer L. Highly efficient neural conversion of human ES and iPS cells by dual inhibition of SMAD signaling. Nat Biotechnol. 2009; 27(3):275-80.


Headings are included herein for reference and to aid in locating certain sections These headings are not intended to limit the scope of the concepts described therein under, and these concepts may have applicability in other sections throughout the entire specification Thus, the present invention is not intended to be limited to the embodiments shown herein but is to be accorded the widest scope consistent with the principles and novel features disclosed herein.


It is understood that the examples and embodiments described herein are for illustrative purposes only and that various modifications or changes in light thereof will be suggested to persons skilled in the art and are to be included within the present invention and scope of the appended claims

Claims
  • 1. A method of obtaining a pancreatic multipotent or unipotent cell, comprising: i) providing a cell of a first type which is not a pancreatic multipotent or unipotent cell, wherein the cell of the first type is an adipose stein cell (ADSC) or a fibroblast cell;ii) contacting the cell of a first type with an agent capable of remodeling the chromatin and/or DNA of the cell, wherein the agent capable of remodeling the chromatin and/or DNA is a historic acetylator, an inhibitor of historic deacetylation, a DNA demethylator, and/or an inhibitor of DNA methylation;iii) introducing into the cell of a first type at least one pancreatic multipotent or unipotent gene regulator polypeptide and/or a polynucleotide encoding said at least one pancreatic multipotent or unipotent gene regulator, to a level at which the at least one pancreatic multipotent or unipotent gene regulator is capable of driving transformation of the cell of a first type into the pancreatic multipotent or unipotent cell, wherein the at least one pancreatic multipotent or unipotent gene regulator is selected from the group consisting of: Sox17, Pdx1, and Ngn3; Oct4, Sox17, Pdx1, and Ngn3; Oct4, Pdx1, and Ngn3; FoxA2, Pdx1, and Sox17; Pdx1 and Ngn3; Pdx1 and FoxA2; and Pdx1 and Oct4; andiv) placing or maintaining the cell in a pancreatic cell culture medium and maintaining intracellular levels of the at least one pancreatic multipotent or unipotent gene regulator for a sufficient period of time to allow a pancreatic multipotent or unipotent cell to be obtained.
  • 2. The method of claim 1, wherein, in step (ii), the remodeling agent is methyl-CpG binding domain protein 2 (MBD2), DNA-damage-inducible beta (Gadd45b), valproic acid or 5-azacytidine.
  • 3. The method of claim 1, wherein, in step (ii), the remodeling agent is methyl-CpG binding domain protein 2 (MBD2).
  • 4. The method of claim 1, wherein the pancreatic multipotent or unipotent cell so obtained possesses one or more of the following characteristics: i) expression of one or more pancreatic progenitor-like cell marker selected from the group consisting of Pdx1, Ngn3, NKX2.2, Gata4 and FoxA2;ii) is capable of differentiation into at least one cell expressing a marker specific for a pancreatic progenitor-like cell;iii) has one or more morphological characteristic of a pancreatic stem cell or pancreatic progenitor cell;iv) expression of at least one pancreatic-specific antigen;v) expression of one or more functional pancreatic markers upon pancreatic differentiation; andvi) is capable of significantly improving or maintaining one or more pancreatic functional measures after injecting an adequate number of said pancreatic multipotent or unipotent cells into a mouse model for pancreatic disease.
  • 5. The method of claim 1, wherein the pancreatic multipotent cell so obtained possesses all of the following characteristics: (i) expresses one or more pancreatic progenitor cell marker;(ii) can self-renew for significantly longer than a somatic cell;(iii) is not a cancerous cell;(iv) is stable and not artificially maintained by forced gene expression and may be maintained in standard pancreatic stem cell media;(v) can differentiate to a pancreatic precursor cell, an islet β-cell or to another differentiated cell type of the pancreatic lineage; and(vi) does not exhibit uncontrolled growth, teratoma formation, or tumor formation in vivo.
  • 6. The method of claim 1, wherein a plurality of pancreatic multipotent or unipotent cells are obtained and wherein the plurality of pancreatic multipotent or unipotent cells are organized within a three-dimensional structure.
  • 7. The method of claim 1, wherein the cell of the first cell type is a human fibroblast cell or a human adipose derived stem cell.
  • 8. The method of claim 1, further comprising treating the cells of a first cell type with a cytoskeleton disruptor.
  • 9. The method of claim 8, wherein the cytoskeleton disruptor is Cytochalasin B or a myosin inhibitor.
  • 10. The method of claim 1, wherein the pancreatic unipotent cell so obtained possesses all of the following characteristics: (i) expresses a pancreatic marker;(ii) is not a cancerous cell;(iii) is stable and not artificially maintained by forced gene expression and may be maintained in standard pancreatic cell media; and(iv) does not exhibit uncontrolled growth, teratoma formation, or tumor formation in vivo.
  • 11. The method of claim 10, wherein the pancreatic marker is selected from the group consisting of Pdx1, Ngn3, NKX2.2, Gata4 and FoxA2.
  • 12. The method of claim 4, wherein the one or more functional pancreatic marker expressed upon pancreatic differentiation is selected from the group consisting of Pdx1, Ngn3, NKX2.2, Gata4 and FoxA2.
  • 13. The method of claim 1, wherein the pancreatic multipotent or unipotent cell obtained is a multipotent cell and capable of differentiating into a pancreatic unipotent or somatic cell.
  • 14. The method of claim 1, wherein the pancreatic multipotent or unipotent cell obtained is a unipotent cell and capable of differentiating into a pancreatic somatic cell.
  • 15. The method of claim 14, wherein the pancreatic somatic cell is an islet β-cell capable of secreting insulin.
  • 16. The method of claim 1, wherein in step the cell of a first type is transfected with at least one expression vector encoding polypeptide(s) selected from the group consisting of: Sox17, Pdx1, and Ngn3; Oct4, Sox17, Pdx1, and Ngn3; Oct4, Pdx1, and Ngn3; FoxA2, Pdx1, and Sox17; Pdx1 and Ngn3; Pdx1 and FoxA2; and Pdx1 and Oct4.
  • 17. The method of claim 1, wherein the fibroblast cell is a fetal fibroblast cell.
US Referenced Citations (36)
Number Name Date Kind
6087168 Levesque et al. Jul 2000 A
6949380 Levesque et al. Sep 2005 B1
9453205 Ahlfors et al. Sep 2016 B2
9528087 Ahlfors et al. Dec 2016 B2
10017737 Ahlfors et al. Jul 2018 B2
10131879 Ahlfors et al. Nov 2018 B2
10260046 Ahlfors et al. Apr 2019 B2
10557123 Ahlfors et al. Feb 2020 B2
10563176 Ahlfors et al. Feb 2020 B2
20020136709 Zahner et al. Sep 2002 A1
20030059939 Page et al. Mar 2003 A1
20030217378 Stice et al. Nov 2003 A1
20080152630 Ginis et al. Jun 2008 A1
20080254004 Terskikh et al. Oct 2008 A1
20090136461 Kim et al. May 2009 A1
20090162329 Anversa et al. Jun 2009 A1
20110206644 Estrov et al. Aug 2011 A1
20110217274 Reld Sep 2011 A1
20110223670 Chwartz et al. Sep 2011 A1
20120129262 West et al. May 2012 A1
20120220034 Ahlfors et al. Aug 2012 A1
20120288936 Ahlfors et al. Nov 2012 A1
20130064799 Rana Mar 2013 A1
20130097717 Zhu Apr 2013 A1
20140301991 Srivastava Oct 2014 A1
20150004145 Lemischka et al. Jan 2015 A1
20160032317 Rossi et al. Feb 2016 A1
20160160184 Ahlfors et al. Jun 2016 A1
20160298086 Terzic et al. Oct 2016 A1
20170029783 Ahlfors et al. Feb 2017 A1
20170101622 Ahlfors et al. Apr 2017 A1
20170101623 Ahlfors et al. Apr 2017 A1
20170101626 Ahlfors et al. Apr 2017 A1
20190024056 Ahlfors et al. Jan 2019 A1
20190085294 Ahlfors et al. Mar 2019 A1
20200165570 Ahlfors et al. May 2020 A1
Foreign Referenced Citations (16)
Number Date Country
2698091 Mar 2009 CA
2660123 Apr 2009 CA
1022330 Jul 2000 EP
2 096 169 Sep 2009 EP
20040016785 Feb 2004 KR
WO 03018767 Mar 2003 WO
WO 2003018767 Mar 2003 WO
WO-2007047509 Apr 2007 WO
WO 2007097494 Aug 2007 WO
WO 2009018832 Feb 2009 WO
WO 2009057831 May 2009 WO
WO 2009079007 Jun 2009 WO
WO 2010052904 May 2010 WO
WO-2010075575 Jul 2010 WO
WO 2010088735 Aug 2010 WO
WO-2010108126 Sep 2010 WO
Non-Patent Literature Citations (86)
Entry
Hanna et al., “Pluripotency and Cellular Reprogramming: Facts, hypotheses, Unresolved Issues,” Cell, Nov. 12, 2010, vol. 143, pp. 508-525.
Hochedlinger; and Plath 2009 “Epigenetic reprogramming and induced plutipotency” Development 136(4): 509-523.
Nishikawa et al., “The promise of human induced pluripotent stem cells for research and therapy” Stem Cells 9: pp. 725-729 (2008).
Ronaghi et al., “Challenges of stem cell therapy for spinal cord injury: Human embryonic stem cells, endogenous neutral stem cells, or induced pluripotent stem cells?” Stem Cells (2010).
Simeonov; and Uppal 2014 “Direct reprogramming of human fibroblasts to hepatocyte-like cells by synthetic modified mRNAs” Plos One 9(6): 1-11.
Wang et al. 2005 “Embryonic stem cell-derived hematopoietic stem cells”, PNAS, Dec. 27, 2005, vol. 102, No. 52, 19081-19086.
Abeliovich, et al. 2009 “Reprogramming therapeutics: IPS cell prospects for neurodegenerative disease” , Neuron.,61 (3): 337-9.
Akazawa, et al. 1995 “A mammalian helix-loop-helix factor structurally related to the product of Drosophila proneural gene atonal is a positive transcriptional regulator expressed in the developing nervous ISystem” J Biol. Chem., 270(15): 8730-8.
Baghbaderani, B.A., et al., 2009 “Bioreactor Expansion of Human Neural Precursor Cells in Serum-Free Media Retains Neurogenic Potential” Biotechnol Bioeng 105: 823-833.
Bertrand, et al. 2002 “Proneural genes and the specification of neural cell types” Nat Rev Neurosci., 3(7): 517-30.
Bouwens, et al. 1998 “Cytokeratins and cell differentiation in the pancreas” J. Pathol. 184: 234-9.
Bouwens, et al. 1998 “Transdifferentiation versus stem cell hypothesis for the regeneration of islet beta-cells in the pancreas” Micro Res Tech 43(4): 332-336.
Brunet, et al. 1999 “Deconstructing cell determination: proneural genes and neuronal identity” BioEssays 21: 313-318.
Detich et al. 2002 “Promoter-specific Activation and Demethylation by MBD2/Demethylase” J Biol Chem 277: 35791-35794.
Caporaso, et al. 2003 “Telomerase activity in the subventricular zone of adult mice” Mol Cell Neurosci,. 23(4): 693-702.
Chambers, et al. 2009 “Highly efficient neural conversion of human ES and ips cells by dual inhibition of SMAD signaling” Nat. Biotech., 27(3): 275-280.
Davis, Robert L. et al., “Expression of a single transfected cDNA converts fibroblasts to myoblasts,” Cell, Dec. 24, 1987, vol. 51, Issue 6, pp. 987-1000.
European Office Action dated Feb. 13, 2017, issued In European Application No. 10 825 907.8, 5 pages.
Extended European Search Report in European Application No. 10 825 907.8, dated Jun. 17, 2013.
Feng, et al. 2009 “Reprogramming of fibroblasts into induced pluripotent stem cells with orphan nuclear receptor Esrrb” Nat Cell Biol. 11(2): 197-203.
Fernandes, et al. 2004 “A dermal niche for multipotent adult skin-derived precursor cells” Nat Cell Biol. 6: 1082-1093.
Fode, et al. 1998 “The bHLH 5 protein Neurogenin 2 is a determination factor for epibranchial placode-derived sensory neurons” Neuron. 20(3): 483-494.
Frohman, et al. 2006 “Most patients with multiple sclerosis or a clinically isolated demyelinating syndrome should be treated at the time of diagnosis” Arch Neurol. 63(4): 614-619.
Gash, et al. 1996 “Functional recovery in parkinsonian monkeys treated with GDNF” Nature 380: 252-255.
Gene ID: 4152 2015 “MBD1 methyl-CpG binding domain protein 1 [ Homo sapiens (human) ]” downloaded on Dec. 19, 2015 from the World-Wide-Web at: ncbi.nlm.nih.gov/gene?cmd=Retrieve&dopt=full_report&list_uids=4152.
Gene ID: 8932 2015 “MBD2 methyl-CpG binding domain protein 2 [ Homo sapiens (human) ]” downloaded on Dec. 19, 2015 from the World-Wide-Web at: ncbi.nlm.nih.gov/gene?cmd=Retrieve&dopt=full_report&list_uids=8932.
Gene ID: 53615 2015 “MBD3 methyl-CpG binding domain protein 3 [ Homo sapiens (human) ]” downloaded on Dec. 19, 2015 from the World-Wide-Web at: ncbi.nlm.nih.gov/gene?cmd=Retrieve&dopt=full report&list_uids=53615.
Gene ID: 8930 2015 “MBD4 methyl-CpG binding domain 4 DNA glycosylase [ Homo sapiens (human) ]” downloaded on Dec. 19, 2015 from the World-Wide-Web at: ncbi.nlm.nih.gov/gene?cmd=Retrieve&dopt=full_report&list_uids=8930.
Gene ID: 4204 2015 “MECP2 methyl-CpG binding protein 2 [ Homo sapiens (human) ]” downloaded on Dec. 19, 2015 from the World-Wide-Web at: ncbi.nim.nih.gov/gene?cmd=Retrieve&dopt=full report&list_uids=4204.
Gene ID: 57379 2015 “AICDA activation-induced cytidine deaminase [ Homo sapiens (human) ]” downloaded on Dec. 19, 2015 from the World-Wide-Web at: ncbi.nlm.nih.gov/gene?cmd=Retrieve&dopt=full_report&list_uids=57379.
Goffin, et al. 2002 “DNA methyltransferase inhibitors—state of the art” Anals of Oncology 13: 1699-1716.
Guillemot, et al. 1993 “Mammalian achaete-scute homolog 1 is required for the early development of olfactory and autonomic neurons” Cell 75(3): 463-476.
Jacobson, et al. 1990 “Polybrene improves transfection efficacy of recombinant arepolication-deficient adenovirus in cutaneous cells and burned skin” J Gene Med. 8: 138-146.
Johnson, et al. 1990 “Two rat homologues of Drosophila achaete-scute specifically expressed in neuronal precursors” Nature 346(6287): 858-861.
Kaji, et al. 2009 “Virus-free induction of pluripotency and subsequent excision of reprogramming factors” Nature 458: 771-775.
Kaneko, et al. 2000 “Musashi1: an evolutionally conserved marker for CNS progenitor cells including neural stem cells” Dev Neurosci. 22(1-2): 139-153.
Keans, et al. 1995 “GDNF protects nigral dopamine neurons against 6 hydroxydopamine in vivo” Brain Res. 672: 104-111.
Kelaini, S. et al., “Direct reprogramming of adult cells: avoiding the pluripotent state,” Stem Cell and Cloning: Advances and Applications, Feb. 1, 2014, pp. 19-29.
Kim, et al. 2009 “Generation of human induced pluripotent stem cells by direct delivery of reoprogramming proteins” Cell Stem Cell 4(6): 472-476.
Kordower, et al. “Neurodegeneration prevented by lentiviral vector delivery of GDNF in ptimate models of parkinsons disease” Disease 290: 5492.
Kuo, et al. 1998 “Roles of histone acetyltransferases and deacetylases in gene regulation” BioEssays 20: 615-626.
Langer-Gould, et al. 2004 “Strategies for managing the side effects of treatments for multiple sclerosis” Neurology 63(suppl5): S35-41.
Lee et al. 2003 “Cellular and Genetic Characterization of human adult bone marrow-derived neural stem-like cells: a potential antiglioma cellular vector” Cancer Research 63: 8877-8889.
Lyko, et al. 2005 “DNA Methyltransferase Inhibitors and the Development of Epigenetic Cancer Therapies” JNCI 97: 1498-1506.
Lyssiotis, et al. “Reprogramming of murine fibroblasts to induced pluripotent stem cells with chemical complementation of Kif” PNAS 106(22): 8912-8917.
Martin et al. 2004 “Neural cell adhesion molecule expression in plasma cells in bone marrow biopsies and aspirates allows discrimination between multiple myeloma, monoclonal gammopathy of uncertain significance and polyclonal plasmacytosis” Histopathology 44: 375-380.
Martino, et al. 2006 “The therapeutic potential of neural stem cells” Nature Res. 7: 395-406.
Martinez-Serrano, et al. 1997 “Immortalized neural progenitor cells for CNS gene transfer and repair” Trends Neurosci. 20: 530-538.
McCormick, et al. 1996 “NeuroD2 and neuroD3: distinct expression patterns and transcriptional activation potentials within the neuroD gene family” Mol Cell Biol. 16(10: 5792-5800.
Méndez-Ferrer et al. 2010 “Mesenchymal and haematopoietic stem cells form a unique bone marrow niche” Nature 466: 829-836.
Mimeault, et al. 2007 “Stem cells: a revolution in therapeutics- recent advances in stem cell biology and their therapeutic applications in regenerative medicine and cancer therapies” Clin Pharmacol Ther. 82: 252-264.
Miyata, et al. 1999 “NeuroD is required for differentiation of the granule cells in the cerebellum and hippocampus” Genes Dev. 13(13): 1647-1652.
Okabe, S. et al. 1996 “Development of neuronal precursor cells and functional postmitotic neurons from embryonic stem cells in vitro.” Mechanisms of Development 59: 89-102.
Okada, Y et al. 2008 “Spatiotemporal recapitulation of central nervous system development by murine embryonic stem cell-derived neural stem/progenitor cells” Stem Cells 26: 3086-3098.
Okano, H., 2009 “A strategy for neuronal regeneration using IPS cells” Geriatric Medicine 47: 1369- 1377.
Okita, et al. 2010 “Generation of mouse-induced pluripotent stem cells with plasmid vectors” Nat. Protoc. 5(3): 418-427.
Ostenfeld, T. et al. 2000 “Human Neural Precursor Cells Express Low Levels of Telomerase in Vitro and Show Diminishing Cell Proliferation with Extensive Axonal Outgrowth following Transplantation” Experimental Neurology 164: 215-226.
Paterson, et al. 1985 “Microtubule-disrupting drugs increase the frequency of conversion of a rat mammary epithelial stem cell line to elongated, myoepithelial-like cells in culture” J Cell Physiol 125(1): 135-150.
Pei, et al. 2009 “Regulation of pluripotency and reprogramming by transcription factors” J Biol. Chem. 284(6): 3365-3369.
Rieske, P. et al. 2005 “Human fibroblast-derived cell lines have characteristics of embryonic stem cells and cells of neuro-ectodermal origin” Differentiation 73: 474-483.
Sato, et al. 1991 “Early and late contraction induced by ouabain in human umbilical arteries” Br J. Pharmacol. 103(2): 1525-1529.
Shea 1990 “Neurogenesis in mouse NB2a/d1 neuroblastoma cells: triggering by calcium influx and involvement of actin and tubulin dynamics” Cell Biol Int Rep. 14(11): 967-979.
Silva, et al. 2009 “Nanog is ther gateway to pluripotent ground state” Cell 138(4): 722-737.
Singec, et al. 2007 “The leading edge of stem cell therapeutics” Annu. Rev. Med 58: 313-328.
Soldner, et al. 2009 “Parkinson's disease patient-derived induced pluripotent stem cells free of viral reprogramming factors” Cell 136(5): 964-977.
Takahashi, et al. 2007 “Induction of pluripotent stem cells from fibroblast cultures” Nat. Protoc 2: 3081-3089.
Takahashi, et al. 2006 “Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors” Cell 126(4): 663-676.
Takebyashi, et al. 1997 “Conversion of ectoderm into a neural fate by ATH-3, a vertebrate basic helix-loop-helix gene homologous to Drosophila proneural gene atonal” EMBO 16(2): 384-395.
Tarnova, et al. 2006 “SOX2 is a dose-dependent regulator of retinal neural progenitor competence” Genes Dev. 20: 1187-1202.
Theise, et al. 2000 “Liver from bone marrow in humans” Hepatology 32(1): 11-16.
UNIPROTKB-Q9GZX7 (AICDA_Human) downloaded on Dec. 19, 2015 from the World-Wide-Web at: uniprot.org/uniprot/Q9GZX7.
Vierbuchen, T. et al., “Direct conversion of fibroblasts to functional neurons by defined factors,” Nature, Feb. 25, 2010, vol. 463, pp. 1035-1041.
Wang et al. 2008 “GADD45B inhibits MKK7-induced cardiac hypertrophy and the polymorphisms of GADD45B is associated with inter-ventricular septum hypertrophy” Biochem Biophys Res Commun 372(4): 623-628.
Warren, et al. 2010 “Highly efficient reprogramming to pluripotency and directed differentiation of human cells with synthetic modified mRNA” Cell Stem Cell 7(5): 618-630.
Wernig, M. et al. 2008 “Neurons derived from reprogrammed fibroblasts functionally integrate into the fetal brain and improve symptoms of rats with Parkinson's disease” Proc Natl Acad Sci USA 105: 5856-5861.
Wiese, C. et al. 2004 “Nestin expression—a property of multi-lineage progenitor cells?” CMLS Cellular and Molecular Life Sciences 61 : 2510-2522.
Woltjen, et al. 2009 “Piggyback transposition reprograms fibroblasts to induced pluripotent stem cells” Nature Letters 458: 766-771.
Woodbury, et al. 2000 “Adult rat and human bone marrow stromal cells differentiate into neurons” J. Neurosci Res. 61(4): 364-370.
Xu et al. (2007 “Histone deacetylase inhibitors: molecular mechanisms of action” Oncogene 26: 5541-5552.
Yeomans, et al. 1976 “Maturation and differentiation of cultured fetal stomach. Effects of corticosteroids, pentagastrin, and cytochalasin B” Gastroenterology 71(5): 770-777.
Yu, et al. 2009 “Human induced pluripotent stem cells free of vector and transgene sequences” Science 324(5928): 797-801.
Yu, et al. 2007 “Induced pluripotent stem cell lines derived from human somatic cells” Science 318(5858): 1917-1920.
Zhou, et al. 2009 “Adenoviral gene delivery can reprogram human fibroblasts to induced pluripotent stem cells” Stem Cells 27(11): 2667-2674.
Zhou et al., “Generation of induced pluripotent stem cells using recombinant proteins,” Cell Stem Cell, May 8, 2009, vol. 4, pp. 381-384.
Zietlow, et al. 2008 “Human stem cells for CNS repair” Cell Tissue Res. 331: 301-322.
NCBI GeneBank Nucleotide Published sequence for GATA binding protein 2 (GATA2) (NM 001145661). Year: 2022.
Related Publications (1)
Number Date Country
20200263140 A1 Aug 2020 US
Provisional Applications (1)
Number Date Country
61256967 Oct 2009 US
Divisions (1)
Number Date Country
Parent 13464987 May 2012 US
Child 15275921 US
Continuations (1)
Number Date Country
Parent 15275921 Sep 2016 US
Child 16752535 US
Continuation in Parts (1)
Number Date Country
Parent 13504988 US
Child 13464987 US