Induced stem cells: Difference between revisions

From Wikipedia, the free encyclopedia
Content deleted Content added
→‎Radical rejuvenation: ref links patent template
Line 98: Line 98:


Due to the fact that the use of iPSC for cell therapy is associated with significant risk of cancer and tumors there is urgent need to develop methods for safer cell lines suitable for use in the clinic.
Due to the fact that the use of iPSC for cell therapy is associated with significant risk of cancer and tumors there is urgent need to develop methods for safer cell lines suitable for use in the clinic.
An alternative to the methods of iPSC technique is the so-called "direct reprogramming" - induced by certain factors direct transdifferentiation of cells without passing through the stages of the pluripotent state.<ref name="Li">Li Qian, Yu Huang, C. Ian Spencer, Amy Foley, Vasanth Vedantham, Lei Liu, Simon J. Conway, Ji-dong Fu & Deepak Srivastava. (2012) In vivo reprogramming of murine cardiac fibroblasts into induced cardiomyocytes. Nature, 2012; DOI: 10.1038/nature11044</ref><ref name="Eva">Eva Szabo,et al & Mickie Bhatia (2010) Direct conversion of human fibroblasts to multilineage blood progenitors. Nature 468, 521-526</ref><ref name="Jem">Jem A. Efe, et al & Sheng Ding (2011) Conversion of mouse fibroblasts into cardiomyocytes using a direct reprogramming strategy Nature Cell Biology 13, 215-222</ref><ref name="Ernesto">Ernesto Lujan, Soham Chanda, Henrik Ahlenius, Thomas C. Südhof and Marius Wernig (2012) Direct conversion of mouse fibroblasts to self-renewing, tripotent neural precursor cells. PNAS Published online before print January 30, 2012, doi: 10.1073/pnas.1121003109</ref><ref name="Marc">Marc Thier, Philipp Wörsdörfer, Yenal B. Lakes, et al. (2012) Direct Conversion of Fibroblasts into Stably Expandable Neural Stem Cells. Cell Stem Cell, 22 March 2012, doi: 10.1016/j.stem.2012.03.003</ref><ref name="Han">Han DW, Tapia N., Hermann A., et al. & Schöler H.R. (2012) Direct Reprogramming of Fibroblasts into Neural Stem Cells by Defined Factors. Cell Stem Cell, April 6, 2012, doi: 10.1016/j.stem.2012.02.021</ref> The basis for this approach laid the study of Taylor and Jones, who showed that 5-azacytidine - reagent causing demethylation of DNA - can cause the formation of myogenic, chondrogenic, and adipogenic clones in the immortal cell line of mouse embryonic fibroblasts<ref>Taylor SM , Jones PA. (1979) Multiple new phenotypes induced in 10T1 / 2 and 3T3 cells treated with 5-azacytidine. Cell; 17:771-779</ref> and Weintraub et al., who found that the activation of a single gene, later named MyoD1, is sufficient for such reprogramming,<ref>Lassar AB, Paterson BM, Weintraub H. (1986) Transfection of a DNA locus that mediates the conversion of 10T1 / 2 fibroblasts to myoblasts. Cell.; 47 (5) :649-56</ref><ref>Davis RL, Weintraub H, Lassar AB. Expression of a single transfected cDNA converts fibroblasts to myoblasts. Cell. 1987; 51:987-1000</ref><ref>Weintraub, H., Tapscott, S. J., Davis, R. L., Thayer, M. J., Adam, M. A., Lassar, A. B. and Miller, A. D. (1989) Activation of muscle-specific genes in pigment, nerve, fat, liver, and fibroblast cell-lines by forced expression of Myod. Proc. Natl. Acad. Sci. U.S.A. 86, 5434-5438</ref>
An alternative to the methods of iPSC technique is the so-called "direct reprogramming" - induced by certain factors direct transdifferentiation of cells without passing through the stages of the pluripotent state.<ref name="Li">{{cite doi|10.1038/nature11044}}</ref><ref name="Eva">Eva Szabo,et al & Mickie Bhatia (2010) Direct conversion of human fibroblasts to multilineage blood progenitors. Nature 468, 521-526</ref><ref name="Jem">Jem A. Efe, et al & Sheng Ding (2011) Conversion of mouse fibroblasts into cardiomyocytes using a direct reprogramming strategy Nature Cell Biology 13, 215-222</ref><ref name="Ernesto">{{cite doi|10.1073/pnas.1121003109}}</ref><ref name="Marc">{{cite doi|10.1016/j.stem.2012.03.003}}</ref><ref name="Han">{{cite doi|10.1016/j.stem.2012.02.021}}</ref> The basis for this approach laid the study of Taylor and Jones, who showed that 5-azacytidine - reagent causing demethylation of DNA - can cause the formation of myogenic, chondrogenic, and adipogenic clones in the immortal cell line of mouse embryonic fibroblasts<ref>Taylor SM , Jones PA. (1979) Multiple new phenotypes induced in 10T1 / 2 and 3T3 cells treated with 5-azacytidine. Cell; 17:771-779</ref> and Weintraub et al., who found that the activation of a single gene, later named MyoD1, is sufficient for such reprogramming,<ref>Lassar AB, Paterson BM, Weintraub H. (1986) Transfection of a DNA locus that mediates the conversion of 10T1 / 2 fibroblasts to myoblasts. Cell.; 47 (5) :649-56</ref><ref>Davis RL, Weintraub H, Lassar AB. Expression of a single transfected cDNA converts fibroblasts to myoblasts. Cell. 1987; 51:987-1000</ref><ref>Weintraub, H., Tapscott, S. J., Davis, R. L., Thayer, M. J., Adam, M. A., Lassar, A. B. and Miller, A. D. (1989) Activation of muscle-specific genes in pigment, nerve, fat, liver, and fibroblast cell-lines by forced expression of Myod. Proc. Natl. Acad. Sci. U.S.A. 86, 5434-5438</ref>
Compared with iPSC whose reprogramming need at least two weeks, the formation of induced progenitor cells occurs relatively quickly - sometimes within a few days and the efficiency of reprogramming are usually many times higher. This reprogramming does not always require cell division.<ref>Thomas Vierbuchen and Marius Wernig (2011) Direct Lineage Conversions: Unnatural but useful? Nat Biotechnol.; 29 (10): 892-907. doi: 10.1038/nbt.1946</ref> But the main thing is that the resulting from such reprogramming multipotent somatic stem cells are more suitable for cell therapy because they do not form teratomas.<ref name="Han"/>
Compared with iPSC whose reprogramming need at least two weeks, the formation of induced progenitor cells occurs relatively quickly - sometimes within a few days and the efficiency of reprogramming are usually many times higher. This reprogramming does not always require cell division.<ref>{{cite doi|10.1038/nbt.1946}}</ref> But the main thing is that the resulting from such reprogramming multipotent somatic stem cells are more suitable for cell therapy because they do not form teratomas.<ref name="Han"/>


==== Single transcription factor transdifferentiation ====
==== Single transcription factor transdifferentiation ====


It was thought that only early embryonic cells could be coaxed into changing their identity. Mature cells are very resistant to changing their identity once they've committed to a specific kind. However, it has been found that brief expression of a single transcription factor, the ELT-7 GATA factor, can convert the identity of fully differentiated, highly specialized non-endodermal cells of the pharynx into fully differentiated intestinal cells in intact larvae and adult roundworm Caenorhabditis elegans. And there was no apparent requirement for a dedifferentiated intermediate during this transdifferentiation process.<ref>M. R. Riddle, A. Weintraub, K. C. Q. Nguyen, D. H. Hall, J. H. Rothman. (2013) Transdifferentiation and remodeling of post-embryonic C. elegans cells by a single transcription factor. Development, 140(24), 4844-4849 doi: 10.1242/dev.103010</ref>
It was thought that only early embryonic cells could be coaxed into changing their identity. Mature cells are very resistant to changing their identity once they've committed to a specific kind. However, it has been found that brief expression of a single transcription factor, the ELT-7 GATA factor, can convert the identity of fully differentiated, highly specialized non-endodermal cells of the pharynx into fully differentiated intestinal cells in intact larvae and adult roundworm Caenorhabditis elegans. And there was no apparent requirement for a dedifferentiated intermediate during this transdifferentiation process.<ref>{{cite doi|10.1242/dev.103010}}</ref>


=== Phased process modeling regeneration ===
=== Phased process modeling regeneration ===


Another way of reprogramming is the simulation of the processes that occur during amphibian limb regeneration. In urodele amphibians, an early step in limb regeneration is skeletal muscle fiber dedifferentiation into a cellulate that proliferates to contribute new limb tissue, whereas mammalian muscle cannot dedifferentiate after injury. However, sequential small molecule treatment of the muscle fiber with myoseverin, reversine (the aurora B kinase inhibitor) and some other chemicals: BIO (glycogen synthase-3 kinase inhibitor), lysophosphatidic acid (pleiotropic activator of G-protein-coupled receptors), SB203580 (p38 MAP kinase inhibitor), or SQ22536 (adenylyl cyclase inhibitor) causes the formation of new muscle cell types as well as other cell types such as precursors to fat, bone, and nervous system cells.<ref>Da-Woon Jung, Darren R. Williams (2011) Novel Chemically Defined Approach To Produce Multipotent Cells from Terminally Differentiated Tissue Syncytia. ACS Chemical Biology, 2011; : 110228124223097 DOI:10.1021/cb2000154</ref><ref>Kim WH, et al. & Williams DR.(2012) Small molecules that recapitulate the early steps of urodele amphibian limb regeneration and confer multipotency. ACS Chem Biol. ;7(4):732-743. DOI: 10.1021/cb300127f</ref>
Another way of reprogramming is the simulation of the processes that occur during amphibian limb regeneration. In urodele amphibians, an early step in limb regeneration is skeletal muscle fiber dedifferentiation into a cellulate that proliferates to contribute new limb tissue, whereas mammalian muscle cannot dedifferentiate after injury. However, sequential small molecule treatment of the muscle fiber with myoseverin, reversine (the aurora B kinase inhibitor) and some other chemicals: BIO (glycogen synthase-3 kinase inhibitor), lysophosphatidic acid (pleiotropic activator of G-protein-coupled receptors), SB203580 (p38 MAP kinase inhibitor), or SQ22536 (adenylyl cyclase inhibitor) causes the formation of new muscle cell types as well as other cell types such as precursors to fat, bone, and nervous system cells.<ref>{{cite doi|10.1021/cb2000154}}</ref><ref>{{cite doi|10.1021/cb300127f}}</ref>


=== Antibody-based transdifferentiation ===
=== Antibody-based transdifferentiation ===
Line 113: Line 113:
The researchers discovered [[granulocyte colony-stimulating factor|GCSF]]-mimicking [[antibody]]
The researchers discovered [[granulocyte colony-stimulating factor|GCSF]]-mimicking [[antibody]]
that can activate a growth-stimulating receptor on marrow cells in a way that induces marrow stem cells—which normally develop into white blood cells—to become neural progenitor cells.
that can activate a growth-stimulating receptor on marrow cells in a way that induces marrow stem cells—which normally develop into white blood cells—to become neural progenitor cells.
The new technique,<ref>Hongkai Zhang, Ian A. Wilson, and Richard A. Lerner (2012) [http://www.pnas.org/content/109/39/15728 Selection of antibodies that regulate phenotype from intracellular combinatorial antibody libraries]. PNAS. 109(39), 15728-15733 doi:10.1073/pnas.1214275109</ref> that enables researchers to search large libraries of antibodies and quickly select the ones with a desired biological effect, has been used for such purpose<ref>[http://www.scripps.edu/news/press/2013/20130422lerner.html Antibody that Transforms Bone Marrow Stem Cells Directly into Brain Cells]</ref><ref>Jia Xie, Hongkai Zhang, Kyungmoo Yea, and Richard A. Lerner (2013) [http://www.pnas.org/content/early/2013/04/23/1306263110.abstract Autocrine signaling based selection of combinatorial antibodies that transdifferentiate human stem cells] PNAS; doi:10.1073/pnas.1306263110</ref>
The new technique,<ref>Hongkai Zhang, Ian A. Wilson, and Richard A. Lerner (2012) [http://www.pnas.org/content/109/39/15728 Selection of antibodies that regulate phenotype from intracellular combinatorial antibody libraries]. PNAS. 109(39), 15728-15733 {{doi|10.1073/pnas.1214275109}}}}</ref> that enables researchers to search large libraries of antibodies and quickly select the ones with a desired biological effect, has been used for such purpose<ref>[http://www.scripps.edu/news/press/2013/20130422lerner.html Antibody that Transforms Bone Marrow Stem Cells Directly into Brain Cells]</ref><ref>Jia Xie, Hongkai Zhang, Kyungmoo Yea, and Richard A. Lerner (2013) [http://www.pnas.org/content/early/2013/04/23/1306263110.abstract Autocrine signaling based selection of combinatorial antibodies that transdifferentiate human stem cells] PNAS; {{doi|10.1073/pnas.1306263110}}</ref>


=== Conditionally reprogrammed cells ===
=== Conditionally reprogrammed cells ===


[[Richard Schlegel|Schlegel]] and Liu <ref name="Xuefeng">Xuefeng Liu, Virginie Ory, Sandra Chapman, et al. & Richard Schlegel ( 2012) ROCK Inhibitor and Feeder Cells Induce the Conditional Reprogramming of Epithelial Cells. The American Journal of Pathology, 180(2), 599-607 http://dx.doi.org/10.1016/j.ajpath.2011.10.036</ref> demonstrated that the combination of feeder cells <ref name="Rheinwald JG">Rheinwald JG, Green H. (1975) Serial cultivation of strains of human epidermal keratinocytes: the formation of keratinizing colonies from single cells. Cell. 1975 Nov;6(3):331-43</ref> (a review is given in<ref>[http://discovery.ucl.ac.uk/1306717/1/1306717.pdf Hiew, Y.-L. (2011) Examining the biological consequences of DNA damage caused by irradiated J2-3T3 fibroblast feeder cells and HPV16: characterisation of the biological functions of Mll. Doctoral thesis, UCL (University College London)]</ref> and<ref>Irena Szumiel (2012) Radiation hormesis: Autophagy and other cellular mechanisms International Journal of Radiation Biology. 88(9), 619-628 doi:10.3109/09553002.2012.699698</ref>) and a Rho kinase inhibitor (Y-27632) <ref>Hiroshi Kurosawa (2012) Application of Rho-associated protein kinase (ROCK) inhibitor to human pluripotent stem cells. Journal of Bioscience and Bioengineering, 114(6), 577–581 http://dx.doi.org/10.1016/j.jbiosc.2012.07.013</ref><ref name="Vogel JC">Terunuma A, Limgala RP, Park CJ, Choudhary I, Vogel JC. (2010) Efficient procurement of epithelial stem cells from human tissue specimens using a Rho-associated protein kinase inhibitor Y-27632. Tissue Eng Part A. ;16(4):1363-1368 doi: 10.1089/ten.tea.2009.0339</ref> induces normal and tumor epithelial cells from many tissues to proliferate
[[Richard Schlegel|Schlegel]] and Liu <ref name="Xuefeng">Xuefeng Liu, Virginie Ory, Sandra Chapman, et al. & Richard Schlegel ( 2012) ROCK Inhibitor and Feeder Cells Induce the Conditional Reprogramming of Epithelial Cells. The American Journal of Pathology, 180(2), 599-607 http://dx.doi.org/10.1016/j.ajpath.2011.10.036</ref> demonstrated that the combination of feeder cells <ref name="Rheinwald JG">Rheinwald JG, Green H. (1975) Serial cultivation of strains of human epidermal keratinocytes: the formation of keratinizing colonies from single cells. Cell. 1975 Nov;6(3):331-43</ref> (a review is given in<ref>[http://discovery.ucl.ac.uk/1306717/1/1306717.pdf Hiew, Y.-L. (2011) Examining the biological consequences of DNA damage caused by irradiated J2-3T3 fibroblast feeder cells and HPV16: characterisation of the biological functions of Mll. Doctoral thesis, UCL (University College London)]</ref> and<ref>{{cite doi|10.3109/09553002.2012.699698}}</ref>) and a Rho kinase inhibitor (Y-27632) <ref>Hiroshi Kurosawa (2012) Application of Rho-associated protein kinase (ROCK) inhibitor to human pluripotent stem cells. Journal of Bioscience and Bioengineering, 114(6), 577–581 http://dx.doi.org/10.1016/j.jbiosc.2012.07.013</ref><ref name="Vogel JC">{{cite doi|10.1089/ten.tea.2009.0339}}</ref> induces normal and tumor epithelial cells from many tissues to proliferate
indefinitely in vitro and this process occurs without the need for transduction of exogenous viral or cellular genes. These cells has been termed as "Conditionally Reprogrammed Cells (CRC)". The induction of CRCs is rapid and results from reprogramming of the entire cell population rather than the selection of a minor subpopulation. CRCs do not express high levels of proteins characteristic of iPSCs or embryonic stem cells (ESCs) (e.g., Sox2, Oct4, Nanog, or Klf4). This induction of CRCs is reversible, and removal of Y-27632 and feeders allows the cells to differentiate normally.<ref name="Xuefeng">Xuefeng Liu, Virginie Ory, Sandra Chapman, et al. & Richard Schlegel ( 2012) ROCK Inhibitor and Feeder Cells Induce the Conditional Reprogramming of Epithelial Cells. The American Journal of Pathology, 180(2), 599-607 http://dx.doi.org/10.1016/j.ajpath.2011.10.036</ref><ref name="Sandra">Sandra Chapman, Xuefeng Liu, Craig Meyers, Richard Schlegel, and Alison A. McBride. ( 2010) Human keratinocytes are efficiently immortalized by a Rho kinase inhibitor http://www.jci.org/articles/view/42297</ref><ref name="Schlegel">Suprynowicz F. A., Upadhyay G., Krawczyk E., et al. and Richard Schlegel. (2012) Conditionally reprogrammed cells represent a stem-like state of adult epithelial cells. PNAS, DOI: 10.1073/pnas.1213241109</ref> CRC technology can generate 2{{e|6}} cells in 5 to 6 days from needle biopsies, and can generate cultures from cryopreserved tissue and from fewer than four viable cells. CRCs retain a normal karyotype and remain nontumorigenic. This technique also efficiently establishes cell cultures from human and rodent tumors.<ref name="Xuefeng">Xuefeng Liu, Virginie Ory, Sandra Chapman, et al. & Richard Schlegel ( 2012) ROCK Inhibitor and Feeder Cells Induce the Conditional Reprogramming of Epithelial Cells. The American Journal of Pathology, 180(2), 599-607 http://dx.doi.org/10.1016/j.ajpath.2011.10.036</ref><ref name="Rimm">Seema Agarwal, David L. Rimm (2012) Making Every Cell Like HeLa: A Giant Step For Cell Culture. The American Journal of Pathology, 180( 2), 443-445 http://dx.doi.org/10.1016/j.ajpath.2011.12.001</ref><ref name="Lisanti">Lisanti MP, Tanowitz HB. (2012) Translational discoveries, personalized medicine, and living biobanks of the future. The American Journal of Pathology, 2012 Apr;180(4):1334-6 http://www.sciencedirect.com/science/article/pii/S0002944012000971</ref> The ability to rapidly generate many tumor cells from small biopsy specimens and frozen tissue provides significant opportunities for cell-based diagnostics and therapeutics (including chemosensitivity testing) and greatly expands the value of biobanking.<ref name="Xuefeng">Xuefeng Liu, Virginie Ory, Sandra Chapman, et al. & Richard Schlegel ( 2012) ROCK Inhibitor and Feeder Cells Induce the Conditional Reprogramming of Epithelial Cells. The American Journal of Pathology, 180(2), 599-607 http://dx.doi.org/10.1016/j.ajpath.2011.10.036</ref><ref name="Rimm">Seema Agarwal, David L. Rimm (2012) Making Every Cell Like HeLa: A Giant Step For Cell Culture. The American Journal of Pathology, 180( 2), 443-445 http://dx.doi.org/10.1016/j.ajpath.2011.12.001</ref><ref name="Lisanti">Lisanti MP, Tanowitz HB. (2012) Translational discoveries, personalized medicine, and living biobanks of the future. The American Journal of Pathology, 2012 Apr;180(4):1334-6 http://www.sciencedirect.com/science/article/pii/S0002944012000971</ref> Using conditionally reprogrammed cells technology, researchers were able to identify an effective therapy for a patient with a rare type of lung tumor.<ref name="Hang Yuan">Hang Yuan, Scott Myers, Jingang Wang, et al & Richard Schlegel. (2012) Use of Reprogrammed Cells to Identify Therapy for Respiratory Papillomatosis. New England Journal of Medicine; 367 (13): 1220-1227 DOI:10.1056/NEJMoa1203055</ref> In addition, the CRC method allows for the genetic manipulation of epithelial cells ex vivo and their subsequent evaluation in vivo in the same host. While their initial studies revealed that co-culturing of epithelial cells with Swiss 3T3 cells J2 was essential for the induction of CRCs, [[Richard Schlegel|Schlegel]] and Liu <ref name="Nancy">Palechor-Ceron N, Suprynowicz FA, Upadhyay G, Dakic A, Minas T, Simic V, Johnson M, Albanese C, Schlegel R, Liu X. (2013) Radiation Induces Diffusible Feeder Cell Factor(s) That Cooperate with ROCK Inhibitor to Conditionally Reprogram and Immortalize Epithelial Cells. Am J Pathol. 2013 Dec;183(6):1862-70. doi: 10.1016/j.ajpath.2013.08.009</ref> have now shown with transwell culture plates that physical contact between feeders and epithelial cells is not required for inducing CRCs and, more important, that irradiation of the feeder cells is required for this induction. Consistent with the transwell experiments, conditioned medium is shown to induce and maintain CRCs, which is accompanied by a concomitant increase of cellular telomerase activity. The activity of the conditioned medium correlated directly
indefinitely in vitro and this process occurs without the need for transduction of exogenous viral or cellular genes. These cells has been termed as "Conditionally Reprogrammed Cells (CRC)". The induction of CRCs is rapid and results from reprogramming of the entire cell population rather than the selection of a minor subpopulation. CRCs do not express high levels of proteins characteristic of iPSCs or embryonic stem cells (ESCs) (e.g., Sox2, Oct4, Nanog, or Klf4). This induction of CRCs is reversible, and removal of Y-27632 and feeders allows the cells to differentiate normally.<ref name="Xuefeng">{{cite doi|10.1016/j.ajpath.2011.10.036}}</ref><ref name="Sandra">Sandra Chapman, Xuefeng Liu, Craig Meyers, Richard Schlegel, and Alison A. McBride. ( 2010) Human keratinocytes are efficiently immortalized by a Rho kinase inhibitor http://www.jci.org/articles/view/42297</ref><ref name="Schlegel">{{cite doi|10.1073/pnas.1213241109}}</ref> CRC technology can generate 2{{e|6}} cells in 5 to 6 days from needle biopsies, and can generate cultures from cryopreserved tissue and from fewer than four viable cells. CRCs retain a normal karyotype and remain nontumorigenic. This technique also efficiently establishes cell cultures from human and rodent tumors.<ref name="Xuefeng">Xuefeng Liu, Virginie Ory, Sandra Chapman, et al. & Richard Schlegel ( 2012) ROCK Inhibitor and Feeder Cells Induce the Conditional Reprogramming of Epithelial Cells. The American Journal of Pathology, 180(2), 599-607 http://dx.doi.org/10.1016/j.ajpath.2011.10.036</ref><ref name="Rimm">Seema Agarwal, David L. Rimm (2012) Making Every Cell Like HeLa: A Giant Step For Cell Culture. The American Journal of Pathology, 180( 2), 443-445 http://dx.doi.org/10.1016/j.ajpath.2011.12.001</ref><ref name="Lisanti">Lisanti MP, Tanowitz HB. (2012) Translational discoveries, personalized medicine, and living biobanks of the future. The American Journal of Pathology, 2012 Apr;180(4):1334-6 http://www.sciencedirect.com/science/article/pii/S0002944012000971</ref> The ability to rapidly generate many tumor cells from small biopsy specimens and frozen tissue provides significant opportunities for cell-based diagnostics and therapeutics (including chemosensitivity testing) and greatly expands the value of biobanking.<ref name="Xuefeng">Xuefeng Liu, Virginie Ory, Sandra Chapman, et al. & Richard Schlegel ( 2012) ROCK Inhibitor and Feeder Cells Induce the Conditional Reprogramming of Epithelial Cells. The American Journal of Pathology, 180(2), 599-607 http://dx.doi.org/10.1016/j.ajpath.2011.10.036</ref><ref name="Rimm">Seema Agarwal, David L. Rimm (2012) Making Every Cell Like HeLa: A Giant Step For Cell Culture. The American Journal of Pathology, 180( 2), 443-445 http://dx.doi.org/10.1016/j.ajpath.2011.12.001</ref><ref name="Lisanti">Lisanti MP, Tanowitz HB. (2012) Translational discoveries, personalized medicine, and living biobanks of the future. The American Journal of Pathology, 2012 Apr;180(4):1334-6 http://www.sciencedirect.com/science/article/pii/S0002944012000971</ref> Using conditionally reprogrammed cells technology, researchers were able to identify an effective therapy for a patient with a rare type of lung tumor.<ref name="Hang Yuan">{{cite doi|10.1056/NEJMoa1203055}}</ref> In addition, the CRC method allows for the genetic manipulation of epithelial cells ex vivo and their subsequent evaluation in vivo in the same host. While their initial studies revealed that co-culturing of epithelial cells with Swiss 3T3 cells J2 was essential for the induction of CRCs, [[Richard Schlegel|Schlegel]] and Liu <ref name="Nancy">{{cite doi|10.1016/j.ajpath.2013.08.009}}</ref> have now shown with transwell culture plates that physical contact between feeders and epithelial cells is not required for inducing CRCs and, more important, that irradiation of the feeder cells is required for this induction. Consistent with the transwell experiments, conditioned medium is shown to induce and maintain CRCs, which is accompanied by a concomitant increase of cellular telomerase activity. The activity of the conditioned medium correlated directly
with radiation-induced apoptosis of the feeder cells. Thus, conditional reprogramming of epithelial cells is mediated by a combination of Y-27632 and a soluble factor(s) released by apoptotic feeder cells.<ref name="Nancy">Palechor-Ceron N, Suprynowicz FA, Upadhyay G, Dakic A, Minas T, Simic V, Johnson M, Albanese C, Schlegel R, Liu X. (2013) Radiation Induces Diffusible Feeder Cell Factor(s) That Cooperate with ROCK Inhibitor to Conditionally Reprogram and Immortalize Epithelial Cells. Am J Pathol. 2013 Dec;183(6):1862-70. doi: 10.1016/j.ajpath.2013.08.009</ref>
with radiation-induced apoptosis of the feeder cells. Thus, conditional reprogramming of epithelial cells is mediated by a combination of Y-27632 and a soluble factor(s) released by apoptotic feeder cells.<ref name="Nancy">{{cite doi|10.1016/j.ajpath.2013.08.009}}</ref>


A different approach to obtain conditionally reprogrammed cells is to inhibit [[CD47]] - a [[membrane protein]] that is the [[thrombospondin-1]] receptor. It was shown that loss of CD47 permits sustained proliferation of primary murine endothelial cells, increases asymmetric division, and enables these cells to spontaneously reprogram to form multipotent [[embryoid body]]-like clusters. CD47 knockdown acutely increases mRNA levels of c-Myc and other stem cell transcription factors in cells in vitro and in vivo. It is obvious that thrombospondin-1 is a key environmental signal that inhibits stem cell self-renewal via CD47. Thus, CD47 antagonists enable cell self-renewal and reprogramming by overcoming negative regulation of c-Myc and other stem cell transcription factors.<ref>Sukhbir Kaur, David R. Soto-Pantoja, Erica V. Stein et al. & David D. Roberts.( 2013) [http://www.nature.com/srep/2013/130417/srep01673/full/srep01673.html Thrombospondin-1 Signaling through CD47 Inhibits Self-renewal by Regulating c-Myc and Other Stem Cell Transcription Factors]. Scientific Reports; 3, Article number: 1673 DOI:10.1038/srep01673</ref> Interesting to note that in vivo blockade of CD47 using an antisense [[morpholino]] increases survival of mice exposed to lethal total body irradiation due to increased proliferative capacity of bone marrow derived cells and radioprotection of radiosensitive gastrointestinal tissues.<ref>Soto-Pantoja, D. R., Ridnour, L. A., Wink, D. A. & Roberts, D. D. (2013) Blockade of CD47 increases survival of mice exposed to lethal total body irradiation. Sci Rep 3, 1038 doi:10.1038/srep01038</ref>
A different approach to obtain conditionally reprogrammed cells is to inhibit [[CD47]] - a [[membrane protein]] that is the [[thrombospondin-1]] receptor. It was shown that loss of CD47 permits sustained proliferation of primary murine endothelial cells, increases asymmetric division, and enables these cells to spontaneously reprogram to form multipotent [[embryoid body]]-like clusters. CD47 knockdown acutely increases mRNA levels of c-Myc and other stem cell transcription factors in cells in vitro and in vivo. It is obvious that thrombospondin-1 is a key environmental signal that inhibits stem cell self-renewal via CD47. Thus, CD47 antagonists enable cell self-renewal and reprogramming by overcoming negative regulation of c-Myc and other stem cell transcription factors.<ref>Sukhbir Kaur, David R. Soto-Pantoja, Erica V. Stein et al. & David D. Roberts.( 2013) [http://www.nature.com/srep/2013/130417/srep01673/full/srep01673.html Thrombospondin-1 Signaling through CD47 Inhibits Self-renewal by Regulating c-Myc and Other Stem Cell Transcription Factors]. Scientific Reports; 3, Article number: 1673 DOI:10.1038/srep01673</ref> Interesting to note that in vivo blockade of CD47 using an antisense [[morpholino]] increases survival of mice exposed to lethal total body irradiation due to increased proliferative capacity of bone marrow derived cells and radioprotection of radiosensitive gastrointestinal tissues.<ref>{{cite doi|10.1038/srep01038}}</ref>


=== Indirect lineage conversion ===
=== Indirect lineage conversion ===
Line 132: Line 132:
=== Outer membrane glycoprotein ===
=== Outer membrane glycoprotein ===


A common feature of pluripotent stem cells, taken from different sources, is the specific nature of protein [[glycosylation]] of their outer membrane. That distinguishes them from most ( with the exception of [[white blood cells]] ) nonpluripotent cells.<ref>Wang, Y. C., Nakagawa, M., Garitaonandia. et al. & Loring, J. F. (2011). Specific lectin biomarkers for isolation of human pluripotent stem cells identified through array-based glycomic analysis.Cell research, 21 (11 ) , 1551-1563 . doi: 10.1038/cr.2011.148</ref>
A common feature of pluripotent stem cells, taken from different sources, is the specific nature of protein [[glycosylation]] of their outer membrane. That distinguishes them from most ( with the exception of [[white blood cells]] ) nonpluripotent cells.<ref>{{cite doi|10.1038/cr.2011.148}}</ref>
Obviously, changes in the glycosylation of outer membrane proteins are markers of the state of the cells connected in some way with pluripotency and differentiation. The "shift" in the nature of glycosylation, apparently, is not just the result of the initialization of some genes expression, but the mechanism performing the role of an important regulator of genes involved in the acquisition and maintenance of the undifferentiated state.<ref>Hasehira, K., Tateno, H., Onuma, Y., Ito, Y., Asashima, M., & Hirabayashi, J. ( 2012). Structural and Quantitative Evidence for Dynamic Glycome Shift on Production of Induced Pluripotent Stem Cells. Molecular & Cellular Proteomics, 11 (12 ) 1913-1923 . doi: 10.1074/mcp.M112.020586</ref>
Obviously, changes in the glycosylation of outer membrane proteins are markers of the state of the cells connected in some way with pluripotency and differentiation. The "shift" in the nature of glycosylation, apparently, is not just the result of the initialization of some genes expression, but the mechanism performing the role of an important regulator of genes involved in the acquisition and maintenance of the undifferentiated state.<ref>{{cite doi|10.1074/mcp.M112.020586}}</ref>
For example, it is shown that activation of the glycoprotein ACA,<ref>Becker-Kojic, Z. A., & Terness, P. (2002). A novel human erythrocyte GPI anchored glycoprotein ACA. Isolation, purification, primary structure determination, molecular parameters of its lipid structure. . Journal of Biological Chemistry, 277, 40472-40478 . doi: 10.1074/jbc.M202416200</ref>
For example, it is shown that activation of the glycoprotein ACA,<ref>{{cite doi|10.1074/jbc.M202416200}}</ref>
linking glycosylphosphatidylinositol on the surface of the progenitor cells in human peripheral blood, induces increased expression of genes [[Wnt signaling pathway|Wnt]], [[Notch-1]], [[BMI1]] and [[HOXB4]] through a signaling cascade [[PI3K]]/[[Akt]]/[[Mammalian target of rapamycin|mTor]]/[[PTEN (gene)|PTEN]], and promotes the formation of self-renewing population of hematopoietic stem cells<ref>ZABecker-Kojič, J . Ureña-Peralta, R.Saffrich et al. & M.Stojkovič ( 2013 ) A new glycoprotein ACA - the main regulator of human hematopoiesis . CELL TECHNOLOGIES IN BIOLOGY AND MEDICINE , 9 (2 ) , 69-84 (rus.)</ref>
linking glycosylphosphatidylinositol on the surface of the progenitor cells in human peripheral blood, induces increased expression of genes [[Wnt signaling pathway|Wnt]], [[Notch-1]], [[BMI1]] and [[HOXB4]] through a signaling cascade [[PI3K]]/[[Akt]]/[[Mammalian target of rapamycin|mTor]]/[[PTEN (gene)|PTEN]], and promotes the formation of self-renewing population of hematopoietic stem cells<ref>ZABecker-Kojič, J . Ureña-Peralta, R.Saffrich et al. & M.Stojkovič ( 2013 ) A new glycoprotein ACA - the main regulator of human hematopoiesis . CELL TECHNOLOGIES IN BIOLOGY AND MEDICINE , 9 (2 ) , 69-84 (rus.)</ref>
Furthermore, it is shown that dedifferentiation of progenitor cells induced by ACA- dependent signaling pathway leads to ACA- induced pluripotent stem cells, capable of differentiating in vitro into cells of all three [[germ layers]].<ref>ZABecker-Kojič, JRUreña-Peralta, I.Zipančić, et al. & M.Stojkovič ( 2013 ) Activation of surface glycoprotein ACA induced pluripotent hematopoietic progenitor cells. CELL TECHNOLOGIES IN BIOLOGY AND MEDICINE , 9 (2 ) , 85-101 (rus.)</ref>
Furthermore, it is shown that dedifferentiation of progenitor cells induced by ACA- dependent signaling pathway leads to ACA- induced pluripotent stem cells, capable of differentiating in vitro into cells of all three [[germ layers]].<ref>ZABecker-Kojič, JRUreña-Peralta, I.Zipančić, et al. & M.Stojkovič ( 2013 ) Activation of surface glycoprotein ACA induced pluripotent hematopoietic progenitor cells. CELL TECHNOLOGIES IN BIOLOGY AND MEDICINE , 9 (2 ) , 85-101 (rus.)</ref>
Line 159: Line 159:
=== Reprogramming through a physical approach ===
=== Reprogramming through a physical approach ===


Cell adhesion protein [[CDH1 (gene)|E-cadherin]] is indispensable for a robust pluripotent phenotype.<ref>Torben Redmer, Sebastian Diecke, Tamara Grigoryan, Angel Quiroga-Negreira, Walter Birchmeier, Daniel Besser (2011) E-cadherin is crucial for embryonic stem cell pluripotency and can replace OCT4 during somatic cell reprogramming. EMBO reports , 12, 720 - 726, doi:10.1038/embor.2011.88</ref> During reprogramming for iPS cell generation, [[CDH2|N-cadherin]] can replace function of E-cadherin.<ref>Bedzhov, I., Alotaibi, H., Basilicata, M. F. et al. & Stemmler, M. P. (2013). Adhesion, but not a specific cadherin code, is indispensable for ES cell and induced pluripotency. Stem cell research, 11(3), 1250-1263. http://dx.doi.org/10.1016/j.scr.2013.08.009</ref> These functions of cadherins obviously are not directly related to adhesion because, according to Guannan Su et al.<ref>Guannan Su, Yannan Zhao, Jianshu Wei, et al. & Jianwu Dai (2013) Direct conversion of fibroblasts into neural progenitor-like cells by forced growth into 3D spheres on low attachment surfaces. Biomaterials, 34(24), 5897–5906 http://dx.doi.org/10.1016/j.biomaterials.2013.04.040</ref> sphere morphology helps maintaining the stemness of stem cells. Moreover, 3D sphere formation, due to forced growth of cells on low attachment surface, sometimes induces reprogramming. For example, neural progenitor cells can be generated from fibroblasts directly through a physical approach without introducing exogenous reprogramming factors.
Cell adhesion protein [[CDH1 (gene)|E-cadherin]] is indispensable for a robust pluripotent phenotype.<ref>{{cite doi|10.1038/embor.2011.88}}</ref> During reprogramming for iPS cell generation, [[CDH2|N-cadherin]] can replace function of E-cadherin.<ref>{{cite doi|10.1016/j.scr.2013.08.009}}</ref> These functions of cadherins obviously are not directly related to adhesion because, according to Guannan Su et al.<ref>Guannan Su, Yannan Zhao, Jianshu Wei, et al. & Jianwu Dai (2013) Direct conversion of fibroblasts into neural progenitor-like cells by forced growth into 3D spheres on low attachment surfaces. Biomaterials, 34(24), 5897–5906 http://dx.doi.org/10.1016/j.biomaterials.2013.04.040</ref> sphere morphology helps maintaining the stemness of stem cells. Moreover, 3D sphere formation, due to forced growth of cells on low attachment surface, sometimes induces reprogramming. For example, neural progenitor cells can be generated from fibroblasts directly through a physical approach without introducing exogenous reprogramming factors.


Physical cues, in the form of parallel microgrooves on the surface of cell-adhesive substrates, can replace the effects of small-molecule epigenetic modifiers and significantly improve reprogramming efficiency. The mechanism relies on the mechanomodulation of the cells’ epigenetic state. To be specific, as the authors of this study believe: "decreased histone deacetylase activity and upregulation of the expression of WD repeat domain 5 (WDR5)—a subunit of H3 methyltranferase—by microgrooved surfaces lead to increased histone H3 acetylation and methylation". Nanofibrous scaffolds with aligned fibre orientation produce effects similar to those produced by microgrooves, suggesting that changes in cell morphology may be responsible for modulation of the epigenetic state.<ref>Timothy L. Downing, Jennifer Soto, Constant Morez, Timothee Houssin, Ashley Fritz, Falei Yuan, Julia Chu, Shyam Patel, David V. Schaffer, Song Li.(2013) Biophysical regulation of epigenetic state and cell reprogramming. Nature Materials, DOI: 10.1038/nmat3777</ref>
Physical cues, in the form of parallel microgrooves on the surface of cell-adhesive substrates, can replace the effects of small-molecule epigenetic modifiers and significantly improve reprogramming efficiency. The mechanism relies on the mechanomodulation of the cells’ epigenetic state. To be specific, as the authors of this study believe: "decreased histone deacetylase activity and upregulation of the expression of WD repeat domain 5 (WDR5)—a subunit of H3 methyltranferase—by microgrooved surfaces lead to increased histone H3 acetylation and methylation". Nanofibrous scaffolds with aligned fibre orientation produce effects similar to those produced by microgrooves, suggesting that changes in cell morphology may be responsible for modulation of the epigenetic state.<ref>{{cite doi|10.1038/nmat3777}}</ref>


Mouse embryonic stem cells (mESCs) undergo self-renewal in the presence of the [[cytokine]], [[leukemia inhibitory factor]] (LIF). Following LIF withdrawal, mESCs differentiate, and this is accompanied by an increase in cell–substratum adhesion and cell spreading. Restricted cell spreading in the absence of LIF by either culturing mESCs on chemically defined, weakly adhesive biomaterial substrates, or by manipulating the [[cytoskeleton]] allowed to keep the cells in an undifferentiated and pluripotent state. The effect of restricted cell spreading on mESC self-renewal is not mediated by increased intercellular adhesion, as evidenced by the observations that inhibition of mESC adhesion using a function blocking anti E-cadherin antibody or siRNA do not promote differentiation.<ref>Patricia Murray, Marina Prewitz, Isabel Hopp, et al. (2013) The self-renewal of mouse embryonic stem cells is regulated by cell–substratum adhesion and cell spreading. The International Journal of Biochemistry & Cell Biology, 45(11), 2698–2705 http://dx.doi.org/10.1016/j.biocel.2013.07.001</ref>
Mouse embryonic stem cells (mESCs) undergo self-renewal in the presence of the [[cytokine]], [[leukemia inhibitory factor]] (LIF). Following LIF withdrawal, mESCs differentiate, and this is accompanied by an increase in cell–substratum adhesion and cell spreading. Restricted cell spreading in the absence of LIF by either culturing mESCs on chemically defined, weakly adhesive biomaterial substrates, or by manipulating the [[cytoskeleton]] allowed to keep the cells in an undifferentiated and pluripotent state. The effect of restricted cell spreading on mESC self-renewal is not mediated by increased intercellular adhesion, as evidenced by the observations that inhibition of mESC adhesion using a function blocking anti E-cadherin antibody or siRNA do not promote differentiation.<ref>Patricia Murray, Marina Prewitz, Isabel Hopp, et al. (2013) The self-renewal of mouse embryonic stem cells is regulated by cell–substratum adhesion and cell spreading. The International Journal of Biochemistry & Cell Biology, 45(11), 2698–2705 http://dx.doi.org/10.1016/j.biocel.2013.07.001</ref>


The possible mechanisms of stem cell fate predetermining by physical interactions with the extracellular matrix see in reviews:<ref>Guilak, F., Cohen, D. M., Estes, B. T., et al. & Chen, C. S. (2009) [http://www.ncbi.nlm.nih.gov/pmc/articles/PMC2768283/ Control of stem cell fate by physical interactions with the extracellular matrix]. Cell stem cell, 5(1), 17-26. doi: 10.1016/j.stem.2009.06.016</ref><ref>Worley, K., Certo, A., & Wan, L. Q. (2013). Geometry–Force Control of Stem Cell Fate. BioNanoScience, 3(1), 43-51. DOI: 10.1007/s12668-012-0067-0</ref>
The possible mechanisms of stem cell fate predetermining by physical interactions with the extracellular matrix see in reviews:<ref>Guilak, F., Cohen, D. M., Estes, B. T., et al. & Chen, C. S. (2009) [http://www.ncbi.nlm.nih.gov/pmc/articles/PMC2768283/ Control of stem cell fate by physical interactions with the extracellular matrix]. Cell stem cell, 5(1), 17-26. doi: 10.1016/j.stem.2009.06.016</ref><ref>{{cite doi|10.1007/s12668-012-0067-0}}</ref>


=== Induced neural stem cells ===
=== Induced neural stem cells ===


Stroke and many neurodegenerative disorders such a: Parkinson's disease, Alzheimer’s disease, amyotrophic lateral sclerosis need cell replacement therapy. The successful use of converted neural cells (cNs) in transplantations open a new avenue to treat such diseases.<ref name="Qu">Zhang W, Duan S, Li Y. et al. and Jing Qu (2012) Converted neural cells: induced to a cure? Protein Cell; 3(2), 91–97. DOI: 10.1007/s13238-012-2029-2</ref> Nevertheless, induced neurons (iNs), directly converted from fibroblasts are terminally committed neurons that exhibit very limited proliferative ability and may not provide enough autologous donor cells for transplantation.<ref name="Yang">Yang N, Ng YH, Pang ZP, Sudhof TC, Wernig M.(2011) Induced neuronal cells: how to make and define a neuron. Cell Stem Cell; 9(6), 517–525. doi: 10.1016/j.stem.2011.11.015</ref> The generation of self-renewing induced neural stem cells (iNSCs) provides additional advantages over iNs for both basic research and clinical applications.<ref name="Ernesto">Lujan E, Chanda S, Ahlenius H, Sudhof TC, Wernig M.(2012) Direct conversion of mouse fibroblasts to self-renewing, tripotent neural precursor cells. PNAS; 109(7), 2527–2532. doi: 10.1073/pnas.1121003109</ref><ref name="Marc">Thier M, Wörsdörfer P, Lakes YB, et al. Direct conversion of fibroblasts into stably expandable neural stem cells. Cell Stem Cell 2012; 10(4),473–479 doi: 10.1016/j.stem.2012.03.003</ref><ref name="Han">Han DW, Tapia N., Hermann A., et al. & Schöler H.R. (2012) Direct Reprogramming of Fibroblasts into Neural Stem Cells by Defined Factors. Cell Stem Cell, 10(4), 465–472, doi: 10.1016/j.stem.2012.02.021</ref><ref name="Wu">Sheng C, Zheng Q, Wu J, et al. and Qi Zhou (2012) Generation of dopaminergic neurons directly from mouse fibroblasts and fibroblast-derived neural progenitors. Cell Res; 22:769–772. doi:10.1038/cr.2012.32</ref><ref name="Maucksch, C. 2012">Maucksch, C., E. Firmin, et al. (2012). "Non-viral generation of neural precursor-like cells from adult human fibroblasts." J Stem Cells Regen Med 8(3): 1-9.</ref> For example, under specific growth conditions, mouse fibroblasts can be reprogrammed with a single factor, Sox2, to form induced neural stem cells (iNSCs) that self-renew in culture and after transplantation can survive, integrate and do not form tumors in mouse brains.<ref name="Yadong">Ring K L, Leslie M. Tong L M, Balestra M E et al. & Yadong Huang (2012) Direct Reprogramming of Mouse and Human Fibroblasts into Multipotent Neural Stem Cells with a Single Factor. Cell Stem Cell, 11(1), 100-109 doi: 10.1016/j.stem.2012.05.018</ref> Induced neural stem cells can also be derived from adult human fibroblasts by non-viral techniques, thus offering a safe method to generate iNSCs for autologous transplantation or for the development of cell-based disease models.<ref name="Maucksch, C. 2012"/> Methods of direct transformation of somatic cells into induced neural stem cells differ in their technique as shown in the review.<ref name="Belmonte">Liu G-H , Yi F, Suzuki K, Qu J. and Izpisua Belmonte J C. (2012) Induced neural stem cells: a new tool for studying neural development and neurological disorders. Cell Research 22, 1087–1091. doi:10.1038/cr.2012.73</ref>
Stroke and many neurodegenerative disorders such a: Parkinson's disease, Alzheimer’s disease, amyotrophic lateral sclerosis need cell replacement therapy. The successful use of converted neural cells (cNs) in transplantations open a new avenue to treat such diseases.<ref name="Qu">{{cite doi|10.1007/s13238-012-2029-2}}</ref> Nevertheless, induced neurons (iNs), directly converted from fibroblasts are terminally committed neurons that exhibit very limited proliferative ability and may not provide enough autologous donor cells for transplantation.<ref name="Yang">{{cite doi|10.1016/j.stem.2011.11.015}}</ref> The generation of self-renewing induced neural stem cells (iNSCs) provides additional advantages over iNs for both basic research and clinical applications.<ref name="Ernesto">{{cite doi|10.1073/pnas.1121003109}}</ref><ref name="Marc">{{cite doi|10.1016/j.stem.2012.03.003}}</ref><ref name="Han">{{cite doi|10.1016/j.stem.2012.02.021}}</ref><ref name="Wu">{{cite doi|10.1038/cr.2012.32}}</ref><ref name="Maucksch, C. 2012">Maucksch, C., E. Firmin, et al. (2012). "Non-viral generation of neural precursor-like cells from adult human fibroblasts." J Stem Cells Regen Med 8(3): 1-9.</ref> For example, under specific growth conditions, mouse fibroblasts can be reprogrammed with a single factor, Sox2, to form induced neural stem cells (iNSCs) that self-renew in culture and after transplantation can survive, integrate and do not form tumors in mouse brains.<ref name="Yadong">{{cite doi|10.1016/j.stem.2012.05.018}}</ref> Induced neural stem cells can also be derived from adult human fibroblasts by non-viral techniques, thus offering a safe method to generate iNSCs for autologous transplantation or for the development of cell-based disease models.<ref name="Maucksch, C. 2012"/> Methods of direct transformation of somatic cells into induced neural stem cells differ in their technique as shown in the review.<ref name="Belmonte">{{cite doi|10.1038/cr.2012.73}}</ref>


Some data provide proof of principle that it is possible, directly in the brain, to convert transplanted human fibroblasts and human astrocytes, which are engineered to express inducible forms of neural reprogramming genes, into neurons, when reprogramming genes (Ascl11, Brn2a, and Myt1l) are activated after transplantation using a drug in the animals’ drinking water.<ref>Olof Torper, Ulrich Pfisterer, Daniel A. Wolf, et al. and Malin Parmar (2013) Generation of induced neurons via direct conversion in vivo. PNAS, DOI:10.1073/pnas.1303829110</ref> It was also shown that endogenous mouse astrocytes can be directly converted into neural nuclei (NeuN)-expressing neurons in situ.
Some data provide proof of principle that it is possible, directly in the brain, to convert transplanted human fibroblasts and human astrocytes, which are engineered to express inducible forms of neural reprogramming genes, into neurons, when reprogramming genes (Ascl11, Brn2a, and Myt1l) are activated after transplantation using a drug in the animals’ drinking water.<ref>{{cite doi|10.1073/pnas.1303829110}}</ref> It was also shown that endogenous mouse astrocytes can be directly converted into neural nuclei (NeuN)-expressing neurons in situ.


Future studies will show which of these approaches proved to be most appropriate for clinic.
Future studies will show which of these approaches proved to be most appropriate for clinic.
Line 177: Line 177:
=== Oligodendrocyte precursor cells ===
=== Oligodendrocyte precursor cells ===


Without myelin to insulate neurons, signals sent down nerve cell axons quickly lose power. Diseases that attack myelin, such as multiple sclerosis, result in nerve signals that are not efficient, because cannot propagate to nerve endings, and as a consequence to cognitive, motor, and sensory problems. Transplantation of oligodendrocyte precursor cells (OPCs), which can successfully create myelin sheaths around nerve cells, is a promising potential therapeutic strategy for diseases affecting myelin. However, there was no available source of engraftable OPCs. Therefore, direct lineage conversion of mouse and rat fibroblasts into oligodendroglial cells by forced expression of either eight<ref>Fadi J Najm, Angela M Lager, Anita Zaremba, et al. & Paul J Tesar (2013) Transcription factor–mediated reprogramming of fibroblasts to expandable, myelinogenic oligodendrocyte progenitor cells. Nature Biotechnology, doi:10.1038/nbt.2561</ref> or of the three<ref>Nan Yang, J Bradley Zuchero, Henrik Ahlenius, et al. & Marius Wernig (2013) Generation of oligodendroglial cells by direct lineage conversion. Nature Biotechnology, doi:10.1038/nbt.2564</ref> transcription factors Sox10, Olig2 and Zfp536, laid the groundwork for therapies of a wide array of myelin disorders and spinal cord injury.
Without myelin to insulate neurons, signals sent down nerve cell axons quickly lose power. Diseases that attack myelin, such as multiple sclerosis, result in nerve signals that are not efficient, because cannot propagate to nerve endings, and as a consequence to cognitive, motor, and sensory problems. Transplantation of oligodendrocyte precursor cells (OPCs), which can successfully create myelin sheaths around nerve cells, is a promising potential therapeutic strategy for diseases affecting myelin. However, there was no available source of engraftable OPCs. Therefore, direct lineage conversion of mouse and rat fibroblasts into oligodendroglial cells by forced expression of either eight<ref>{{cite doi|10.1038/nbt.2561}}</ref> or of the three<ref>{{cite doi|10.1038/nbt.2564}}</ref> transcription factors Sox10, Olig2 and Zfp536, laid the groundwork for therapies of a wide array of myelin disorders and spinal cord injury.


=== Induced cardiomyocytes ===
=== Induced cardiomyocytes ===


Development of regenerative therapeutic strategies to reverse the progression of advanced [[heart failure]] is one of the most urgent clinical needs of this century. Cell-based in vivo therapies may provide a transformative approach to augment vascular and muscle growth and to prevent non-contractile scar formation by delivering transcription factors<ref name="Li">Li Qian, Yu Huang, C. Ian Spencer, Amy Foley, Vasanth Vedantham, Lei Liu, Simon J. Conway, Ji-dong Fu & Deepak Srivastava. (2012) In vivo reprogramming of murine cardiac fibroblasts into induced cardiomyocytes. Nature, Nature; 485, 593–598. DOI: 10.1038/nature11044</ref> or microRNAs<ref name="Dzau"/> to the heart.<ref name="Chunhui">Chunhui (2012) Turning cardiac fibroblasts into cardiomyocytes in vivo Trends in Molecular Medicine, doi:10.1016/j.molmed.2012.06.009</ref> For example: Qian L. et al. & Srivastava D. demonstrate that cardiac fibroblasts, which represent 50% of the cells in the mammalian heart, can be reprogrammed into [[Cardiac muscle|cardiomyocyte]]-like cells in vivo by local delivery of cardiac core transcription factors ( GATA4, MEF2C, TBX5 and for improved reprogramming plus ESRRG, MESP1, Myocardin and ZFPM2) after coronary ligation.<ref name="Li">Li Qian, Yu Huang, C. Ian Spencer, Amy Foley, Vasanth Vedantham, Lei Liu, Simon J. Conway, Ji-dong Fu & Deepak Srivastava. (2012) In vivo reprogramming of murine cardiac fibroblasts into induced cardiomyocytes. Nature, Nature; 485, 593–598. DOI:10.1038/nature11044</ref><ref>Ji-Dong Fu, Nicole R. Stone, Lei Liu, et al. & Deepak Srivastava (2013) Direct Reprogramming of Human Fibroblasts toward a Cardiomyocyte-like State. Stem Cell Reports, doi: 10.1016/j.stemcr.2013.07.005</ref> These results implicated therapies that can directly remuscularize the heart without the need for cell transplantation. However, the efficiency of direct cardiac reprogramming in vivo by overexpression of cardiac core transcription factors in cardiac fibroblasts turned out to be very low and phenotype of received cardiomyocyte-like cells does not resemble those of a bona fide mature cardiomyocyte. Furthermore, transplantation of cardiac transcription factors into injured murine hearts resulted in poor cell survival and minimal expression of cardiac genes.<ref name="Sean">Chen J X., Krane M, Deutsch M-A, et al. and Sean M. Wu (2012)Inefficient Reprogramming of Fibroblasts into Cardiomyocytes Using Gata4, Mef2c, and Tbx5. Circulation Research.;111: 50-55, doi:10.1161/CIRCRESAHA.112.270264</ref> So, further technical improvements are needed to make this technology more applicable in situ.
Development of regenerative therapeutic strategies to reverse the progression of advanced [[heart failure]] is one of the most urgent clinical needs of this century. Cell-based in vivo therapies may provide a transformative approach to augment vascular and muscle growth and to prevent non-contractile scar formation by delivering transcription factors<ref name="Li">{{cite H10.1038/nature11044}}</ref> or microRNAs<ref name="Dzau"/> to the heart.<ref name="Chunhui">{{cite doi|10.1016/j.molmed.2012.06.009}}</ref> For example: Qian L. et al. & Srivastava D. demonstrate that cardiac fibroblasts, which represent 50% of the cells in the mammalian heart, can be reprogrammed into [[Cardiac muscle|cardiomyocyte]]-like cells in vivo by local delivery of cardiac core transcription factors ( GATA4, MEF2C, TBX5 and for improved reprogramming plus ESRRG, MESP1, Myocardin and ZFPM2) after coronary ligation.<ref name="Li">{{cite doi|10.1038/nature11044}}</ref><ref>{{cite doi|10.1016/j.stemcr.2013.07.005}}</ref> These results implicated therapies that can directly remuscularize the heart without the need for cell transplantation. However, the efficiency of direct cardiac reprogramming in vivo by overexpression of cardiac core transcription factors in cardiac fibroblasts turned out to be very low and phenotype of received cardiomyocyte-like cells does not resemble those of a bona fide mature cardiomyocyte. Furthermore, transplantation of cardiac transcription factors into injured murine hearts resulted in poor cell survival and minimal expression of cardiac genes.<ref name="Sean">{{cite doi|10.1161/CIRCRESAHA.112.270264}}</ref> So, further technical improvements are needed to make this technology more applicable in situ.
Meanwhile, there have been some advances in the methods of obtaining cardiac myocytes in vitro.<ref name="Paul">Paul W. Burridge, Gordon Keller, Joseph D. Gold, Joseph C. Wu (2012) Production of De Novo Cardiomyocytes: Human Pluripotent Stem Cell Differentiation and Direct Reprogramming. Review Article. Cell Stem Cell, 10 (1), 16-28</ref> For example, Carpenter et al. demonstrated efficient cardiac differentiation of human iPS cells that gave rise to progenitors that were retained within the infarcted rat heart, and reduced remodeling of the heart after ischemic damage.<ref name="Carpenter">Carpenter L. et al. and Watt S. M. (2012) Efficient Differentiation of Human Induced Pluripotent Stem Cells Generates Cardiac Cells That Provide Protection Following Myocardial Infarction in the Rat. Stem Cells and Development. 21 (6): 977-986. doi: 10.1089/scd.2011.0075.</ref>
Meanwhile, there have been some advances in the methods of obtaining cardiac myocytes in vitro.<ref name="Paul">Paul W. Burridge, Gordon Keller, Joseph D. Gold, Joseph C. Wu (2012) Production of De Novo Cardiomyocytes: Human Pluripotent Stem Cell Differentiation and Direct Reprogramming. Review Article. Cell Stem Cell, 10 (1), 16-28</ref> For example, Carpenter et al. demonstrated efficient cardiac differentiation of human iPS cells that gave rise to progenitors that were retained within the infarcted rat heart, and reduced remodeling of the heart after ischemic damage.<ref name="Carpenter">{{cite doi|10.1089/scd.2011.0075}}</ref>
Furthermore, in ischaemic cardiomyopathy, caused by the murine infarction model, targeted iPS cell transplantation synchronized failing ventricles, offering a regenerative strategy to achieve resynchronization and protection from decompensation by dint of improved left ventricular conduction and contractility, reduced scar, and reversal of structural remodelling.<ref>Satsuki Yamada, Timothy J. Nelson, Garvan C. Kane et al. & Andre Terzic (2013) iPS Cell Intervention Rescues Wall Motion Disparity Achieving Biological Cardiac Resynchronization Post-Infarction.The Journal of Physiology, 591, 4335-4349.; DOI:10.1113/jphysiol.2013.252288</ref>
Furthermore, in ischaemic cardiomyopathy, caused by the murine infarction model, targeted iPS cell transplantation synchronized failing ventricles, offering a regenerative strategy to achieve resynchronization and protection from decompensation by dint of improved left ventricular conduction and contractility, reduced scar, and reversal of structural remodelling.<ref>{{cite doi|10.1113/jphysiol.2013.252288}}</ref>
Palecek S P et al., have developed a protocol for generating almost pure populations of cardiomyocytes (up to 98% cardiomyocytes) from human pluripotent stem cells simply by modulating canonical [[Wnt signaling pathway]] at defined time points in the differentiation process, using readily accessible small molecule compounds.<ref name="Palecek">Lian X, Hsiao C, Gisela Wilson, et al and Palecek S P. (2012) Robust cardiomyocyte differentiation from human pluripotent stem cells via temporal modulation of canonical Wnt signaling. PNAS 2012 109 (27) E1848-E1857, doi: 10.1073/pnas.1200250109.</ref> Discovery of the mechanisms controlling the formation of cardiomyocytes led to the development of the drug (ITD-1), which effectively clears the cell surface from [[Transforming growth factor beta|TGF-β]] receptor type II and selectively inhibits the intracellular TGF-β signaling and, thus, selectively enhances the differentiation of uncommitted [[mesoderm]] to cardiomyocytes, but not to vascular smooth muscle and endothelial cells.<ref name="Mercola">Willems E, Cabral-Teixeira J, et al. & Mark Mercola. (2012) Small Molecule-Mediated TGF-β Type II Receptor Degradation Promotes Cardiomyogenesis in Embryonic Stem Cells. Cell Stem Cell,; 11 (2): 242-252 DOI: 10.1016/j.stem.2012.04.025
Palecek S P et al., have developed a protocol for generating almost pure populations of cardiomyocytes (up to 98% cardiomyocytes) from human pluripotent stem cells simply by modulating canonical [[Wnt signaling pathway]] at defined time points in the differentiation process, using readily accessible small molecule compounds.<ref name="Palecek">{{cite doi|10.1073/pnas.1200250109}}</ref> Discovery of the mechanisms controlling the formation of cardiomyocytes led to the development of the drug (ITD-1), which effectively clears the cell surface from [[Transforming growth factor beta|TGF-β]] receptor type II and selectively inhibits the intracellular TGF-β signaling and, thus, selectively enhances the differentiation of uncommitted [[mesoderm]] to cardiomyocytes, but not to vascular smooth muscle and endothelial cells.<ref name="Mercola">{{cite doi|10.1016/j.stem.2012.04.025}}</ref>
</ref>


Lu et al.<ref>Tung-Ying Lu, Bo Lin, Jong Kim, et al. & Lei Yang (2013) Repopulation of decellularized mouse heart with human induced pluripotent stem cell-derived cardiovascular progenitor cells. Nature Communications, 4, Article number: 2307 doi:10.1038/ncomms3307</ref> create heart constructs by repopulating decellularized mouse hearts with human induced pluripotent stem cell-derived multipotential cardiovascular progenitor cells. They found that the seeded multipotential cardiovascular progenitor cells migrate, proliferate and differentiate in situ into cardiomyocytes, smooth muscle cells and endothelial cells to reconstruct the decellularized hearts. In addition, they observed that heart's extracellular matrix of mouse (the substrate of heart scaffold) can send signals to guide the repopulated human multipotential cardiovascular progenitor cells into becoming the specialised cells needed for proper heart function. After 20 days of perfusion with growth factors, the engineered heart tissues started to beat again and were responsive to drugs.
Lu et al.<ref>{{cite doi|10.1038/ncomms3307}}</ref> create heart constructs by repopulating decellularized mouse hearts with human induced pluripotent stem cell-derived multipotential cardiovascular progenitor cells. They found that the seeded multipotential cardiovascular progenitor cells migrate, proliferate and differentiate in situ into cardiomyocytes, smooth muscle cells and endothelial cells to reconstruct the decellularized hearts. In addition, they observed that heart's extracellular matrix of mouse (the substrate of heart scaffold) can send signals to guide the repopulated human multipotential cardiovascular progenitor cells into becoming the specialised cells needed for proper heart function. After 20 days of perfusion with growth factors, the engineered heart tissues started to beat again and were responsive to drugs.


See also: [http://heart.bmj.com/content/early/2012/08/31/heartjnl-2011-301540.full?g=widget cell therapy in cardiovascular disease: selected research that has driven recent advances in clinical cardiology]
See also: [http://heart.bmj.com/content/early/2012/08/31/heartjnl-2011-301540.full?g=widget Cell therapy in cardiovascular disease: selected research that has driven recent advances in clinical cardiology]


=== Nephron Progenitors ===
=== Nephron Progenitors ===


Adult proximal tubule cells could be directly transcriptionally reprogrammed to [[nephron]] progenitors of the embryonic [[kidney]], using a pool of six genes of instructive transcription factors (SIX1, SIX2, OSR1, Eyes absent homolog 1(EYA1), Homeobox A11 (HOXA11) and Snail homolog 2 (SNAI2)) that activated a network of genes consistent with a cap mesenchyme/nephron progenitor phenotype in the adult proximal tubule cell line.<ref>Caroline E. Hendry, Jessica M. Vanslambrouck, Jessica Ineson, et al. and Melissa H. Little (2013) Direct Transcriptional Reprogramming of Adult Cells to Embryonic Nephron Progenitors. JASN ASN.2012121143; ,doi:10.1681/ASN.2012121143</ref>
Adult proximal tubule cells could be directly transcriptionally reprogrammed to [[nephron]] progenitors of the embryonic [[kidney]], using a pool of six genes of instructive transcription factors (SIX1, SIX2, OSR1, Eyes absent homolog 1(EYA1), Homeobox A11 (HOXA11) and Snail homolog 2 (SNAI2)) that activated a network of genes consistent with a cap mesenchyme/nephron progenitor phenotype in the adult proximal tubule cell line.<ref>{{cite doi|10.1681/ASN.2012121143}}</ref>
The generation of such cells may lead to cellular therapies for adult [[renal disease]]. Indeed, it has recently been shown that embryonic kidney organoids placed into adult rat kidney can undergo onward development and vascular development.<ref>Xinaris C, Benedetti V, Rizzo P, et al. and Giuseppe Remuzzi (2012) [http://211.144.68.84:9998/91keshi/Public/File/18/23-11/pdf/1857.full.pdf In vivo maturation of functional renal organoids formed from embryonic cell suspensions]. J Am Soc Nephrol 23: 1857–1868, doi: 10.1681/ASN.2012050505</ref>
The generation of such cells may lead to cellular therapies for adult [[renal disease]]. Indeed, it has recently been shown that embryonic kidney organoids placed into adult rat kidney can undergo onward development and vascular development.<ref>Xinaris C, Benedetti V, Rizzo P, et al. and Giuseppe Remuzzi (2012) [http://211.144.68.84:9998/91keshi/Public/File/18/23-11/pdf/1857.full.pdf In vivo maturation of functional renal organoids formed from embryonic cell suspensions]. J Am Soc Nephrol 23: 1857–1868, doi: 10.1681/ASN.2012050505</ref>


=== Bioengineering of the cells of blood vessels ===
=== Bioengineering of the cells of blood vessels ===


Blood vessels build extensive networks that supply all cells with nutrients and oxygen throughout life. As blood vessels get older, they often become abnormal in structure and function, thereby contributing to numerous age-associated diseases including myocardial infarction, ischemic stroke and atherosclerosis of arteries supplying the heart, brain, and lower extremities. So, an important goal is to stimulate vascular growth for the [[collateral circulation]] to prevent the exacerbation of these diseases. A useful cell type for cell-based therapy designed to stimulate coronary collateral growth are Induced Vascular Progenitor Cells (iVPCs), generated by partially reprogramming endothelial cells.<ref name="Kurian" /> The vascular commitment of iVPCs is related to the epigenetic memory of endothelial cells, which engenders them as cellular components of growing blood vessels. That is why, when iVPCs were implanted into myocardium, they engrafted in blood vessels and increased coronary collateral flow better than iPSCs, mesenchymal stem cells, or native endothelial cells.<ref name="Chilian">Yin L, Ohanyan V, Pung Y F, and Chilian W M. (2012) Induction of Vascular Progenitor Cells From Endothelial Cells Stimulates Coronary Collateral Growth. Circulation Research.;110:241-252, doi:10.1161/CIRCRESAHA.111.250126</ref>
Blood vessels build extensive networks that supply all cells with nutrients and oxygen throughout life. As blood vessels get older, they often become abnormal in structure and function, thereby contributing to numerous age-associated diseases including myocardial infarction, ischemic stroke and atherosclerosis of arteries supplying the heart, brain, and lower extremities. So, an important goal is to stimulate vascular growth for the [[collateral circulation]] to prevent the exacerbation of these diseases. A useful cell type for cell-based therapy designed to stimulate coronary collateral growth are Induced Vascular Progenitor Cells (iVPCs), generated by partially reprogramming endothelial cells.<ref name="Kurian" /> The vascular commitment of iVPCs is related to the epigenetic memory of endothelial cells, which engenders them as cellular components of growing blood vessels. That is why, when iVPCs were implanted into myocardium, they engrafted in blood vessels and increased coronary collateral flow better than iPSCs, mesenchymal stem cells, or native endothelial cells.<ref name="Chilian">{{cite doi|10.1161/CIRCRESAHA.111.250126}}</ref>
An effective strategy to enhance stem cell function is ex vivo genetic modification. For example, genetic modification with Pim-1 kinase (a downstream effector of Akt, which positively regulates neovasculogenesis) of bone marrow–derived cells<ref name=" Toko">Quijada P, Toko H, Fischer K M., et al. and Sussman M, A. (2012) Preservation of Myocardial Structure Is Enhanced by Pim-1 Engineering of Bone Marrow Cells. Circulation Research. ;111:77-86, doi:10.1161/CIRCRESAHA.112.265207</ref> or human cardiac progenitor cells, isolated from failing myocardium<ref name="Sussman">Mohsin S, Mohsin K, Toko H, et al & Sussman M A. (2012) Human Cardiac Progenitor Cells Engineered With Pim-I Kinase Enhance Myocardial Repair. J Am Coll Cardiol. 2012; doi:10.1016/j.jacc.2012.04.047</ref> results in durability of repair, together with the superior improvement of functional parameters of myocardial hemodynamic performance after cellular therapy of the injured myocardium with modified progenitor cells.
An effective strategy to enhance stem cell function is ex vivo genetic modification. For example, genetic modification with Pim-1 kinase (a downstream effector of Akt, which positively regulates neovasculogenesis) of bone marrow–derived cells<ref name=" Toko">{{cite doi|10.1161/CIRCRESAHA.112.265207}}</ref> or human cardiac progenitor cells, isolated from failing myocardium<ref name="Sussman">{{cite doi|10.1016/j.jacc.2012.04.047}}</ref> results in durability of repair, together with the superior improvement of functional parameters of myocardial hemodynamic performance after cellular therapy of the injured myocardium with modified progenitor cells.
Stem cells extracted from fat tissue after [[liposuction]] may be coaxed into progenitor [[Vascular smooth muscle|smooth muscle]] cells (iPVSMCs) found in arteries and veins.<ref name="Nollert M">American Heart Association (2012, July 25). Adult stem cells from liposuction used to create blood vessels in the lab. ScienceDaily.</ref> In the future, iVPCs and iPVSMCs may become a necessary source for the creation of blood vessels networks for tissue engineering and reconstruction of organs for transplantation. For instance, it was found that the 2D culture system of human iPS cells<ref>ZZ Wang, P Au, T Chen, et al. (2007) Endothelial cells derived from human embryonic stem cells form durable blood vessels in vivo, Nature Biotechnology 25, 317-318, doi:10.1038/nbt1287</ref> in conjunction with the CD34 (a surface glycophosphoprotein expressed on developmentally early embryonic fibroblast), NP1 (receptor - neuropilin 1) and KDR (kinase insert domain-containing receptor) triple marker selection (for the isolation of vasculogenic precursor cells from human induced pluripotent stem cells) was capable to generate endothelial cells that, after transplantation, were able to form stable functional blood vessels in vivo, lasting for 280 d in mice.<ref>Rekha Samuel, Laurence Daheron, Shan Liao, et al. and Rakesh K. Jain (2013 ) Generation of functionally competent and durable engineered blood vessels from human induced pluripotent stem cells. PNAS doi:10.1073/pnas.1310675110</ref>
Stem cells extracted from fat tissue after [[liposuction]] may be coaxed into progenitor [[Vascular smooth muscle|smooth muscle]] cells (iPVSMCs) found in arteries and veins.<ref name="Nollert M">American Heart Association (2012, July 25). Adult stem cells from liposuction used to create blood vessels in the lab. ScienceDaily.</ref> In the future, iVPCs and iPVSMCs may become a necessary source for the creation of blood vessels networks for tissue engineering and reconstruction of organs for transplantation. For instance, it was found that the 2D culture system of human iPS cells<ref>{{cite doi|10.1038/nbt1287}}</ref> in conjunction with the CD34 (a surface glycophosphoprotein expressed on developmentally early embryonic fibroblast), NP1 (receptor - neuropilin 1) and KDR (kinase insert domain-containing receptor) triple marker selection (for the isolation of vasculogenic precursor cells from human induced pluripotent stem cells) was capable to generate endothelial cells that, after transplantation, were able to form stable functional blood vessels in vivo, lasting for 280 d in mice.<ref>{{cite doi|10.1073/pnas.1310675110}}</ref>


In the treatment of infarction, it is important to prevent the formation of fibrotic scar tissue. This can be achieved in vivo by means of transient application of paracrine factors changing the fate of the native heart progenitor stem cells from contributing to cardiac fibrotic scar tissue and toward cardiovascular tissue. For example, it was shown in a mouse myocardial infarction model that a single intramyocardial injection of human vascular endothelial growth factor-A mRNA (VEGF-A modRNA), synthetically modified so that it escapes the normal defense system of the body, results in long-term improvement of heart function due to mobilization of epicardial progenitor cells and redirection of their differentiation toward cardiovascular cell types <ref>Lior Zangi, Kathy O Lui, Alexander von Gise, et al. & Kenneth R Chien.( 2013) Modified mRNA directs the fate of heart progenitor cells and induces vascular regeneration after myocardial infarction. Nature Biotechnology,; DOI:10.1038/nbt.2682</ref>
In the treatment of infarction, it is important to prevent the formation of fibrotic scar tissue. This can be achieved in vivo by means of transient application of paracrine factors changing the fate of the native heart progenitor stem cells from contributing to cardiac fibrotic scar tissue and toward cardiovascular tissue. For example, it was shown in a mouse myocardial infarction model that a single intramyocardial injection of human vascular endothelial growth factor-A mRNA (VEGF-A modRNA), synthetically modified so that it escapes the normal defense system of the body, results in long-term improvement of heart function due to mobilization of epicardial progenitor cells and redirection of their differentiation toward cardiovascular cell types <ref>{{cite doi|10.1038/nbt.2682}}</ref>


=== Bioengineering of blood stem cells ===
=== Bioengineering of blood stem cells ===
Line 210: Line 209:
==== Red blood cells ====
==== Red blood cells ====


[[Red blood cells]] (RBC) [[Blood transfusion|transfusion]] is necessary for many patients with emergency or hematological disorders. However, to date the supply of RBCs remains labile and dependent on voluntary donations. In addition, the transmission of infectious disease via blood transfusion from unspecified donors remains a risk. Establishing a large quantity of safe RBCs would help to address this issue. New technologies for ex vivo erythroid cell generation will hopefully provide alternative transfusion products to meet present and future clinical requirements.<ref name="Zeuner">Zeuner, A., Martelli, F., et al. and Migliaccio, A. R. (2012), Concise Review: Stem Cell-Derived Erythrocytes as Upcoming Players in Blood Transfusion. STEM CELLS, 30: 1587–1596. doi: 10.1002/stem.1136</ref><ref>Hirose Sho-ichi, Takayama Naoya, Nakamura Sou, et al. & Eto Koji (2013) Immortalization of Erythroblasts by c-MYC and BCL-XL Enables Large-Scale Erythrocyte Production from Human Pluripotent Stem Cells. Stem Cell Reports, doi: 10.1016/j.stemcr.2013.10.010</ref> In favor of this suggest data that RBCs generated in vitro from mobilized [[CD34]] positive cells have normal survival when transfused into an autologous recipient.<ref name="Douay">Giarratana MC, Rouard H, Dumont A, et al & Luc Douay (2011) Proof of principle for transfusion of in vitro generated red blood cells. Blood; 118(19): 5071-5079. doi: 10.1182/blood-2011-06-362038.</ref> However, the RBC produced in vitro contained exclusively fetal hemoglobin (HbF), which rescues the functionality of these RBCs. However, in vivo the switch of fetal to adult hemoglobin after infusion of nucleated erythroid precursors derived from induced pluripotent stem cells was observed.<ref name="Kobari">Ladan Kobari, Frank Yates, Noufissa Oudrhiri et al. and Luc Douay (2012) Human induced pluripotent stem cells can reach complete terminal maturation: in vivo and in vitro evidence in the erythropoietic differentiation model. Haematologica. 2012; 97:xxx DOI: 10.3324/haematol.2011.055566</ref> But then there is another problem: although RBCs do not have nuclei, and therefore can not form a tumor, their immediate precursors the erythroblasts do. The terminal maturation of erythroblasts into functional RBCs requires a complex remodeling process that ends with extrusion of the nucleus and the formation of an enucleated RBC.<ref name="Crispino">Keerthivasan Ganesan , Wickrema A, and Crispino J D (2011) Erythroblast Enucleation Stem Cells Int.; 2011: 139851. doi: 10.4061/2011/139851</ref> Alas, cell reprogramming methodologies at present often disrupt these processes of enucleation, and therefore transfusion of in vitro-generated RBCs or their immediate precursors the erythroblasts still insufficiently protected against the possibility of tumors formation.
[[Red blood cells]] (RBC) [[Blood transfusion|transfusion]] is necessary for many patients with emergency or hematological disorders. However, to date the supply of RBCs remains labile and dependent on voluntary donations. In addition, the transmission of infectious disease via blood transfusion from unspecified donors remains a risk. Establishing a large quantity of safe RBCs would help to address this issue. New technologies for ex vivo erythroid cell generation will hopefully provide alternative transfusion products to meet present and future clinical requirements.<ref name="Zeuner">{{cite doi|10.1002/stem.1136}}</ref><ref>{{cite doi|10.1016/j.stemcr.2013.10.010}}</ref> In favor of this suggest data that RBCs generated in vitro from mobilized [[CD34]] positive cells have normal survival when transfused into an autologous recipient.<ref name="Douay">{{cite doi| 10.1182/blood-2011-06-362038}}</ref> However, the RBC produced in vitro contained exclusively fetal hemoglobin (HbF), which rescues the functionality of these RBCs. However, in vivo the switch of fetal to adult hemoglobin after infusion of nucleated erythroid precursors derived from induced pluripotent stem cells was observed.<ref name="Kobari">{{cite doi| 10.3324/haematol.2011.055566}}</ref> But then there is another problem: although RBCs do not have nuclei, and therefore can not form a tumor, their immediate precursors the erythroblasts do. The terminal maturation of erythroblasts into functional RBCs requires a complex remodeling process that ends with extrusion of the nucleus and the formation of an enucleated RBC.<ref name="Crispino">{{cite doi| 10.4061/2011/139851}}</ref> Alas, cell reprogramming methodologies at present often disrupt these processes of enucleation, and therefore transfusion of in vitro-generated RBCs or their immediate precursors the erythroblasts still insufficiently protected against the possibility of tumors formation.
However, Bouhassira and colleagues recently found that exposing CD34 positive cells to a short pulse of cytokines favorable for erythroid differentiation prior to stem cell expansion followed by progenitor expansion produced on the order more yield of erythroid cells than the yields observed previously. And the most important: These red blood cells expressed a globin profile similar to that of the developmental age of the CD34 positive cells<ref name="Bouhassira">Emmanuel Olivier, Caihong Qiu, Eric E. Bouhassira (2012) Protocols and Manufacturing for Cell-Based TherapiesNovel, High-Yield Red Blood Cell Production Methods from CD34-Positive Cells Derived from Human Embryonic Stem, Yolk Sac, Fetal Liver, Cord Blood, and Peripheral Blood Stem Cells Trans Med first published on August 2, 2012;doi:10.5966/sctm.2012-0059</ref>
However, Bouhassira and colleagues recently found that exposing CD34 positive cells to a short pulse of cytokines favorable for erythroid differentiation prior to stem cell expansion followed by progenitor expansion produced on the order more yield of erythroid cells than the yields observed previously. And the most important: These red blood cells expressed a globin profile similar to that of the developmental age of the CD34 positive cells<ref name="Bouhassira">{{cite doi|10.5966/sctm.2012-0059}}</ref>
It is also interesting to note that an important role in normal blood cell development plays the aryl hydrocarbon receptor (AhR) pathway (which has been shown to be involved in the promotion of cancer cell development). AhR activation in human hematopoietic progenitor cells (HPs) drives an unprecedented expansion of HPs, megakaryocyte- and erythroid-lineage cells.<ref>Brenden W. Smith, Sarah S. Rozelle, Amy Leung, et al and George J. Murphy (2013) The aryl hydrocarbon receptor directs hematopoietic progenitor cell expansion and differentiation. Blood, blood-2012-11-466722, doi: 10.1182/blood-2012-11-466722</ref>
It is also interesting to note that an important role in normal blood cell development plays the aryl hydrocarbon receptor (AhR) pathway (which has been shown to be involved in the promotion of cancer cell development). AhR activation in human hematopoietic progenitor cells (HPs) drives an unprecedented expansion of HPs, megakaryocyte- and erythroid-lineage cells.<ref>{{cite doi|10.1182/blood-2012-11-466722}}</ref>


See also: Migliaccio AR, Whitsett C, Papayannopoulou T, Sadelain M. (2012) [http://www.cell.com/cell-stem-cell/abstract/S1934-5909(12)00003-3 The potential of stem cells as an in vitro source of red blood cells for transfusion.] Review. Cell Stem Cell.;10(2):115-9
See also: Migliaccio AR, Whitsett C, Papayannopoulou T, Sadelain M. (2012) [http://www.cell.com/cell-stem-cell/abstract/S1934-5909(12)00003-3 The potential of stem cells as an in vitro source of red blood cells for transfusion.] Review. Cell Stem Cell.;10(2):115-9
Line 218: Line 217:
==== Platelets ====
==== Platelets ====


[[Platelets]] play an important role in preventing hemorrhage in thrombocytopenic patients and patients with thrombocythemia. A significant problem for multitransfused patients is refractoriness to platelet transfusions. Thus, the ability to generate platelet products ex vivo and platelet products lacking [[Human leukocyte antigen|HLA antigens]] in serum free media, would have great clinical value. Some success in this direction has achieved Figueiredo et al. Using an [[RNA interference]]-based mechanism in which a [[Viral vector|lentiviral vector]] was used to express short-hairpin RNAi targeting β2-microglobulin transcripts in CD34-positive cells, they generated platelets demonstrating an 85% reduction in class I HLA antigens. These platelets appeared to have normal function in vitro<ref name=" Figueiredo ">Figueiredo C, Goudeva L., Horn P. A., et al and Seltsam A. (2010) Generation of HLA-deficient platelets from hematopoietic progenitor cells. Transfusion.; 50(8): 1690-701. doi: 10.1111/j.1537-2995.2010.02644.x.</ref>
[[Platelets]] play an important role in preventing hemorrhage in thrombocytopenic patients and patients with thrombocythemia. A significant problem for multitransfused patients is refractoriness to platelet transfusions. Thus, the ability to generate platelet products ex vivo and platelet products lacking [[Human leukocyte antigen|HLA antigens]] in serum free media, would have great clinical value. Some success in this direction has achieved Figueiredo et al. Using an [[RNA interference]]-based mechanism in which a [[Viral vector|lentiviral vector]] was used to express short-hairpin RNAi targeting β2-microglobulin transcripts in CD34-positive cells, they generated platelets demonstrating an 85% reduction in class I HLA antigens. These platelets appeared to have normal function in vitro<ref name=" Figueiredo ">{{cite doi|10.1111/j.1537-2995.2010.02644.x}}</ref>
A clinically applicable strategy for the derivation of functional platelets from human iPSC was developed, that involves the establishment of stable immortalized megakaryocyte progenitor cell lines (imMKCLs) through the [[doxycycline]]-dependent overexpression of [[BMI1]] and [[Bcl-xL|BCL-XL]]. The resulting imMKCLs can be expanded in culture over extended periods (4–5 months), even after cryopreservation. Halting the overexpression (by the removal of doxycycline from the medium) of c-MYC, [[BMI1]], and [[Bcl-xL|BCL-XL]] in growing imMKCLs led to the production of [[GP1BA|CD42b]]+ platelets with functionality comparable to that of native platelets on the basis of a range of assays in vitro and in vivo.<ref>Sou Nakamura, Naoya Takayama, Shinji Hirata, et al. & Koji Eto (2014). Expandable Megakaryocyte Cell Lines Enable Clinically Applicable Generation of Platelets from Human Induced Pluripotent Stem Cells. Cell Stem Cell; DOI: 10.1016/j.stem.2014.01.011</ref>
A clinically applicable strategy for the derivation of functional platelets from human iPSC was developed, that involves the establishment of stable immortalized megakaryocyte progenitor cell lines (imMKCLs) through the [[doxycycline]]-dependent overexpression of [[BMI1]] and [[Bcl-xL|BCL-XL]]. The resulting imMKCLs can be expanded in culture over extended periods (4–5 months), even after cryopreservation. Halting the overexpression (by the removal of doxycycline from the medium) of c-MYC, [[BMI1]], and [[Bcl-xL|BCL-XL]] in growing imMKCLs led to the production of [[GP1BA|CD42b]]+ platelets with functionality comparable to that of native platelets on the basis of a range of assays in vitro and in vivo.<ref>{{cite doi|10.1016/j.stem.2014.01.011}}</ref>


==== Immune cells ====
==== Immune cells ====


A specialised type of [[white blood cell]], known as '''[[cytotoxic T cell|cytotoxic T]] [[lymphocytes]] (CTLs)''', are produced by the [[immune system]] and are able to recognise specific markers on the surface of various infectious or tumour cells, causing them to launch an attack to kill the harmful cells. Thence, immunotherapy with functional antigen-specific T cells has potential to be an effective therapeutic strategy for combating many types of cancer and viral infection.<ref>Riddell, S.R. & Greenberg, P.D. (1995) Principles for adoptive T cell therapy of human viral diseases. Annu. Rev. Immunol. 13, 545–586 DOI: 10.1146/annurev.iy.13.040195.002553</ref> However, sources of such cells for therapeutic purposes are limited, because they are produced in small numbers naturally and have a short lifespan.
A specialised type of [[white blood cell]], known as '''[[cytotoxic T cell|cytotoxic T]] [[lymphocytes]] (CTLs)''', are produced by the [[immune system]] and are able to recognise specific markers on the surface of various infectious or tumour cells, causing them to launch an attack to kill the harmful cells. Thence, immunotherapy with functional antigen-specific T cells has potential to be an effective therapeutic strategy for combating many types of cancer and viral infection.<ref>{{cite doi|10.1146/annurev.iy.13.040195.002553}}</ref> However, sources of such cells for therapeutic purposes are limited, because they are produced in small numbers naturally and have a short lifespan.
A potentially efficient approach for generating antigen-specific CTLs is to revert mature immune T cells into iPSCs, which possess indefinite proliferative capacity in vitro, and after their multiplication to coaxed them to redifferentiate back into T cells<ref name="Hiromitsu">Toshinobu Nishimura, Shin Kaneko, Ai Kawana-Tachikawa et al. & Hiromitsu Nakauchi (2013) Generation of rejuvenated antigen-specific T cells by pluripotency reprogramming and redifferentiation. Cell Stem Cell, 12(1), 114-126 DOI: 10.1016/j.stem.2012.11.002</ref><ref name="Kawamoto">Raul Vizcardo, Kyoko Masuda, Daisuke Yamada, et al. & Hiroshi Kawamoto (2013) Regeneration of Human Tumor Antigen-Specific T Cells from iPSCs Derived from Mature CD8+ T Cells . Cell Stem Cell, 12(1), 31-36 DOI: http://dx.doi.org/10.1016/j.stem.2012.12.006</ref><ref name="Crompton">Joseph G. Crompton, Mahendra Rao, Nicholas P. Restifo (2013) Memoirs of a Reincarnated T Cell. Cell Stem Cell, 12(1), 6-8 DOI: 10.1016/j.stem.2012.12.009</ref><ref name="Xiong">Lei F, Haque R, Xiong X, Song J. (2012) Directed differentiation of induced pluripotent stem cells towards T lymphocytes. J Vis Exp. ;(63):e3986. doi: 10.3791/3986</ref>
A potentially efficient approach for generating antigen-specific CTLs is to revert mature immune T cells into iPSCs, which possess indefinite proliferative capacity in vitro, and after their multiplication to coaxed them to redifferentiate back into T cells<ref name="Hiromitsu">{{cite doi|10.1016/j.stem.2012.11.002}}</ref><ref name="Kawamoto">{{cite doi|10.1016/j.stem.2012.12.006}}</ref><ref name="Crompton">{{cite doi|10.3791/3986}}</ref>
Even more opportunities promises a method that combine iPSC and chimeric antigen receptor (CAR) <ref>Sadelain, M., Brentjens, R. & Riviere, I. (2013). The basic principles of chimeric antigen receptor design. Cancer Discov. 3, 388–398 doi: 10.1158/2159-8290.CD-12-054</ref> technologies to generate human T cells targeted to CD19, an antigen expressed by malignant B cells, in tissue culture.<ref>Maria Themeli, Christopher C Kloss, Giovanni Ciriello, et al. & Michel Sadelain (2013) Generation of tumor-targeted human T lymphocytes from induced pluripotent stem cells for cancer therapy. Nature Biotechnology, doi:10.1038/nbt.2678</ref> This approach of generating therapeutic human T cells 'in the dish' may be useful for cancer immunotherapy and other medical applications because such 'living drugs' have fewer side-effects, once injected - stay in the body and move around to stop recurrences.
Even more opportunities promises a method that combine iPSC and chimeric antigen receptor (CAR) <ref>{{cite doi|10.1158/2159-8290.CD-12-054}}</ref> technologies to generate human T cells targeted to CD19, an antigen expressed by malignant B cells, in tissue culture.<ref>{{cite doi|10.1038/nbt.2678}}</ref> This approach of generating therapeutic human T cells 'in the dish' may be useful for cancer immunotherapy and other medical applications because such 'living drugs' have fewer side-effects, once injected - stay in the body and move around to stop recurrences.


'''Invariant [[Natural killer T cell|natural killer T]] (iNKT) cells''' has great clinical potential as [[adjuvant]] for cancer immunotherapy. iNKT cells act as innate T lymphocytes and serve as a bridge between the [[Innate immune system|innate]] and [[acquired immune system]]s. They augment anti-tumor responses by producing [[interferon-gamma]] (IFN-γ).<ref>Karsten A. Pilones, Joseph Aryankalayil, and Sandra Demaria (2012) Invariant NKT Cells as Novel Targets for Immunotherapy in Solid Tumors. Clinical and Developmental Immunology, 2012 , Article ID 720803, doi:10.1155/2012/720803</ref>
'''Invariant [[Natural killer T cell|natural killer T]] (iNKT) cells''' has great clinical potential as [[adjuvant]] for cancer immunotherapy. iNKT cells act as innate T lymphocytes and serve as a bridge between the [[Innate immune system|innate]] and [[acquired immune system]]s. They augment anti-tumor responses by producing [[interferon-gamma]] (IFN-γ).<ref>{{cite doi|10.1155/2012/720803}}</ref>
A conceptual method is proposed for the use of iPSC-derived iNKT cells for adjuvant cell therapy against cancer, which is composed of four segments: (1) collection of iNKT cells, (2) reprogramming of iNKT cells into iPSCs, (3) re-differentiation of iNKT cell-derived iPSCs into iNKT cells and their expansion in vitro, and (4) injection of iPSC-derived iNKT cells into tumor-bearing animals.<ref name="Koseki">Watarai H, Yamada D, Fujii S, Taniguchi M, Koseki H. (2012) Induced pluripotency as a potential path towards iNKT cell-mediated cancer immunotherapy. Int J Hematol. ;95(6):624-631. doi: 10.1007/s12185-012-1091-0</ref>
A conceptual method is proposed for the use of iPSC-derived iNKT cells for adjuvant cell therapy against cancer, which is composed of four segments: (1) collection of iNKT cells, (2) reprogramming of iNKT cells into iPSCs, (3) re-differentiation of iNKT cell-derived iPSCs into iNKT cells and their expansion in vitro, and (4) injection of iPSC-derived iNKT cells into tumor-bearing animals.<ref name="Koseki">{{cite doi|10.1007/s12185-012-1091-0}}</ref>


'''[[Dendritic cells]] (DC)''' are specialized to control T-cell responses. DC with appropriate genetic modifications may survive long enough to stimulate antigen-specific CTL and, after that, be completely eliminated. It was shown that DC-like antigen-presenting cells obtained from human induced pluripotent stem cells may serve as an unlimited source for [[vaccination]] therapy.<ref name="Senju">M Haruta, Y Tomita, A Yuno, et al. and S Senju (2012) TAP-deficient human iPS cell-derived myeloid cell lines as unlimited cell source for dendritic cell-like antigen-presenting cells. Gene Therapy , doi:10.1038/gt.2012.59</ref>
'''[[Dendritic cells]] (DC)''' are specialized to control T-cell responses. DC with appropriate genetic modifications may survive long enough to stimulate antigen-specific CTL and, after that, be completely eliminated. It was shown that DC-like antigen-presenting cells obtained from human induced pluripotent stem cells may serve as an unlimited source for [[vaccination]] therapy.<ref name="Senju">{{cite doi|10.1038/gt.2012.59}}</ref>


'''B cells'''. [[Ccaat-enhancer-binding proteins|CCAAT/enhancer binding protein-α (C/EBPα)]] induces transdifferentiation of [[B cell]]s into [[macrophage]]s at high efficiencies<ref>Xie, H., Ye, M., Feng, R. & Graf, T (2004) [http://www.sciencedirect.com/science/article/pii/S0092867404004192 Stepwise reprogramming of B cells into macrophages]. Cell 117(5), 663–676 doi: 10.1016/S0092-8674(04)00419-2</ref><ref>Bussmann, L.H., Schubert, A., Vu Manh, T.P et al. and Graf, T. (2009). A robust and highly efficient immune cell reprogramming system. Cell Stem Cell, 5(5), 554–566. doi: 10.1016/j.stem.2009.10.004</ref> and enhances reprogramming into iPS cells when co-expressed with the transcription factors Oct4, Sox2, Klf4 and Myc<ref>Hanna, J., Markoulaki, S., Schorderet, P., et al. & Jaenisch, R. (2008). Direct reprogramming of terminally differentiated mature B lymphocytes to pluripotency. Cell, 133(2), 250-264. doi: 10.1016/j.cell.2008.03.028</ref> with a 100-fold increase in iPS cell reprogramming efficiency, involving 95% of the population.<ref>Bruno Di Stefano, Jose Luis Sardina, Chris van Oevelen, et al. & Thomas Graf. ( 2013) C/EBPα poises B cells for rapid reprogramming into induced pluripotent stem cells. Nature,; DOI:10.1038/nature12885</ref>
'''B cells'''. [[Ccaat-enhancer-binding proteins|CCAAT/enhancer binding protein-α (C/EBPα)]] induces transdifferentiation of [[B cell]]s into [[macrophage]]s at high efficiencies<ref{{cite doi|10.1016/S0092-8674(04)00419-2}}</ref><ref>{{cite doi|10.1016/j.stem.2009.10.004}}</ref> and enhances reprogramming into iPS cells when co-expressed with the transcription factors Oct4, Sox2, Klf4 and Myc<ref>{{cite doi|10.1016/j.cell.2008.03.028}}</ref> with a 100-fold increase in iPS cell reprogramming efficiency, involving 95% of the population.<ref>{{cite doi|10.1038/nature12885}}</ref>
Furthermore, C/EBPa can convert at high efficiencies selected human B cell lymphoma and leukemia cell lines into macrophage-like cells, impairing the cells’ tumor-forming capacity.<ref>Rapino et al., (2013) C/EBPaInduces Highly Efficient Macrophage Transdifferentiation of B Lymphoma and Leukemia Cell Lines and Impairs Their Tumorigenicity, Cell Reports, http://dx.doi.org/10.1016/j.celrep.2013.03.003</ref>
Furthermore, C/EBPa can convert at high efficiencies selected human B cell lymphoma and leukemia cell lines into macrophage-like cells, impairing the cells’ tumor-forming capacity.<ref>Rapino et al., (2013) C/EBPaInduces Highly Efficient Macrophage Transdifferentiation of B Lymphoma and Leukemia Cell Lines and Impairs Their Tumorigenicity, Cell Reports, http://dx.doi.org/10.1016/j.celrep.2013.03.003</ref>


Line 242: Line 241:
range of mesenchymal-lineage tissues, [[Mesenchymal stem cell|mesenchymal stem/stromal cells]] (MSCs) are under
range of mesenchymal-lineage tissues, [[Mesenchymal stem cell|mesenchymal stem/stromal cells]] (MSCs) are under
intense investigation for applications in cardiac, renal, neural, joint, and bone repair, as
intense investigation for applications in cardiac, renal, neural, joint, and bone repair, as
well as in inflammatory conditions and hemopoietic cotransplantation.<ref name="Fu X">Peng Y, Huang S, Cheng B, et al. and Fu X. (2012) Mesenchymal stem cells: A revolution in therapeutic strategies of age-related diseases Review Article. Ageing Research Reviews, , Available online 30 April 2012, .doi.org/10.1016/j.arr.2012.04.005</ref> MSCs are typically harvested from adult bone marrow or fat, but these not only require painful invasive procedures but are low-frequency sources, with MSCs making up only 0.001%– 0.01% of bone marrow cells and 0.05% in liposuction aspirates.<ref name="Bieback">Bieback K, Kern S, Kocaomer A et al. (2008) Comparing mesenchymal stromal cells from different human tissues: Bone marrow, adipose tissue and umbilical cord blood. Biomed Mater Eng; 18:S71–S76</ref> Of concern for autologous use, in particular in the elderly most in need of tissue repair, MSCs decline in quantity and quality with age.<ref name="Fu X" /><ref>Anastasia Efimenko, Nina Dzhoyashvili, Natalia Kalinina, Tatiana Kochegura, Renat Akchurin, Vsevolod Tkachuk, Yelena Parfyonova (2014), Adipose-Derived Mesenchymal Stromal Cells From Aged Patients With Coronary Artery Disease Keep Mesenchymal Stromal Cell Properties but Exhibit Characteristics of Aging and Have Impaired Angiogenic Potential. Stem Cells Trans Med ; 3:32-41; doi:10.5966/sctm.2013-0014</ref><ref name="McGonagle">Stolzing A, Jones E, McGonagle D et al. (2008) Age-related changes in human bone marrow derived mesenchymal stem cells: Consequences for cell therapies. Mech Ageing Dev;129:163–173</ref>
well as in inflammatory conditions and hemopoietic cotransplantation.<ref name="Fu X"{{cite doi|10.1016/j.arr.2012.04.005}}</ref> MSCs are typically harvested from adult bone marrow or fat, but these not only require painful invasive procedures but are low-frequency sources, with MSCs making up only 0.001%– 0.01% of bone marrow cells and 0.05% in liposuction aspirates.<ref name="Bieback">Bieback K, Kern S, Kocaomer A et al. (2008) Comparing mesenchymal stromal cells from different human tissues: Bone marrow, adipose tissue and umbilical cord blood. Biomed Mater Eng; 18:S71–S76</ref> Of concern for autologous use, in particular in the elderly most in need of tissue repair, MSCs decline in quantity and quality with age.<ref name="Fu X" /><ref>{{cite doi|10.5966/sctm.2013-0014}}</ref><ref name="McGonagle">Stolzing A, Jones E, McGonagle D et al. (2008) Age-related changes in human bone marrow derived mesenchymal stem cells: Consequences for cell therapies. Mech Ageing Dev;129:163–173</ref>
However, induced pluripotent stem cells (iPSCs) could be obtained by the cells rejuvenation of even centenarian human.<ref name="Laure">Laure Lapasset et al. and Jean-Marc Lemaitre (2011) Rejuvenating senescent and centenarian human cells by reprogramming through the pluripotent state. Genes Dev. 25: 2248-2253; doi: 10.1101/gad.173922.111</ref> Because iPSCs can be harvested free of ethical constraints and culture can be expanded indefinitely, they are an advantageous source of MSCs.<ref name="Cantz">Irina Eberle, Mohsen Moslem, Reinhard Henschler, Tobias Cantz (2012) [http://link.springer.com/chapter/10.1007%2F10_2012_156?LI=true Engineered MSCs from Patient-Specific iPS Cells]. Advances in Biochemical Engineering Biotechnology</ref> Chen and colleagues discovered that iPSCs treatment with [[SB-431542]] (that inhibits the activin/TGF- pathways by blocking phosphorylation of ALK4, ALK5, and ALK7 receptors) leads to rapid and uniform MSC generation from human iPSCs. These iPS-MSCs possibly lack teratoma-forming ability, display a normal stable karyotype in culture, and exhibit growth and differentiation characteristics that closely resemble those of primary MSCs described, has considerable potential for the in vitro scale-up required to enable a wide range of MSC-based therapies.<ref name=" Fisk">Chen Y S, Pelekanos R A., Ellis R L., et al and Nicholas M. Fisk (2012) Small Molecule Mesengenic Induction of Human Induced Pluripotent Stem Cells to Generate Mesenchymal Stem/Stromal Cells Stem Cells Trans Med published online February 7, 2012 doi:10.5966/sctm.2011-0022</ref> Unfortunately, currently there is no data on the in vivo efficacy and long-term safety of iPSC-derived MSCs generated by this method.
However, induced pluripotent stem cells (iPSCs) could be obtained by the cells rejuvenation of even centenarian human.<ref name="Laure">{{cite doi|10.1101/gad.173922.111}}</ref> Because iPSCs can be harvested free of ethical constraints and culture can be expanded indefinitely, they are an advantageous source of MSCs.<ref name="Cantz">Irina Eberle, Mohsen Moslem, Reinhard Henschler, Tobias Cantz (2012) [http://link.springer.com/chapter/10.1007%2F10_2012_156?LI=true Engineered MSCs from Patient-Specific iPS Cells]. Advances in Biochemical Engineering Biotechnology</ref> Chen and colleagues discovered that iPSCs treatment with [[SB-431542]] (that inhibits the activin/TGF- pathways by blocking phosphorylation of ALK4, ALK5, and ALK7 receptors) leads to rapid and uniform MSC generation from human iPSCs. These iPS-MSCs possibly lack teratoma-forming ability, display a normal stable karyotype in culture, and exhibit growth and differentiation characteristics that closely resemble those of primary MSCs described, has considerable potential for the in vitro scale-up required to enable a wide range of MSC-based therapies.<ref name=" Fisk">{{cite doi|10.5966/sctm.2011-0022}}</ref> Unfortunately, currently there is no data on the in vivo efficacy and long-term safety of iPSC-derived MSCs generated by this method.


It is interesting to note that besides cell therapy in vivo the culture of human mesenchymal stem cells can be used in vitro for mass-production of [[Exosome (vesicle)|exosomes]], which, as it turned out, are ideal vehicles for drug delivery.<ref>Ruenn Chai Lai, Ronne Wee Yeh Yeo, Soon Sim Tan, Bin Zhang, et al. and Sai Kiang Lim (2013) Mesenchymal Stem Cell Exosomes: The Future MSC-Based Therapy? In: Mesenchymal Stem Cell Therapy. Chase, Lucas G.; Vemuri, Mohan C. (Eds.). 39-61 DOI
It is interesting to note that besides cell therapy in vivo the culture of human mesenchymal stem cells can be used in vitro for mass-production of [[Exosome (vesicle)|exosomes]], which, as it turned out, are ideal vehicles for drug delivery.<ref>Ruenn Chai Lai, Ronne Wee Yeh Yeo, Soon Sim Tan, Bin Zhang, et al. and Sai Kiang Lim (2013) Mesenchymal Stem Cell Exosomes: The Future MSC-Based Therapy? In: Mesenchymal Stem Cell Therapy. Chase, Lucas G.; Vemuri, Mohan C. (Eds.). 39-61 DOI
Line 250: Line 249:
==== Dedifferentiated adipocytes ====
==== Dedifferentiated adipocytes ====


Adipose tissue, because of its abundance and relatively less invasive harvest methods, represents a practical and appealing source of mesenchymal stem cells (MSCs). Unfortunately, there are only 0.05% MSCs in liposuction aspirates.<ref name="Bieback"/> However, a large amount of mature adipocytes, which, in general, lost their proliferative abilities, and therefore often discarded, can be easily isolated from the adipose cell suspension and dedifferentiated into lipid-free fibroblast-like cells, named dedifferentiated fat (DFAT) cells. DFAT cells re-establish active proliferation ability and undertake multipotent capacities.<ref>Medet Jumabay, Raushan Abdmaulen, Albert Ly, et al. and Kristina I. Boström (January 2014). Pluripotent Stem Cells Derived From Mouse and Human White Mature Adipocytes. Stem Cells Trans Med. sctm.2013-0107 doi:10.5966/sctm.2013-0107</ref> Compared with adult stem cells, DFAT cells showed unique advantages in their abundance, isolation, and homogeneity. Under proper induction culture in vitro or environment in vivo, DFAT cells could demonstrate adipogenic, osteogenic, chondrogenic, and myogenic potentials. They also could exhibit perivascular characteristics and elicit neovascularization.<ref name="Cinti">Poloni A, Maurizi G, Leoni P, et al. & Cinti S (2012) Human Dedifferentiated Adipocytes Show Similar Properties to Bone Marrow-Derived Mesenchymal Stem Cells. Stem Cells. ;30(5):965-74. doi: 10.1002/stem.1067.</ref><ref name="Sugawara">Shen JF, Sugawara A, Yamashita J, Ogura H, Sato S. (2011) Dedifferentiated fat cells: an alternative source of adult multipotent cells from the adipose tissues. Int J Oral Sci.;3(3):117-24</ref><ref>Sara M. Melief, Jaap Jan Zwaginga, Willem E. Fibbe and Helene Roelofs ( 2013) Adipose Tissue-Derived Multipotent Stromal Cells Have a Higher Immunomodulatory Capacity Than Their Bone Marrow-Derived Counterparts. Stem Cells Trans Med May 2013 sctm.2012-0184 doi:10.5966/sctm.2012-0184</ref>
Adipose tissue, because of its abundance and relatively less invasive harvest methods, represents a practical and appealing source of mesenchymal stem cells (MSCs). Unfortunately, there are only 0.05% MSCs in liposuction aspirates.<ref name="Bieback"/> However, a large amount of mature adipocytes, which, in general, lost their proliferative abilities, and therefore often discarded, can be easily isolated from the adipose cell suspension and dedifferentiated into lipid-free fibroblast-like cells, named dedifferentiated fat (DFAT) cells. DFAT cells re-establish active proliferation ability and undertake multipotent capacities.<ref>{{cite doi|10.5966/sctm.2013-0107}}</ref> Compared with adult stem cells, DFAT cells showed unique advantages in their abundance, isolation, and homogeneity. Under proper induction culture in vitro or environment in vivo, DFAT cells could demonstrate adipogenic, osteogenic, chondrogenic, and myogenic potentials. They also could exhibit perivascular characteristics and elicit neovascularization.<ref name="Cinti">{{cite doi|10.1002/stem.1067}}</ref><ref name="Sugawara">Shen JF, Sugawara A, Yamashita J, Ogura H, Sato S. (2011) Dedifferentiated fat cells: an alternative source of adult multipotent cells from the adipose tissues. Int J Oral Sci.;3(3):117-24</ref><ref>{{cite doi|10.5966/sctm.2012-0184}}</ref>


=== Induced chondrogenic cells ===
=== Induced chondrogenic cells ===


Cartilage is the connective tissue responsible for frictionless joint movement. Its degeneration ultimately results in complete loss of joint function in the late stages of osteoarthritis. As an avascular and hypocellular tissue, cartilage has a very limited capacity for self-repair. Chondrocytes are the only cell type in cartilage, in which they are surrounded by the extracellular matrix that they secrete and assemble.
Cartilage is the connective tissue responsible for frictionless joint movement. Its degeneration ultimately results in complete loss of joint function in the late stages of osteoarthritis. As an avascular and hypocellular tissue, cartilage has a very limited capacity for self-repair. Chondrocytes are the only cell type in cartilage, in which they are surrounded by the extracellular matrix that they secrete and assemble.
One conceivable method of producing cartilage is to induce it from iPS cells.<ref>Cheng, A., Hardingham, T. E., & Kimber, S. J. (2013). Generating Cartilage Repair from Pluripotent Stem Cells. Tissue Engineering Part B: Reviews. doi:10.1089/ten.teb.2012.0757.</ref>
One conceivable method of producing cartilage is to induce it from iPS cells.<ref>{{cite doi|10.1089/ten.teb.2012.0757}}</ref>
Alternatively, it is possible to convert fibroblasts directly into induced chondrogenic cells (iChon cells) without an intermediate iPS cell stage, by inserting three reprogramming factors (c-MYC, KLF4, and SOX9).<ref>Outani H, Okada M, Yamashita A, Nakagawa K, Yoshikawa H, et al. (2013) Direct Induction of Chondrogenic Cells from Human Dermal Fibroblast Culture by Defined Factors. PLoS ONE 8(10): e77365. doi:10.1371/journal.pone.0077365</ref>
Alternatively, it is possible to convert fibroblasts directly into induced chondrogenic cells (iChon cells) without an intermediate iPS cell stage, by inserting three reprogramming factors (c-MYC, KLF4, and SOX9).<ref>{{cite doi|10.1371/journal.pone.0077365}}</ref>
The human iChon cells expressed marker genes for chondrocytes (type II collagen) but not fibroblasts.
Implanted into defects created in the articular cartilage of rats’, the human iChon cells were able to survive and form cartilaginous tissue for at least four weeks, but not tumors. Nevertheless, the method used in the study makes use of c-MYC, which is thought to have a major role in tumorigenesis, and employs a retrovirus to introduce the reprogramming factors. So, it could not be applied without modification in human therapy.{{cite doi|10.1016/j.stem.2012.11.002}}</ref>{{cite doi|10.1016/j.stem.2012.12.006}}</ref><ref name="Crompton">Joseph G. Crompton, Mahendra Rao, Nicholas P. Restifo (2013) Memoirs of a Reincarnated T Cell. Cell Stem Cell, 12(1), 6-8 >{{cite doi 10.1016/j.stem.2012.12.009}}</ref><ref name="Xiong">Lei F, Haque R, Xiong X, Song J. (2012) Directed differentiation of induced pluripotent stem cells towards T lymphocytes. J Vis Exp. ;(63):e3986. >{{cite doi|10.3791/3986}}</ref>
Even more opportunities promises a method that combine iPSC and chimeric antigen receptor (CAR) <ref>Sadelain, M., Brentjens, R. & Riviere, I. (2013). The basic principles of chimeric antigen receptor design. Cancer Discov. 3, 388–398 >{{cite doi|10.1158/2159-8290.CD-12-054}}</ref> technologies to generate human T cells targeted to CD19, an antigen expressed by malignant B cells, in tissue culture.<ref>{{cite doi|10.1038/nbt.2678}}</ref> This approach of generating therapeutic human T cells 'in the dish' may be useful for cancer immunotherapy and other medical applications because such 'living drugs' have fewer side-effects, once injected - stay in the body and move around to stop recurrences.

'''Invariant [[Natural killer T cell|natural killer T]] (iNKT) cells''' has great clinical potential as [[adjuvant]] for cancer immunotherapy. iNKT cells act as innate T lymphocytes and serve as a bridge between the [[Innate immune system|innate]] and [[acquired immune system]]s. They augment anti-tumor responses by producing [[interferon-gamma]] (IFN-γ).<ref>{{cite doi|10.1155/2012/720803}}</ref>
A conceptual method is proposed for the use of iPSC-derived iNKT cells for adjuvant cell therapy against cancer, which is composed of four segments: (1) collection of iNKT cells, (2) reprogramming of iNKT cells into iPSCs, (3) re-differentiation of iNKT cell-derived iPSCs into iNKT cells and their expansion in vitro, and (4) injection of iPSC-derived iNKT cells into tumor-bearing animals.<ref name="Koseki">Watarai H, Yamada D, Fujii S, Taniguchi M, Koseki H. (2012) Induced pluripotency as a potential path towards iNKT cell-mediated cancer immunotherapy. Int J Hematol. ;95(6):624-631. >{{cite doi|10.1007/s12185-012-1091-0}}</ref>

'''[[Dendritic cells]] (DC)''' are specialized to control T-cell responses. DC with appropriate genetic modifications may survive long enough to stimulate antigen-specific CTL and, after that, be completely eliminated. It was shown that DC-like antigen-presenting cells obtained from human induced pluripotent stem cells may serve as an unlimited source for [[vaccination]] therapy.<ref name="Senju">{{cite doi|10.1038/gt.2012.59}}</ref>

'''B cells'''. [[Ccaat-enhancer-binding proteins|CCAAT/enhancer binding protein-α (C/EBPα)]] induces transdifferentiation of [[B cell]]s into [[macrophage]]s at high efficiencies<ref>Xie, H., Ye, M., Feng, R. & Graf, T (2004) [http://www.sciencedirect.com/science/article/pii/S0092867404004192 Stepwise reprogramming of B cells into macrophages]. Cell 117(5), 663–676 >{{cite doi|10.1016/S0092-8674(04)00419-2}}</ref><ref>{{cite doi|10.1016/j.stem.2009.10.004}}ref>{{cite doi|10.1016/j.cell.2008.03.028}}</ref> with a 100-fold increase in iPS cell reprogramming efficiency, involving 95% of the population.<ref>{{cite doi|10.1038/nature12885}}</ref>
Furthermore, C/EBPa can convert at high efficiencies selected human B cell lymphoma and leukemia cell lines into macrophage-like cells, impairing the cells’ tumor-forming capacity.<ref>{{cite doi|10.1016/j.celrep.2013.03.003}}</ref>

=== Mesenchymal stem cells ===

==== Induction ====

Because of their immunosuppressive properties and ability to differentiate into a wide
range of mesenchymal-lineage tissues, [[Mesenchymal stem cell|mesenchymal stem/stromal cells]] (MSCs) are under
intense investigation for applications in cardiac, renal, neural, joint, and bone repair, as
well as in inflammatory conditions and hemopoietic cotransplantation.<ref name="Fu X">{{CITE DOI|10.1016/j.arr.2012.04.005}}</ref> MSCs are typically harvested from adult bone marrow or fat, but these not only require painful invasive procedures but are low-frequency sources, with MSCs making up only 0.001%– 0.01% of bone marrow cells and 0.05% in liposuction aspirates.<ref name="Bieback">Bieback K, Kern S, Kocaomer A et al. (2008) Comparing mesenchymal stromal cells from different human tissues: Bone marrow, adipose tissue and umbilical cord blood. Biomed Mater Eng; 18:S71–S76</ref> Of concern for autologous use, in particular in the elderly most in need of tissue repair, MSCs decline in quantity and quality with age.<ref name="Fu X" /><ref>{{cite doi|10.5966/sctm.2013-0014}}</ref><ref name="McGonagle">Stolzing A, Jones E, McGonagle D et al. (2008) Age-related changes in human bone marrow derived mesenchymal stem cells: Consequences for cell therapies. Mech Ageing Dev;129:163–173</ref>
However, induced pluripotent stem cells (iPSCs) could be obtained by the cells rejuvenation of even centenarian human.<ref name="Laure">{{cite doi|10.1101/gad.173922.111}}</ref> Because iPSCs can be harvested free of ethical constraints and culture can be expanded indefinitely, they are an advantageous source of MSCs.<ref name="Cantz">Irina Eberle, Mohsen Moslem, Reinhard Henschler, Tobias Cantz (2012) [http://link.springer.com/chapter/10.1007%2F10_2012_156?LI=true Engineered MSCs from Patient-Specific iPS Cells]. Advances in Biochemical Engineering Biotechnology</ref> Chen and colleagues discovered that iPSCs treatment with [[SB-431542]] (that inhibits the activin/TGF- pathways by blocking phosphorylation of ALK4, ALK5, and ALK7 receptors) leads to rapid and uniform MSC generation from human iPSCs. These iPS-MSCs possibly lack teratoma-forming ability, display a normal stable karyotype in culture, and exhibit growth and differentiation characteristics that closely resemble those of primary MSCs described, has considerable potential for the in vitro scale-up required to enable a wide range of MSC-based therapies.<ref name=" Fisk">{{cite doi|10.5966/sctm.2011-0022}}</ref> Unfortunately, currently there is no data on the in vivo efficacy and long-term safety of iPSC-derived MSCs generated by this method.

It is interesting to note that besides cell therapy in vivo the culture of human mesenchymal stem cells can be used in vitro for mass-production of [[Exosome (vesicle)|exosomes]], which, as it turned out, are ideal vehicles for drug delivery.<ref>Ruenn Chai Lai, Ronne Wee Yeh Yeo, Soon Sim Tan, Bin Zhang, et al. and Sai Kiang Lim (2013) Mesenchymal Stem Cell Exosomes: The Future MSC-Based Therapy? In: Mesenchymal Stem Cell Therapy. Chase, Lucas G.; Vemuri, Mohan C. (Eds.). 39-61 DOI
10.1007/978-1-62703-200-1_3</ref><ref>Ruenn Chai Lai, Ronne Wee Yeh Yeo, Kok Hian Tan, Sai Kiang Lim (2013) Exosomes for drug delivery — a novel application for the mesenchymal stem cell. Biotechnology Advances.http://dx.doi.org/10.1016/j.biotechadv.2012.08.008</ref><ref>Ronne Wee Yeh Yeoa, Ruenn Chai Laia, Bin Zhanga, et al. & Sai Kiang Lim (2012)Mesenchymal stem cell: An efficient mass producer of exosomes for drug delivery. Advanced Drug Delivery Reviewshttp://dx.doi.org/10.1016/j.addr.2012.07.001</ref><ref>Nobuyoshi Kosaka, Fumitaka Takeshita, Yusuke Yoshioka, et al. & Takahiro Ochiya (2012) Exosomal tumor-suppressive microRNAs as novel cancer therapy: «Exocure» is another choice for cancer treatment. Advanced Drug Delivery Reviewshttp://dx.doi.org/10.1016/j.addr.2012.07.011</ref>

==== Dedifferentiated adipocytes ====

Adipose tissue, because of its abundance and relatively less invasive harvest methods, represents a practical and appealing source of mesenchymal stem cells (MSCs). Unfortunately, there are only 0.05% MSCs in liposuction aspirates.<ref name="Bieback"/>{{cite doi|10.1002/stem.1067}}</ref><ref name="Sugawara">Shen JF, Sugawara A, Yamashita J, Ogura H, Sato S. (2011) Dedifferentiated fat cells: an alternative source of adult multipotent cells from the adipose tissues. Int J Oral Sci.;3(3):117-24</ref><ref>{{cite doi|10.5966/sctm.2012-0184}}</ref>

=== Induced chondrogenic cells ===

Cartilage is the connective tissue responsible for frictionless joint movement. Its degeneration ultimately results in complete loss of joint function in the late stages of osteoarthritis. As an avascular and hypocellular tissue, cartilage has a very limited capacity for self-repair. Chondrocytes are the only cell type in cartilage, in which they are surrounded by the extracellular matrix that they secrete and assemble.
One conceivable method of producing cartilage is to induce it from iPS cells.<ref>{{cite doi|10.1089/ten.teb.2012.0757}}</ref>
Alternatively, it is possible to convert fibroblasts directly into induced chondrogenic cells (iChon cells) without an intermediate iPS cell stage, by inserting three reprogramming factors (c-MYC, KLF4, and SOX9).<ref>{{cite doi|10.1371/journal.pone.0077365}}</ref>
The human iChon cells expressed marker genes for chondrocytes (type II collagen) but not fibroblasts.
The human iChon cells expressed marker genes for chondrocytes (type II collagen) but not fibroblasts.
Implanted into defects created in the articular cartilage of rats’, the human iChon cells were able to survive and form cartilaginous tissue for at least four weeks, but not tumors. Nevertheless, the method used in the study makes use of c-MYC, which is thought to have a major role in tumorigenesis, and employs a retrovirus to introduce the reprogramming factors. So, it could not be applied without modification in human therapy.
Implanted into defects created in the articular cartilage of rats’, the human iChon cells were able to survive and form cartilaginous tissue for at least four weeks, but not tumors. Nevertheless, the method used in the study makes use of c-MYC, which is thought to have a major role in tumorigenesis, and employs a retrovirus to introduce the reprogramming factors. So, it could not be applied without modification in human therapy.

Revision as of 06:09, 20 February 2014

Induced stem cells (iSC) are stem cells artificially derived from some other (somatic, reproductive, pluripotent etc.) cell types by induced (i.e., initiated, forced) epigenetic reprogramming. In accordance to the developmental potentiality and the degree of cell dedifferentiation caused by induced reprogramming they are distinguished and subdivided as: induced totipotent, induced pluripotent stem cells (iPSc) and, obtained by so-called direct reprogramming or directed forced differentiation, induced progenitor (multipotent or unipotent) stem cells, also called induced somatic stem cells. Three techniques are widely recognized:[1]

Metaplasia and Transdetermination

The reversible transformation of one differentiated cell type to another type of mature differentiated cells is called metaplasia.[21] This transition from one cell type to another can be a part of the normal maturation process, or caused by some of its inducing stimulus. For example: transformation of cells of the iris to the lens in the process of maturation and transformation of the retinal pigment epithelium cells into the neural retina during regeneration in adult newt eyes. This process allows the body to replace the original cells not suitable to new conditions, into new cells that are more suited to new conditions. In experiments on cells in Drosophila imaginal discs, it was found that there are a limited number of standard discrete states of differentiation and the cells have to choose one of them. The fact that transdetermination (change of the path of differentiation) often take place not in one, but in a group of cells shows that it is not caused by a mutation but is induced.[22][23]

Some types of mature, specialized adult cells can naturally revert to stem cells. For example, differentiated cells, which are called chief cells and express the stem cell marker Troy, normally produce digestive fluids for the stomach, yet they can change back into stem cells to make temporary repairs in significant stomach injuries, such as a cut or damage from infection. Moreover, they are making this transition even in the absence of noticeable injuries and are capable of replenishing entire gastric units, in essence serving as quiescent “reserve” stem cells.[24] Differentiated airway epithelial cells can revert into stable and functional stem cells in vivo.[25] After injury, mature terminally differentiated kidney cells dedifferentiate into more primordial versions of themselves, and then differentiate into the cell types needing replacement in the damaged tissue[26] Macrophages can self-renew by local proliferation of mature differentiated cells.[27] In Newts, muscle tissue is regenerated from specialized muscle cells that dedifferentiate and forget what type of cell they have been. This capacity to regenerate tissue does not decline with age, which may be linked to their ability to make new stem cells from muscle cells on demand.[28]

There are also a variety of nontumorigenic stem cells with the ability to generate the multiple cell types. For instance, multilineage-differentiating stress-enduring (Muse) cells are the stress-tolerant adult human stem cells that can self-renew; form characteristic cell clusters in suspension culture that express a set of genes associated with pluripotency; and can differentiate into endodermal, ectodermal, and mesodermal cells both in vitro and in vivo.[29][30][31][32][33]

Detailed description of some other well-documented examples of transdifferentiation, and their significance in development and regeneration are reviewed in.[34]

Induced totipotent cells usually can be obtained by reprogramming somatic cells by somatic-cell nuclear transfer (SCNT).

Cloning

Induced totipotent cells usually can be obtained by reprogramming somatic cells by somatic-cell nuclear transfer (SCNT) to the recipient eggs or oocytes.[4][35][36][37][38] Sometimes the oocytes of species such as sheep are used.[39][40] New possibilities for creating genetically modified animals opens method of induced androgenetic haploid embryonic stem cells, which can be used instead of sperm. These cells, synchronized in M phase and injected into the oocyte can produce viable offspring.[41] These developments, together with data on the possibility to obtain unlimited number of oocytes from mitotically active reproductive stem cells[42] offer the possibility of industrial production of transgenic farm animals. Repeated recloning of viable mice through a somatic cell nuclear transfer method that includes a histone deacetylase inhibitor – trichostatin, added to the cell culture medium,[43] show that it may be possible to reclone animals indefinitely without any visible accumulation of reprogramming or genomic errors [44] However, research into technologies to develop sperm and egg cells from stem cells bring up bioethical issues.

Such technologies may also have far-reaching clinical applications for overcoming cytoplasmic defects in human oocytes.[45][46] For example, the technology have been developed that could prevent inherited mitochondrial disease being passed on to the next generation. Mitochondria, often described as the powerhouse of the cell, contain genetic material, which is passed from mother to child. Mutations on mitochondrial DNA can cause diabetes, deafness, eye disorders, gastrointestinal disorders, heart disease, dementia and several other neurological diseases. The nucleus from one human egg cell have been transferred to another egg, in effect swapping the cell cytoplasm, which includes the mitochondria (and their DNA), creating a cell that could be regarded as having two mothers. The eggs were then fertilised, and the resulting embryonic stem cells carried the swapped mitochondrial DNA.[47]

In the case of late-generation telomerase-deficient (Terc−/−) mice, SCNT-mediated reprogramming mitigates telomere dysfunction and mitochondrial defects to a greater extent than iPSC-based reprogramming.[48]

Read more about the latest achievements of the cloning techniques and the generation of totipotent cells, in:[49]

Radical rejuvenation

First iPSc were obtained in the form of transplantable teratocarcinoma induced by the graft taken from mouse embryos.[50] It was shown that teratocarcinoma formed from somatic cells.[51] The fact that normal genetically mosaic mice can be obtained from malignant teratocarcinoma cells confirmed their pluripotency.[52][53][54] It turned out that teratocarcinoma cells are able to maintain a culture of pluripotent embryonic stem cells in an undifferentiated state, by supplying the culture medium with various factors.[55] Thus, as early as in the 1980s, it became clear that the transplantation of pluripotent or embryonic stem cells into the body of adult mammals, usually leads to the formation of teratomas, which can then turn into a malignant tumor teratocarcinoma.[56] If, however, to put the teratocarcinoma cells into the early mammal embryo (at the blastocyst stage), they became incorporated in the cell mass of blastocysts and from such a chimeric (i.e. composed of cells from different organisms) blastocyst often develops normal chimeric animal.[57][58][59] This indicated that the cause of the teratoma is a dissonance - mutual misunderstanding of "speech" of young donor cells and surrounding adult cells (so-called niche) of the recipient.

In August 2006, Japanese researchers circumvented the need for an oocyte, which is required for SCNT-mediated nuclear reprogramming method. By reprograming mouse embryonic fibroblasts into pluripotent stem cell via the ectopic expression of only four transcription factors, namely Oct4, Sox2, Klf4, and c-Myc, they proved that the overexpression of a small number of factors can sometimes push the cell to transition to a new stable state that is associated with changes in the activity of thousands of genes.[6] Obviously, mechanisms of reprogramming are linked, rather than independent, and are centered on a small number of genes.[60] The properties of IPSC were very similar to embryonic stem cells (ESCs). iPSCs have been shown to support the development of all-iPSC mice using tetraploid (4n) embryo,[61][62] the most stringent assay for developmental potential. However, some genetically normal iPSCs failed to give rise to all-iPSC mice on the score of aberrant epigenetic silencing of the imprinted Dlk1-Dio3 gene cluster.[18] An important advantage of iPSC to ESC is that they can be derived from adult cells, rather than from embryos. Therefore, it became possible to obtain iPSC from adults and even elderly patients[8]

Reprogramming somatic cells to iPSC leads to rejuvenation. It was found that reprogramming to iPSC leads to telomere lengthening and shortening after their subsequent differentiation back into fibroblast-like derivatives.[63] Thus, reprogramming of somatic cells to iPSC leads to the restoration of embryonic telomere length,[64] and hence increases the potential number of cell divisions limited by Hayflick limit.[65][66] Therefore, technology of iPSC should be seen as a radical way to rejuvenation. However, because of the dissonance between rejuvenated cells and their surrounding (so-called niche ) of older cells of the recipient, the injection of the patient by his own iPSC usually leads to an immune response,[67] which can be used for medical purposes,[68] or the formation of tumors such as teratoma.[69] The reason for this probably lies in the fact that some cells differentiated from ESC and iPSC in vivo continue to synthesize the embryonic isoforms of protein.[70] So, the immune system might detect and attack cells that are not cooperating properly.

Hope gives us study that showed that a small molecule called MitoBloCK-6 can force the pluripotent stem cells to die by triggering apoptosis (via cytochrome c release across the mitochondrial outer membrane in human pluripotent stem cells but not in differentiated cells), while shortly after differentiation their daughter cells became resistant to death. When MitoBloCK-6 introduced to differentiated cell lines, the cells remained healthy. The key to the survival of the differentiated cells, when the cells are exposed to MitoBloCK-6, may be due to the changes undergone by pluripotent stem cell mitochondria in the process of cell differentiation. This ability of MitoBloCK-6 to separate the pluripotent and differentiated cell lines have potential to reduce the risk of teratomas and other problems in regenerative medicine treatment strategies.[71] Recently been identified others small molecules (selective cytotoxic inhibitors of hPSCs) that prevented human pluripotent stem cells from forming teratomas in mice following transplantation. The most potent and selective compound of them (PluriSIn #1) inhibits stearoyl-coA desaturase (the key enzyme in oleic acid biosynthesis), which finally results in apoptosis. With the help of this molecule the undifferentiated cells can be selectively removed from culture.[72][73] An efficient strategy to selectively eliminate pluripotent cells with teratoma potential is targeting pluripotent stem cell-specific antiapoptotic factor(s) (i.e., survivin or Bcl10). Indeed, a single treatment of mixed population with chemical inhibitors of survivin (e.g., quercetin or YM155) can induce selective and complete cell death of undifferentiated hPSCs and, according to authors, is sufficient to prevent teratoma formation after transplantation of iPSC-derived cells.[74] However, it is unlikely that any kind of preliminary clearance, even the most sophisticated pre-treatment,[75] is able to secure the replanting iPSC or ESC, as after the selective removal of pluripotent cells, they re-emerge quickly by converting differentiated cells back into stem cells, which leads to the formation of tumors,[76][77] This may be due to the disorder of let-7 regulation of its target Nr6a1 (also known as Germ cell nuclear factor - GCNF), an embryonic transcriptional repressor of pluripotency genes that regulates gene expression in adult fibroblasts following miRNA loss.[78][79]

Teratoma formation by pluripotent stem cells may be caused by low activity of PTEN enzyme, reported to promote the survival of a small population (0,1-5% of total population) of highly tumorigenic, aggressive, teratoma-initiating embryonic-like carcinoma cells during differentiation. The survival of these teratoma-initiating cells is associated with failed repression of Nanog as well as a propensity for increased glucose and cholesterol metabolism.[80] These teratoma-initiating cells also expressed a lower ratio of p53/p21 when compared to non-tumorigenic cells.[81] In connection with the above safety problems, the use iPSC for cell therapy is still limited.[82][83] However, they can be used for a variety of other purposes - including the modeling of disease,[84][85] screening (selective selection) of drugs, toxicity testing of various drugs.[86][87]

It is interesting to note that the tissue grown from iPSCs, placed in the "chimeric" embryos in the early stages of mouse development, practically do not cause an immune response (after the embryos have grown into adult mice) and are suitable for autologous transplantation[88][89] At the same time, full reprogramming of adult cells in vivo within tissues by transitory induction of the four factors Oct4, Sox2, Klf4 and c-Myc in mice results in teratomas emerging from multiple organs.[90][91]

Chemical inducement

By using solely small molecules, Deng Hongkui and colleagues demonstrated that endogenous “master genes” are enough for cell fate reprogramming. They induced a pluripotent state in adult cells from mice using seven small-molecule compounds.[16] The effectiveness of the method is quite high: it was able to convert 0.2% of the adult tissue cells into iPSCs, which is comparable to the gene insertion conversion rate. The authors note that the mice generated from CiPSCs were "100% viable and apparently healthy for up to 6 months”.So. This chemical reprogramming strategy has potential use in generating functional desirable cell types for clinical applications.[92][93]

Differentiation from induced teratoma

The fact that human iPSCs capable of forming teratomas not only in humans but also in some animal body, in particular in mice or pigs, allowed to develop a method for differentiation of iPSCs in vivo. For this purpose, iPSCs with an agent for inducing differentiation into target cells are injected to genetically modified pig or mouse that has suppressed immune system activation on human cells. The formed after a while teratoma is cut out and used for the isolation of the necessary differentiated human cells[94] by means of monoclonal antibody to tissue-specific markers on the surface of these cells. This method has been successfully used for the production of functional myeloid, erythroid, and lymphoid human cells suitable for transplantation (yet only to mice).[95] Mice engrafted with human iPSC teratoma-derived hematopoietic cells produced human B and T cells capable of functional immune responses. These results offer hope that in vivo generation of patient customized cells is feasible, providing materials that could be useful for transplantation, human antibody generation, and drug screening applications. Using MitoBloCK-6 [71] and / or PluriSIn # 1 the differentiated progenitor cells can be further purified from teratoma forming pluripotent cells. The fact, that the differentiation takes place in a teratoma, offers hope that the resulting cells are sufficiently stable to stimuli able to cause their transition back to the dedifferentiated (pluripotent) state, and therefore safe. A similar in vivo differentiation system, yielding engraftable hematopoietic stem cells from mouse and human iPSCs in teratoma-bearing animals in combination with a maneuver to facilitate hematopoiesis, was described by Suzuki et al.[96] They noted that neither leukemia nor tumors were observed in recipients after intravenous injection of iPSC-derived hematopoietic stem cells into irradiated recipients. Moreover, this injection resulted in multilineage and long-term reconstitution of the hematolymphopoietic system in serial transfers. Such system provides a useful tool for practical application of iPSCs in the treatment of hematologic and immunologic diseases.[97]

For further development of this method animal in which is grown the human cell graft, for example mouse, must have so modified genome that all its cells express and have on its surface human SIRPα.[98] To prevent rejection after transplantation to the patient of the allogenic organ or tissue, grown from the pluripotent stem cells in vivo in the animal, these cells should express two molecules: CTLA4-Ig, which disrupts T cell costimulatory pathways, and PD-L1, which activates T cell inhibitory pathway.[99]

See also: US 20130058900  patent.

Differentiated cell types

Retinal cells

In the near-future, clinical trials designed to demonstrate the safety of the use of iPSCs for cell therapy of the people with age-related macular degeneration, a disease causing blindness through retina damaging, will begin. There are several articles describing methods for producing retinal cells from iPSCs[100] [101] and how to use them for cell therapy.[102][103] Reports of iPSC-derived retinal pigmented epithelium transplantation showed enhanced visual-guided behaviors of experimental animals for 6 weeks after transplantation.[104] However, clinical trials have been successful: ten patients suffering from retinitis pigmentosa have had their eyesight restored—including a woman who had only 17 percent of her vision left. [105]

Lung and airway epithelial cells

Chronic lung diseases such as idiopathic pulmonary fibrosis and cystic fibrosis or chronic obstructive pulmonary disease and asthma are leading causes of morbidity and mortality worldwide with a considerable human, societal, and financial burden. So there is an urgent need for effective cell therapy and lung tissue engineering.[106][107] Several protocols have been developed for generation of the most cell types of the respiratory system, which may be useful for deriving patient-specific therapeutic cells.[108][109][110][111][112]

Reproductive cells

Some lines of iPSCs have the potentiality to differentiate into male germ cells and oocyte-like cells in an appropriate niche (by culturing in retinoic acid and porcine follicular fluid differentiation medium or seminiferous tubule transplantation). Moreover, iPSC transplantation make a contribution to repairing the testis of infertile mice, demonstrating the potentiality of gamete derivation from iPSCs in vivo and in vitro.[113][114][115]

Induced progenitor stem cells

Direct transdifferentiation

Due to the fact that the use of iPSC for cell therapy is associated with significant risk of cancer and tumors there is urgent need to develop methods for safer cell lines suitable for use in the clinic. An alternative to the methods of iPSC technique is the so-called "direct reprogramming" - induced by certain factors direct transdifferentiation of cells without passing through the stages of the pluripotent state.[116][117][118][119][120][121] The basis for this approach laid the study of Taylor and Jones, who showed that 5-azacytidine - reagent causing demethylation of DNA - can cause the formation of myogenic, chondrogenic, and adipogenic clones in the immortal cell line of mouse embryonic fibroblasts[122] and Weintraub et al., who found that the activation of a single gene, later named MyoD1, is sufficient for such reprogramming,[123][124][125] Compared with iPSC whose reprogramming need at least two weeks, the formation of induced progenitor cells occurs relatively quickly - sometimes within a few days and the efficiency of reprogramming are usually many times higher. This reprogramming does not always require cell division.[126] But the main thing is that the resulting from such reprogramming multipotent somatic stem cells are more suitable for cell therapy because they do not form teratomas.[121]

Single transcription factor transdifferentiation

It was thought that only early embryonic cells could be coaxed into changing their identity. Mature cells are very resistant to changing their identity once they've committed to a specific kind. However, it has been found that brief expression of a single transcription factor, the ELT-7 GATA factor, can convert the identity of fully differentiated, highly specialized non-endodermal cells of the pharynx into fully differentiated intestinal cells in intact larvae and adult roundworm Caenorhabditis elegans. And there was no apparent requirement for a dedifferentiated intermediate during this transdifferentiation process.[127]

Phased process modeling regeneration

Another way of reprogramming is the simulation of the processes that occur during amphibian limb regeneration. In urodele amphibians, an early step in limb regeneration is skeletal muscle fiber dedifferentiation into a cellulate that proliferates to contribute new limb tissue, whereas mammalian muscle cannot dedifferentiate after injury. However, sequential small molecule treatment of the muscle fiber with myoseverin, reversine (the aurora B kinase inhibitor) and some other chemicals: BIO (glycogen synthase-3 kinase inhibitor), lysophosphatidic acid (pleiotropic activator of G-protein-coupled receptors), SB203580 (p38 MAP kinase inhibitor), or SQ22536 (adenylyl cyclase inhibitor) causes the formation of new muscle cell types as well as other cell types such as precursors to fat, bone, and nervous system cells.[128][129]

Antibody-based transdifferentiation

The researchers discovered GCSF-mimicking antibody that can activate a growth-stimulating receptor on marrow cells in a way that induces marrow stem cells—which normally develop into white blood cells—to become neural progenitor cells. The new technique,[130] that enables researchers to search large libraries of antibodies and quickly select the ones with a desired biological effect, has been used for such purpose[131][132]

Conditionally reprogrammed cells

Schlegel and Liu [133] demonstrated that the combination of feeder cells [134] (a review is given in[135] and[136]) and a Rho kinase inhibitor (Y-27632) [137][138] induces normal and tumor epithelial cells from many tissues to proliferate indefinitely in vitro and this process occurs without the need for transduction of exogenous viral or cellular genes. These cells has been termed as "Conditionally Reprogrammed Cells (CRC)". The induction of CRCs is rapid and results from reprogramming of the entire cell population rather than the selection of a minor subpopulation. CRCs do not express high levels of proteins characteristic of iPSCs or embryonic stem cells (ESCs) (e.g., Sox2, Oct4, Nanog, or Klf4). This induction of CRCs is reversible, and removal of Y-27632 and feeders allows the cells to differentiate normally.[133][139][140] CRC technology can generate 2×106 cells in 5 to 6 days from needle biopsies, and can generate cultures from cryopreserved tissue and from fewer than four viable cells. CRCs retain a normal karyotype and remain nontumorigenic. This technique also efficiently establishes cell cultures from human and rodent tumors.[133][141][142] The ability to rapidly generate many tumor cells from small biopsy specimens and frozen tissue provides significant opportunities for cell-based diagnostics and therapeutics (including chemosensitivity testing) and greatly expands the value of biobanking.[133][141][142] Using conditionally reprogrammed cells technology, researchers were able to identify an effective therapy for a patient with a rare type of lung tumor.[143] In addition, the CRC method allows for the genetic manipulation of epithelial cells ex vivo and their subsequent evaluation in vivo in the same host. While their initial studies revealed that co-culturing of epithelial cells with Swiss 3T3 cells J2 was essential for the induction of CRCs, Schlegel and Liu [144] have now shown with transwell culture plates that physical contact between feeders and epithelial cells is not required for inducing CRCs and, more important, that irradiation of the feeder cells is required for this induction. Consistent with the transwell experiments, conditioned medium is shown to induce and maintain CRCs, which is accompanied by a concomitant increase of cellular telomerase activity. The activity of the conditioned medium correlated directly with radiation-induced apoptosis of the feeder cells. Thus, conditional reprogramming of epithelial cells is mediated by a combination of Y-27632 and a soluble factor(s) released by apoptotic feeder cells.[144]

A different approach to obtain conditionally reprogrammed cells is to inhibit CD47 - a membrane protein that is the thrombospondin-1 receptor. It was shown that loss of CD47 permits sustained proliferation of primary murine endothelial cells, increases asymmetric division, and enables these cells to spontaneously reprogram to form multipotent embryoid body-like clusters. CD47 knockdown acutely increases mRNA levels of c-Myc and other stem cell transcription factors in cells in vitro and in vivo. It is obvious that thrombospondin-1 is a key environmental signal that inhibits stem cell self-renewal via CD47. Thus, CD47 antagonists enable cell self-renewal and reprogramming by overcoming negative regulation of c-Myc and other stem cell transcription factors.[145] Interesting to note that in vivo blockade of CD47 using an antisense morpholino increases survival of mice exposed to lethal total body irradiation due to increased proliferative capacity of bone marrow derived cells and radioprotection of radiosensitive gastrointestinal tissues.[146]

Indirect lineage conversion

A reprogramming methodology developed in which somatic cells transition through a plastic intermediate state of partially reprogrammed cells (pre-iPSC), induced by brief exposure to reprogramming factors, followed by differentiation by specially developed chemical environment (artificial niche).[147] It is assumed that this new method could be both more efficient and safer, since it does not seem to produce tumors or other undesirable genetic changes, and results in much greater yield than other methods. However, the safety of these cells all the same questionable - considering that lineage conversion from pre-iPSC relies on the use of iPSC reprogramming conditions, it can not be excluded that a fraction of the cells could acquire pluripotent properties if they do not stop the de-differentation process in vitro or due to further de-differentiation in vivo.

Outer membrane glycoprotein

A common feature of pluripotent stem cells, taken from different sources, is the specific nature of protein glycosylation of their outer membrane. That distinguishes them from most ( with the exception of white blood cells ) nonpluripotent cells.[148] Obviously, changes in the glycosylation of outer membrane proteins are markers of the state of the cells connected in some way with pluripotency and differentiation. The "shift" in the nature of glycosylation, apparently, is not just the result of the initialization of some genes expression, but the mechanism performing the role of an important regulator of genes involved in the acquisition and maintenance of the undifferentiated state.[149] For example, it is shown that activation of the glycoprotein ACA,[150] linking glycosylphosphatidylinositol on the surface of the progenitor cells in human peripheral blood, induces increased expression of genes Wnt, Notch-1, BMI1 and HOXB4 through a signaling cascade PI3K/Akt/mTor/PTEN, and promotes the formation of self-renewing population of hematopoietic stem cells[151] Furthermore, it is shown that dedifferentiation of progenitor cells induced by ACA- dependent signaling pathway leads to ACA- induced pluripotent stem cells, capable of differentiating in vitro into cells of all three germ layers.[152] The study of lectins on their ability to maintain a culture of pluripotent human stem cells has led to the discovery of lectin Erythrina Cristagalli (ECA), capable of serving as a simple and highly effective matrix for the cultivation of human pluripotent stem cells.[153]

Stimulus-triggered acquisition of pluripotency

File:Muse Cell Cluster.jpg
Cluster forming of pluripotent Muse/Stem cell

In Stimulus-triggered acquisition of pluripotency cell (STAP), strong external stimuli such as a transient low-pH stresspr reprogrammed mammalian somatic cells, produce pluripotent cells. STAP does not involve touching the DNA, nuclear transfer or the introduction of transcription factors.[154] Blastocyst injection showed that

STAP cells efficiently contribute to chimaeric embryos and to offspring via germline transmission. STAP cells, unlike embryonic stem cells and iPSC, have only a limited capacity for self-renewal, but can contribute to both embryonic and placental tissues. Mouse STAP cells lose the ability to contribute to the placenta as well as trophoblast marker expression on converting into ES-like stem cells by treatment with adrenocorticotropic hormone (ACTH) and leukemia inhibitory factor (LIF). In contrast, when cultured with Fgf4, STAP cells give rise to proliferative stem cells with enhanced trophoblastic characteristics. Unlike conventional trophoblast stem cells, the Fgf4-induced stem cells from STAP cells contribute to both embryonic and placental tissues in vivo and transform into ES-like cells (so-called STAP stem cells) when cultured with LIF-containing medium.[155]

Brain, skin, lung and liver cells have all been transformed using the technique. Conversion rates of 7.5% have been achieved, given that 25% of the cells survive the stress and 30% of those convert. Notably, three stressors — a bacterial toxin that perforates the cell membrane, exposure to low pH and physical squeezing — were each able to coax the cells to show markers of pluripotency.[29][30][31][32][33][156][157]

Muse cells are easily reprogrammed to iPSC.[158][159] In mature cells a strong stress activates a natural repair and transdetermination processes that permit them to revert to their "naive" cell state.

The remarkable properties demonstrated by STAP cells and STAP stem cells suggest that they could offer a simple and unobtrusive mechanism by which researchers can generate pluripotent cells for research—and perhaps even clinical—applications that previously required the laborious isolation of ES or iPS cells.[160]

Other labs have posted their experiences in attempting to reproduce the initial results on a blog.[161] As of February 2014 none had claimed to have successfully reprogrammed cells. Several scientists claimed to have contacted one or some of the authors for more details on the protocol with no response. Hongkui Deng, a stem-cell biologist at Peking University in Beijing, was told that “the authors will publish a detailed protocol soon”. Vacanti says he has had no problem repeating the experiment and says he will let Obokata supply the protocol “to avoid any potential for variation that could lead to confusion.”[162]

Reprogramming through a physical approach

Cell adhesion protein E-cadherin is indispensable for a robust pluripotent phenotype.[163] During reprogramming for iPS cell generation, N-cadherin can replace function of E-cadherin.[164] These functions of cadherins obviously are not directly related to adhesion because, according to Guannan Su et al.[165] sphere morphology helps maintaining the stemness of stem cells. Moreover, 3D sphere formation, due to forced growth of cells on low attachment surface, sometimes induces reprogramming. For example, neural progenitor cells can be generated from fibroblasts directly through a physical approach without introducing exogenous reprogramming factors.

Physical cues, in the form of parallel microgrooves on the surface of cell-adhesive substrates, can replace the effects of small-molecule epigenetic modifiers and significantly improve reprogramming efficiency. The mechanism relies on the mechanomodulation of the cells’ epigenetic state. To be specific, as the authors of this study believe: "decreased histone deacetylase activity and upregulation of the expression of WD repeat domain 5 (WDR5)—a subunit of H3 methyltranferase—by microgrooved surfaces lead to increased histone H3 acetylation and methylation". Nanofibrous scaffolds with aligned fibre orientation produce effects similar to those produced by microgrooves, suggesting that changes in cell morphology may be responsible for modulation of the epigenetic state.[166]

Mouse embryonic stem cells (mESCs) undergo self-renewal in the presence of the cytokine, leukemia inhibitory factor (LIF). Following LIF withdrawal, mESCs differentiate, and this is accompanied by an increase in cell–substratum adhesion and cell spreading. Restricted cell spreading in the absence of LIF by either culturing mESCs on chemically defined, weakly adhesive biomaterial substrates, or by manipulating the cytoskeleton allowed to keep the cells in an undifferentiated and pluripotent state. The effect of restricted cell spreading on mESC self-renewal is not mediated by increased intercellular adhesion, as evidenced by the observations that inhibition of mESC adhesion using a function blocking anti E-cadherin antibody or siRNA do not promote differentiation.[167]

The possible mechanisms of stem cell fate predetermining by physical interactions with the extracellular matrix see in reviews:[168][169]

Induced neural stem cells

Stroke and many neurodegenerative disorders such a: Parkinson's disease, Alzheimer’s disease, amyotrophic lateral sclerosis need cell replacement therapy. The successful use of converted neural cells (cNs) in transplantations open a new avenue to treat such diseases.[170] Nevertheless, induced neurons (iNs), directly converted from fibroblasts are terminally committed neurons that exhibit very limited proliferative ability and may not provide enough autologous donor cells for transplantation.[171] The generation of self-renewing induced neural stem cells (iNSCs) provides additional advantages over iNs for both basic research and clinical applications.[119][120][121][172][173] For example, under specific growth conditions, mouse fibroblasts can be reprogrammed with a single factor, Sox2, to form induced neural stem cells (iNSCs) that self-renew in culture and after transplantation can survive, integrate and do not form tumors in mouse brains.[174] Induced neural stem cells can also be derived from adult human fibroblasts by non-viral techniques, thus offering a safe method to generate iNSCs for autologous transplantation or for the development of cell-based disease models.[173] Methods of direct transformation of somatic cells into induced neural stem cells differ in their technique as shown in the review.[175]

Some data provide proof of principle that it is possible, directly in the brain, to convert transplanted human fibroblasts and human astrocytes, which are engineered to express inducible forms of neural reprogramming genes, into neurons, when reprogramming genes (Ascl11, Brn2a, and Myt1l) are activated after transplantation using a drug in the animals’ drinking water.[176] It was also shown that endogenous mouse astrocytes can be directly converted into neural nuclei (NeuN)-expressing neurons in situ.

Future studies will show which of these approaches proved to be most appropriate for clinic.

Oligodendrocyte precursor cells

Without myelin to insulate neurons, signals sent down nerve cell axons quickly lose power. Diseases that attack myelin, such as multiple sclerosis, result in nerve signals that are not efficient, because cannot propagate to nerve endings, and as a consequence to cognitive, motor, and sensory problems. Transplantation of oligodendrocyte precursor cells (OPCs), which can successfully create myelin sheaths around nerve cells, is a promising potential therapeutic strategy for diseases affecting myelin. However, there was no available source of engraftable OPCs. Therefore, direct lineage conversion of mouse and rat fibroblasts into oligodendroglial cells by forced expression of either eight[177] or of the three[178] transcription factors Sox10, Olig2 and Zfp536, laid the groundwork for therapies of a wide array of myelin disorders and spinal cord injury.

Induced cardiomyocytes

Development of regenerative therapeutic strategies to reverse the progression of advanced heart failure is one of the most urgent clinical needs of this century. Cell-based in vivo therapies may provide a transformative approach to augment vascular and muscle growth and to prevent non-contractile scar formation by delivering transcription factors[116] or microRNAs[13] to the heart.[179] For example: Qian L. et al. & Srivastava D. demonstrate that cardiac fibroblasts, which represent 50% of the cells in the mammalian heart, can be reprogrammed into cardiomyocyte-like cells in vivo by local delivery of cardiac core transcription factors ( GATA4, MEF2C, TBX5 and for improved reprogramming plus ESRRG, MESP1, Myocardin and ZFPM2) after coronary ligation.[116][180] These results implicated therapies that can directly remuscularize the heart without the need for cell transplantation. However, the efficiency of direct cardiac reprogramming in vivo by overexpression of cardiac core transcription factors in cardiac fibroblasts turned out to be very low and phenotype of received cardiomyocyte-like cells does not resemble those of a bona fide mature cardiomyocyte. Furthermore, transplantation of cardiac transcription factors into injured murine hearts resulted in poor cell survival and minimal expression of cardiac genes.[181] So, further technical improvements are needed to make this technology more applicable in situ. Meanwhile, there have been some advances in the methods of obtaining cardiac myocytes in vitro.[182] For example, Carpenter et al. demonstrated efficient cardiac differentiation of human iPS cells that gave rise to progenitors that were retained within the infarcted rat heart, and reduced remodeling of the heart after ischemic damage.[183] Furthermore, in ischaemic cardiomyopathy, caused by the murine infarction model, targeted iPS cell transplantation synchronized failing ventricles, offering a regenerative strategy to achieve resynchronization and protection from decompensation by dint of improved left ventricular conduction and contractility, reduced scar, and reversal of structural remodelling.[184] Palecek S P et al., have developed a protocol for generating almost pure populations of cardiomyocytes (up to 98% cardiomyocytes) from human pluripotent stem cells simply by modulating canonical Wnt signaling pathway at defined time points in the differentiation process, using readily accessible small molecule compounds.[185] Discovery of the mechanisms controlling the formation of cardiomyocytes led to the development of the drug (ITD-1), which effectively clears the cell surface from TGF-β receptor type II and selectively inhibits the intracellular TGF-β signaling and, thus, selectively enhances the differentiation of uncommitted mesoderm to cardiomyocytes, but not to vascular smooth muscle and endothelial cells.[186]

Lu et al.[187] create heart constructs by repopulating decellularized mouse hearts with human induced pluripotent stem cell-derived multipotential cardiovascular progenitor cells. They found that the seeded multipotential cardiovascular progenitor cells migrate, proliferate and differentiate in situ into cardiomyocytes, smooth muscle cells and endothelial cells to reconstruct the decellularized hearts. In addition, they observed that heart's extracellular matrix of mouse (the substrate of heart scaffold) can send signals to guide the repopulated human multipotential cardiovascular progenitor cells into becoming the specialised cells needed for proper heart function. After 20 days of perfusion with growth factors, the engineered heart tissues started to beat again and were responsive to drugs.

See also: Cell therapy in cardiovascular disease: selected research that has driven recent advances in clinical cardiology

Nephron Progenitors

Adult proximal tubule cells could be directly transcriptionally reprogrammed to nephron progenitors of the embryonic kidney, using a pool of six genes of instructive transcription factors (SIX1, SIX2, OSR1, Eyes absent homolog 1(EYA1), Homeobox A11 (HOXA11) and Snail homolog 2 (SNAI2)) that activated a network of genes consistent with a cap mesenchyme/nephron progenitor phenotype in the adult proximal tubule cell line.[188] The generation of such cells may lead to cellular therapies for adult renal disease. Indeed, it has recently been shown that embryonic kidney organoids placed into adult rat kidney can undergo onward development and vascular development.[189]

Bioengineering of the cells of blood vessels

Blood vessels build extensive networks that supply all cells with nutrients and oxygen throughout life. As blood vessels get older, they often become abnormal in structure and function, thereby contributing to numerous age-associated diseases including myocardial infarction, ischemic stroke and atherosclerosis of arteries supplying the heart, brain, and lower extremities. So, an important goal is to stimulate vascular growth for the collateral circulation to prevent the exacerbation of these diseases. A useful cell type for cell-based therapy designed to stimulate coronary collateral growth are Induced Vascular Progenitor Cells (iVPCs), generated by partially reprogramming endothelial cells.[147] The vascular commitment of iVPCs is related to the epigenetic memory of endothelial cells, which engenders them as cellular components of growing blood vessels. That is why, when iVPCs were implanted into myocardium, they engrafted in blood vessels and increased coronary collateral flow better than iPSCs, mesenchymal stem cells, or native endothelial cells.[190] An effective strategy to enhance stem cell function is ex vivo genetic modification. For example, genetic modification with Pim-1 kinase (a downstream effector of Akt, which positively regulates neovasculogenesis) of bone marrow–derived cells[191] or human cardiac progenitor cells, isolated from failing myocardium[192] results in durability of repair, together with the superior improvement of functional parameters of myocardial hemodynamic performance after cellular therapy of the injured myocardium with modified progenitor cells. Stem cells extracted from fat tissue after liposuction may be coaxed into progenitor smooth muscle cells (iPVSMCs) found in arteries and veins.[193] In the future, iVPCs and iPVSMCs may become a necessary source for the creation of blood vessels networks for tissue engineering and reconstruction of organs for transplantation. For instance, it was found that the 2D culture system of human iPS cells[194] in conjunction with the CD34 (a surface glycophosphoprotein expressed on developmentally early embryonic fibroblast), NP1 (receptor - neuropilin 1) and KDR (kinase insert domain-containing receptor) triple marker selection (for the isolation of vasculogenic precursor cells from human induced pluripotent stem cells) was capable to generate endothelial cells that, after transplantation, were able to form stable functional blood vessels in vivo, lasting for 280 d in mice.[195]

In the treatment of infarction, it is important to prevent the formation of fibrotic scar tissue. This can be achieved in vivo by means of transient application of paracrine factors changing the fate of the native heart progenitor stem cells from contributing to cardiac fibrotic scar tissue and toward cardiovascular tissue. For example, it was shown in a mouse myocardial infarction model that a single intramyocardial injection of human vascular endothelial growth factor-A mRNA (VEGF-A modRNA), synthetically modified so that it escapes the normal defense system of the body, results in long-term improvement of heart function due to mobilization of epicardial progenitor cells and redirection of their differentiation toward cardiovascular cell types [196]

Bioengineering of blood stem cells

Definitive hematopoiesis emerges during embryogenesis via an endothelial-to-hematopoietic transition. Fairly simple combination of four transcription factors, Gata2, Gfi1b, cFos, and Etv6, is sufficient to induce in vitro this complex, dynamic, and multistep developmental program leading to the formation of endothelial-like precursor cells, with the subsequent appearance of hematopoietic cells.[197]

Red blood cells

Red blood cells (RBC) transfusion is necessary for many patients with emergency or hematological disorders. However, to date the supply of RBCs remains labile and dependent on voluntary donations. In addition, the transmission of infectious disease via blood transfusion from unspecified donors remains a risk. Establishing a large quantity of safe RBCs would help to address this issue. New technologies for ex vivo erythroid cell generation will hopefully provide alternative transfusion products to meet present and future clinical requirements.[198][199] In favor of this suggest data that RBCs generated in vitro from mobilized CD34 positive cells have normal survival when transfused into an autologous recipient.[200] However, the RBC produced in vitro contained exclusively fetal hemoglobin (HbF), which rescues the functionality of these RBCs. However, in vivo the switch of fetal to adult hemoglobin after infusion of nucleated erythroid precursors derived from induced pluripotent stem cells was observed.[201] But then there is another problem: although RBCs do not have nuclei, and therefore can not form a tumor, their immediate precursors the erythroblasts do. The terminal maturation of erythroblasts into functional RBCs requires a complex remodeling process that ends with extrusion of the nucleus and the formation of an enucleated RBC.[202] Alas, cell reprogramming methodologies at present often disrupt these processes of enucleation, and therefore transfusion of in vitro-generated RBCs or their immediate precursors the erythroblasts still insufficiently protected against the possibility of tumors formation. However, Bouhassira and colleagues recently found that exposing CD34 positive cells to a short pulse of cytokines favorable for erythroid differentiation prior to stem cell expansion followed by progenitor expansion produced on the order more yield of erythroid cells than the yields observed previously. And the most important: These red blood cells expressed a globin profile similar to that of the developmental age of the CD34 positive cells[203] It is also interesting to note that an important role in normal blood cell development plays the aryl hydrocarbon receptor (AhR) pathway (which has been shown to be involved in the promotion of cancer cell development). AhR activation in human hematopoietic progenitor cells (HPs) drives an unprecedented expansion of HPs, megakaryocyte- and erythroid-lineage cells.[204]

See also: Migliaccio AR, Whitsett C, Papayannopoulou T, Sadelain M. (2012) The potential of stem cells as an in vitro source of red blood cells for transfusion. Review. Cell Stem Cell.;10(2):115-9

Platelets

Platelets play an important role in preventing hemorrhage in thrombocytopenic patients and patients with thrombocythemia. A significant problem for multitransfused patients is refractoriness to platelet transfusions. Thus, the ability to generate platelet products ex vivo and platelet products lacking HLA antigens in serum free media, would have great clinical value. Some success in this direction has achieved Figueiredo et al. Using an RNA interference-based mechanism in which a lentiviral vector was used to express short-hairpin RNAi targeting β2-microglobulin transcripts in CD34-positive cells, they generated platelets demonstrating an 85% reduction in class I HLA antigens. These platelets appeared to have normal function in vitro[205] A clinically applicable strategy for the derivation of functional platelets from human iPSC was developed, that involves the establishment of stable immortalized megakaryocyte progenitor cell lines (imMKCLs) through the doxycycline-dependent overexpression of BMI1 and BCL-XL. The resulting imMKCLs can be expanded in culture over extended periods (4–5 months), even after cryopreservation. Halting the overexpression (by the removal of doxycycline from the medium) of c-MYC, BMI1, and BCL-XL in growing imMKCLs led to the production of CD42b+ platelets with functionality comparable to that of native platelets on the basis of a range of assays in vitro and in vivo.[206]

Immune cells

A specialised type of white blood cell, known as cytotoxic T lymphocytes (CTLs), are produced by the immune system and are able to recognise specific markers on the surface of various infectious or tumour cells, causing them to launch an attack to kill the harmful cells. Thence, immunotherapy with functional antigen-specific T cells has potential to be an effective therapeutic strategy for combating many types of cancer and viral infection.[207] However, sources of such cells for therapeutic purposes are limited, because they are produced in small numbers naturally and have a short lifespan. A potentially efficient approach for generating antigen-specific CTLs is to revert mature immune T cells into iPSCs, which possess indefinite proliferative capacity in vitro, and after their multiplication to coaxed them to redifferentiate back into T cells[208][209][210] Even more opportunities promises a method that combine iPSC and chimeric antigen receptor (CAR) [211] technologies to generate human T cells targeted to CD19, an antigen expressed by malignant B cells, in tissue culture.[212] This approach of generating therapeutic human T cells 'in the dish' may be useful for cancer immunotherapy and other medical applications because such 'living drugs' have fewer side-effects, once injected - stay in the body and move around to stop recurrences.

Invariant natural killer T (iNKT) cells has great clinical potential as adjuvant for cancer immunotherapy. iNKT cells act as innate T lymphocytes and serve as a bridge between the innate and acquired immune systems. They augment anti-tumor responses by producing interferon-gamma (IFN-γ).[213] A conceptual method is proposed for the use of iPSC-derived iNKT cells for adjuvant cell therapy against cancer, which is composed of four segments: (1) collection of iNKT cells, (2) reprogramming of iNKT cells into iPSCs, (3) re-differentiation of iNKT cell-derived iPSCs into iNKT cells and their expansion in vitro, and (4) injection of iPSC-derived iNKT cells into tumor-bearing animals.[214]

Dendritic cells (DC) are specialized to control T-cell responses. DC with appropriate genetic modifications may survive long enough to stimulate antigen-specific CTL and, after that, be completely eliminated. It was shown that DC-like antigen-presenting cells obtained from human induced pluripotent stem cells may serve as an unlimited source for vaccination therapy.[215]

B cells. CCAAT/enhancer binding protein-α (C/EBPα) induces transdifferentiation of B cells into macrophages at high efficiencies<refAttention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1016/S0092-8674(04)00419-2, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1016/S0092-8674(04)00419-2 instead.</ref>[216] and enhances reprogramming into iPS cells when co-expressed with the transcription factors Oct4, Sox2, Klf4 and Myc[217] with a 100-fold increase in iPS cell reprogramming efficiency, involving 95% of the population.[218] Furthermore, C/EBPa can convert at high efficiencies selected human B cell lymphoma and leukemia cell lines into macrophage-like cells, impairing the cells’ tumor-forming capacity.[219]

Mesenchymal stem cells

Induction

Because of their immunosuppressive properties and ability to differentiate into a wide range of mesenchymal-lineage tissues, mesenchymal stem/stromal cells (MSCs) are under intense investigation for applications in cardiac, renal, neural, joint, and bone repair, as well as in inflammatory conditions and hemopoietic cotransplantation.Cite error: The <ref> tag has too many names (see the help page). Of concern for autologous use, in particular in the elderly most in need of tissue repair, MSCs decline in quantity and quality with age.[220][221][222] However, induced pluripotent stem cells (iPSCs) could be obtained by the cells rejuvenation of even centenarian human.[8] Because iPSCs can be harvested free of ethical constraints and culture can be expanded indefinitely, they are an advantageous source of MSCs.[223] Chen and colleagues discovered that iPSCs treatment with SB-431542 (that inhibits the activin/TGF- pathways by blocking phosphorylation of ALK4, ALK5, and ALK7 receptors) leads to rapid and uniform MSC generation from human iPSCs. These iPS-MSCs possibly lack teratoma-forming ability, display a normal stable karyotype in culture, and exhibit growth and differentiation characteristics that closely resemble those of primary MSCs described, has considerable potential for the in vitro scale-up required to enable a wide range of MSC-based therapies.[224] Unfortunately, currently there is no data on the in vivo efficacy and long-term safety of iPSC-derived MSCs generated by this method.

It is interesting to note that besides cell therapy in vivo the culture of human mesenchymal stem cells can be used in vitro for mass-production of exosomes, which, as it turned out, are ideal vehicles for drug delivery.[225][226][227][228]

Dedifferentiated adipocytes

Adipose tissue, because of its abundance and relatively less invasive harvest methods, represents a practical and appealing source of mesenchymal stem cells (MSCs). Unfortunately, there are only 0.05% MSCs in liposuction aspirates.[229] However, a large amount of mature adipocytes, which, in general, lost their proliferative abilities, and therefore often discarded, can be easily isolated from the adipose cell suspension and dedifferentiated into lipid-free fibroblast-like cells, named dedifferentiated fat (DFAT) cells. DFAT cells re-establish active proliferation ability and undertake multipotent capacities.[230] Compared with adult stem cells, DFAT cells showed unique advantages in their abundance, isolation, and homogeneity. Under proper induction culture in vitro or environment in vivo, DFAT cells could demonstrate adipogenic, osteogenic, chondrogenic, and myogenic potentials. They also could exhibit perivascular characteristics and elicit neovascularization.[231][232][233]

Induced chondrogenic cells

Cartilage is the connective tissue responsible for frictionless joint movement. Its degeneration ultimately results in complete loss of joint function in the late stages of osteoarthritis. As an avascular and hypocellular tissue, cartilage has a very limited capacity for self-repair. Chondrocytes are the only cell type in cartilage, in which they are surrounded by the extracellular matrix that they secrete and assemble. One conceivable method of producing cartilage is to induce it from iPS cells.[234] Alternatively, it is possible to convert fibroblasts directly into induced chondrogenic cells (iChon cells) without an intermediate iPS cell stage, by inserting three reprogramming factors (c-MYC, KLF4, and SOX9).[235] The human iChon cells expressed marker genes for chondrocytes (type II collagen) but not fibroblasts. Implanted into defects created in the articular cartilage of rats’, the human iChon cells were able to survive and form cartilaginous tissue for at least four weeks, but not tumors. Nevertheless, the method used in the study makes use of c-MYC, which is thought to have a major role in tumorigenesis, and employs a retrovirus to introduce the reprogramming factors. So, it could not be applied without modification in human therapy.Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1016/j.stem.2012.11.002, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1016/j.stem.2012.11.002 instead.</ref>Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1016/j.stem.2012.12.006, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1016/j.stem.2012.12.006 instead.</ref>[210][236] Even more opportunities promises a method that combine iPSC and chimeric antigen receptor (CAR) [237] technologies to generate human T cells targeted to CD19, an antigen expressed by malignant B cells, in tissue culture.[238] This approach of generating therapeutic human T cells 'in the dish' may be useful for cancer immunotherapy and other medical applications because such 'living drugs' have fewer side-effects, once injected - stay in the body and move around to stop recurrences.

Invariant natural killer T (iNKT) cells has great clinical potential as adjuvant for cancer immunotherapy. iNKT cells act as innate T lymphocytes and serve as a bridge between the innate and acquired immune systems. They augment anti-tumor responses by producing interferon-gamma (IFN-γ).[239] A conceptual method is proposed for the use of iPSC-derived iNKT cells for adjuvant cell therapy against cancer, which is composed of four segments: (1) collection of iNKT cells, (2) reprogramming of iNKT cells into iPSCs, (3) re-differentiation of iNKT cell-derived iPSCs into iNKT cells and their expansion in vitro, and (4) injection of iPSC-derived iNKT cells into tumor-bearing animals.[214]

Dendritic cells (DC) are specialized to control T-cell responses. DC with appropriate genetic modifications may survive long enough to stimulate antigen-specific CTL and, after that, be completely eliminated. It was shown that DC-like antigen-presenting cells obtained from human induced pluripotent stem cells may serve as an unlimited source for vaccination therapy.[215]

B cells. CCAAT/enhancer binding protein-α (C/EBPα) induces transdifferentiation of B cells into macrophages at high efficiencies[240][241] with a 100-fold increase in iPS cell reprogramming efficiency, involving 95% of the population.[242] Furthermore, C/EBPa can convert at high efficiencies selected human B cell lymphoma and leukemia cell lines into macrophage-like cells, impairing the cells’ tumor-forming capacity.[243]

Mesenchymal stem cells

Induction

Because of their immunosuppressive properties and ability to differentiate into a wide range of mesenchymal-lineage tissues, mesenchymal stem/stromal cells (MSCs) are under intense investigation for applications in cardiac, renal, neural, joint, and bone repair, as well as in inflammatory conditions and hemopoietic cotransplantation.[220] MSCs are typically harvested from adult bone marrow or fat, but these not only require painful invasive procedures but are low-frequency sources, with MSCs making up only 0.001%– 0.01% of bone marrow cells and 0.05% in liposuction aspirates.[229] Of concern for autologous use, in particular in the elderly most in need of tissue repair, MSCs decline in quantity and quality with age.[220][244][222] However, induced pluripotent stem cells (iPSCs) could be obtained by the cells rejuvenation of even centenarian human.[8] Because iPSCs can be harvested free of ethical constraints and culture can be expanded indefinitely, they are an advantageous source of MSCs.[223] Chen and colleagues discovered that iPSCs treatment with SB-431542 (that inhibits the activin/TGF- pathways by blocking phosphorylation of ALK4, ALK5, and ALK7 receptors) leads to rapid and uniform MSC generation from human iPSCs. These iPS-MSCs possibly lack teratoma-forming ability, display a normal stable karyotype in culture, and exhibit growth and differentiation characteristics that closely resemble those of primary MSCs described, has considerable potential for the in vitro scale-up required to enable a wide range of MSC-based therapies.[224] Unfortunately, currently there is no data on the in vivo efficacy and long-term safety of iPSC-derived MSCs generated by this method.

It is interesting to note that besides cell therapy in vivo the culture of human mesenchymal stem cells can be used in vitro for mass-production of exosomes, which, as it turned out, are ideal vehicles for drug delivery.[245][246][247][248]

Dedifferentiated adipocytes

Adipose tissue, because of its abundance and relatively less invasive harvest methods, represents a practical and appealing source of mesenchymal stem cells (MSCs). Unfortunately, there are only 0.05% MSCs in liposuction aspirates.[229]Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1002/stem.1067, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1002/stem.1067 instead.</ref>[232][249]

Induced chondrogenic cells

Cartilage is the connective tissue responsible for frictionless joint movement. Its degeneration ultimately results in complete loss of joint function in the late stages of osteoarthritis. As an avascular and hypocellular tissue, cartilage has a very limited capacity for self-repair. Chondrocytes are the only cell type in cartilage, in which they are surrounded by the extracellular matrix that they secrete and assemble. One conceivable method of producing cartilage is to induce it from iPS cells.[250] Alternatively, it is possible to convert fibroblasts directly into induced chondrogenic cells (iChon cells) without an intermediate iPS cell stage, by inserting three reprogramming factors (c-MYC, KLF4, and SOX9).[251] The human iChon cells expressed marker genes for chondrocytes (type II collagen) but not fibroblasts. Implanted into defects created in the articular cartilage of rats’, the human iChon cells were able to survive and form cartilaginous tissue for at least four weeks, but not tumors. Nevertheless, the method used in the study makes use of c-MYC, which is thought to have a major role in tumorigenesis, and employs a retrovirus to introduce the reprogramming factors. So, it could not be applied without modification in human therapy.

Sources of cells for reprogramming

The most frequently used source for reprogramming are blood cells.[252][253][254][255] and fibroblasts, obtained by biopsy of the skin,[256] but more convenient to receive the body cells from the urine.[257][258][259][260] This method does not require a biopsy or blood sampling and therefore harmless to the patient. Urine-derived stem cells have capacity for multipotent differentiation. They are able to differentiate into endothelial, osteogenic, chondrogenic, adipogenic, skeletal myogenic and neurogenic lineages, but did not form teratomas.[261] Therefore, their epigenetic memory better suited to the reprogramming into iPS cells. However, there are very few cells in the urine, the efficiency of turning them into induced stem cells is low, whereas the risk of bacterial contamination higher than with other sources of cells.

Another promising source of cells for reprogramming are mesenchymal stem cells derived from human hair follicles.[262]

It is important to note that the origin of somatic cells used for reprogramming may influence the efficiency of reprogramming,[263][264] the functional properties of the resulting induced stem cells[265] and the ability to form tumors.[266][267]

While selecting the source for reprogramming, into account should be taken the fact that iPSCs retain an epigenetic memory of their tissue of origin, and that this impacts their differentiation potential[210][265][268][269][270][271] This epigenetic memory does not necessarily manifest itself at the pluripotency stage – iPSCs derived from different tissues exhibit proper morphology, express pluripotency markers, and are able to differentiate into the three embryonic layers in vitro and in vivo. However, this epigenetic memory may manifest later, during re-differentiation into specific cell types that require the specific loci that have residual epigenetic marks.

See also

Notes

References for further reading

References

  1. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1038/nature09229, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1038/nature09229 instead.
  2. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1016/j.cell.2013.05.006, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1016/j.cell.2013.05.006 instead.
  3. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1038/nature10397, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1038/nature10397 instead.
  4. ^ a b Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1002/bies.201100174, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1002/bies.201100174 instead.
  5. ^ Attention: This template ({{cite pmid}}) is deprecated. To cite the publication identified by PMID 17218392, please use {{cite journal}} with |pmid=17218392 instead.
  6. ^ a b Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1016/j.cell.2007.11.019, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1016/j.cell.2007.11.019 instead.
  7. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1073/pnas.1100893108, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1073/pnas.1100893108 instead.
  8. ^ a b c d Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1101/gad.173922.111, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1101/gad.173922.111 instead. Cite error: The named reference "Laure" was defined multiple times with different content (see the help page).
  9. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1016/j.stem.2009.04.005, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1016/j.stem.2009.04.005 instead.
  10. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1002/9780470151808.sc04a04s20, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1002/9780470151808.sc04a04s20 instead.
  11. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1016/j.stem.2011.03.001, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1016/j.stem.2011.03.001 instead.
  12. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1016/j.stem.2011.05.001, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1016/j.stem.2011.05.001 instead.
  13. ^ a b Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1161/CIRCRESAHA.112.269035, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1161/CIRCRESAHA.112.269035 instead.
  14. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1016/j.ceb.2012.12.004, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1016/j.ceb.2012.12.004 instead.
  15. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1016/j.stem.2013.06.001, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1016/j.stem.2013.06.001 instead.
  16. ^ a b Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1126/science.1239278, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1126/science.1239278 instead.
  17. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1098/rstb.2011.0006, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1098/rstb.2011.0006 instead.
  18. ^ a b Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1038/ng.1110, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1038/ng.1110 instead.
  19. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1002/biot.201100361, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1002/biot.201100361 instead.
  20. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1038/srep00544, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1038/srep00544 instead.
  21. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1002/path.2442, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1002/path.2442 instead.
  22. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1634/stemcells.18-6-409, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1634/stemcells.18-6-409 instead.
  23. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1146/annurev-genet-110711-155637, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1146/annurev-genet-110711-155637 instead.
  24. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1016/j.cell.2013.09.008, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1016/j.cell.2013.09.008 instead.
  25. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1038/nature12777, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1038/nature12777 instead.
  26. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1073/pnas.1310653110, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1073/pnas.1310653110 instead.
  27. ^ Sieweke, Michael H., Judith E. Allen (2013) Beyond Stem Cells: Self-Renewal of Differentiated Macrophages. Science : 342(6161) DOI: 10.1126/science.1242974
  28. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1016/j.stem.2013.11.007, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1016/j.stem.2013.11.007 instead.
  29. ^ a b Kuroda Y, Wakao S, Kitada M, Murakami T, Nojima M, Dezawa M. (2013). Isolation, culture and evaluation of multilineage-differentiating stress-enduring (Muse) cells. Nat Protoc.;8(7):1391-415. doi: 10.1038/nprot.2013.076.
  30. ^ a b Kuroda, Y., Kitada, M., Wakao, S., et al. & Dezawa, M. (2010) Unique multipotent cells in adult human mesenchymal cell populations. PNAS , 107(19), 8639-8643. doi:10.1073/pnas.0911647107
  31. ^ a b Ogura F, Wakao S, Kuroda Y, Tsuchiyama K, Bagheri M, Heneidi S, Chazenbalk G, Aiba S, Dezawa M. (2014). Human Adipose Tissue Possesses a Unique Population of Pluripotent Stem Cells with Nontumorigenic and Low Telomerase Activities: Potential Implications in Regenerative Medicine. Stem Cells Dev. Epub ahead of print
  32. ^ a b Heneidi S, Simerman AA, Keller E, Singh P, Li X, et al. (2013). Awakened by Cellular Stress: Isolation and Characterization of a Novel Population of Pluripotent Stem Cells Derived from Human Adipose Tissue. PLoS ONE 8(6): e64752. doi:10.1371/journal.pone.0064752
  33. ^ a b Shigemoto T, Kuroda Y, Wakao S, Dezawa M (2013). A Novel Approach to Collecting Satellite Cells From Adult Skeletal Muscles on the Basis of Their Stress Tolerance. Stem Cells Trans Med 2 (7) 488-498 doi:10.5966/sctm.2012-0130
  34. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1007/s00441-012-1403-y, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1007/s00441-012-1403-y instead.
  35. ^ Cite error: The named reference Masahito Tachibana was invoked but never defined (see the help page).
  36. ^ Dinnyes, Andras; Tian, Xiuchun Cindy; Oback, Bj¨orn (17 April 2013). Robert A. Meyers (ed.). Nuclear Transfer for Cloning Animals. John Wiley & Sons. pp. 299–344. ISBN 978-3-527-66854-0. {{cite book}}: |work= ignored (help)
  37. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1038/nrm3140, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1038/nrm3140 instead.
  38. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1046/j.1469-7580.2002.00035.x, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1046/j.1469-7580.2002.00035.x instead.
  39. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1089/cell.2011.0061, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1089/cell.2011.0061 instead.
  40. ^ US 8,647,872  patent
  41. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1016/j.cell.2012.04.002, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1016/j.cell.2012.04.002 instead.
  42. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1126/science.1226889, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1126/science.1226889 instead.
  43. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1016/j.bbrc.2005.11.164, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1016/j.bbrc.2005.11.164 instead.
  44. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1016/j.stem.2013.01.005, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1016/j.stem.2013.01.005 instead.
  45. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1016/j.cell.2013.05.006, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1016/j.cell.2013.05.006 instead.
  46. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1038/nature11800, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1038/nature11800 instead.
  47. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1038/nature11647, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1038/nature11647 instead.
  48. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1016/j.stem.2013.11.005, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1016/j.stem.2013.11.005 instead.
  49. ^ Cibelli, Jose; Lanza, Robert; Campbell, Keith H.S.; West, Michael D. (14 September 2002). Principles of Cloning. Academic Press. ISBN 978-0-08-049215-5.
  50. ^ Attention: This template ({{cite pmid}}) is deprecated. To cite the publication identified by PMID 5436899, please use {{cite journal}} with |pmid=5436899 instead.
  51. ^ Attention: This template ({{cite pmid}}) is deprecated. To cite the publication identified by PMID 275854, please use {{cite journal}} with |pmid=275854 instead.
  52. ^ Mintz B, Illmensee K. (1975) Normal genetically mosaic mice produced from malignant teratocarcinoma cells. Proc Natl Acad Sci U S A; 72 (9) :3585-3589
  53. ^ Template:Cite pmcid MARTIN, G. R. & EVANS, M. J. (1975). Differentiation of clonal lines of teratocarcinoma cells: formation of embryoid bodies in vitro. Proc. Natn. Acad. Sci. U.S.A. 72, 1441-1445
  54. ^ Attention: This template ({{cite pmid}}) is deprecated. To cite the publication identified by PMID 1061157, please use {{cite journal}} with |pmid=1061157 instead.
  55. ^ Template:Cite pmcid Martin, GR (1981) Isolation of a pluripotent cell line from early mouse embryos cultured in medium conditioned by teratocarcinoma stem cells. Proc. Natl. Acad. Sci. USA 78, 7634-7638
  56. ^ Attention: This template ({{cite pmid}}) is deprecated. To cite the publication identified by PMID 6250214, please use {{cite journal}} with |pmid=6250214 instead.
  57. ^ Attention: This template ({{cite pmid}}) is deprecated. To cite the publication identified by PMID 650144, please use {{cite journal}} with |pmid=650144 instead.
  58. ^ GRAHAM, C. F. (January 1977). Teratocarcinoma cells and normal mouse embryogenesiseditor=Michael I. Sherman. MIT Press. ISBN 978-0-262-19158-6. {{cite book}}: |work= ignored (help)
  59. ^ ILLMENSEE, K. (14 June 2012). L. B. Russell (ed.). Reversion of malignancy and normalized differentiation of teratocarcinoma cells in chimeric mice. Springer London, Limited. pp. 3–24. ISBN 978-1-4684-3392-0. {{cite book}}: |work= ignored (help)
  60. ^ Stuart, H. T. et al., (2014). NANOG Amplifies STAT3 Activation and They Synergistically Induce the Naive Pluripotent Program. Current Biology, http://dx.doi.org/10.1016/j.cub.2013.12.040
  61. ^ Attention: This template ({{cite pmid}}) is deprecated. To cite the publication identified by PMID 19672243, please use {{cite journal}} with |pmid=19672243 instead.
  62. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1016/j.stem.2009.07.001, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1016/j.stem.2009.07.001 instead.
  63. ^ Attention: This template ({{cite pmid}}) is deprecated. To cite the publication identified by PMID 20861676, please use {{cite journal}} with |pmid=20861676 instead.
  64. ^ Attention: This template ({{cite pmid}}) is deprecated. To cite the publication identified by PMID 20632849, please use {{cite journal}} with |pmid=20632849 instead.
  65. ^ Attention: This template ({{cite pmid}}) is deprecated. To cite the publication identified by PMID 20176474, please use {{cite journal}} with |pmid=20176474 instead.
  66. ^ Attention: This template ({{cite pmid}}) is deprecated. To cite the publication identified by PMID 21605571, please use {{cite journal}} with |pmid=21605571 instead.
  67. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1038/nature10135, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1038/nature10135 instead.
  68. ^ Attention: This template ({{cite pmid}}) is deprecated. To cite the publication identified by PMID 21104412, please use {{cite journal}} with |pmid=21104412 instead.
  69. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1002/stem.471, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1002/stem.471 instead.
  70. ^ CAttention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1371/journal.pone.0025761, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1371/journal.pone.0025761 instead.
  71. ^ a b Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1016/j.devcel.2013.03.006, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1016/j.devcel.2013.03.006 instead.
  72. ^ Uri Ben-David, Qing-Fen Gan, Tamar Golan-Lev, et al & Nissim Benvenisty (2013)Selective Elimination of Human Pluripotent Stem Cells by an Oleate Synthesis Inhibitor Discovered in a High-Throughput Screen Cell Stem Cell, 12(2), 167-179 http://dx.doi.org/10.1016/j.stem.2012.11.015
  73. ^ {{cite doi|10.1038/scibx.2013.158
  74. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1073/pnas.1303669110, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1073/pnas.1303669110 instead.
  75. ^ Attention: This template ({{cite pmid}}) is deprecated. To cite the publication identified by PMID 22294556, please use {{cite journal}} with |pmid=22294556 instead.
  76. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1073/pnas.1102454108, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1073/pnas.1102454108 instead.
  77. ^ doi:10.1089/scd.2011.0131
  78. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1101/gad.215376.113, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1101/gad.215376.113 instead.
  79. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1002/stem.1367, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1002/stem.1367 instead.
  80. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1371/journal.pone.0016478, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1371/journal.pone.0016478 instead.
  81. ^ Grad, I., Hibaoui, Y., Jaconi,. et al. & Feki, A. (2011) NANOG priming before full reprogramming may generate germ cell tumours. Eur. Cell Mater, 22, 258-274 PMID 22071697
  82. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1161/CIRCRESAHA.111.256149, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1161/CIRCRESAHA.111.256149 instead.
  83. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1038/nbt.2329, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1038/nbt.2329 instead.
  84. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1038/nrm3448, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1038/nrm3448 instead.
  85. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1038/nn.3425, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1038/nn.3425 instead.
  86. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1242/dev.092551, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1242/dev.092551 instead.
  87. ^ Attention: This template ({{cite pmid}}) is deprecated. To cite the publication identified by PMID 22741526, please use {{cite journal}} with |pmid=22741526 instead.
  88. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1038/nature11807, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1038/nature11807 instead.
  89. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1182/blood-2012-11-469080, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1182/blood-2012-11-469080 instead.
  90. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1038/nature12586, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1038/nature12586 instead.
  91. ^ Naik, Gautam (2013-09-11). "New Promise for Stem Cells - WSJ.com". Online.wsj.com. Retrieved 2014-01-30.
  92. ^ De Los Angeles, A., & Daley, G. Q. (2013) A chemical logic for reprogramming to pluripotency doi:10.1038/cr.2013.119
  93. ^ Federation, A. J., Bradner, J. E., & Meissner, A. (2013) The use of small molecules in somatic-cell reprogramming. Trends in cell biology. doi:10.1016/j.tcb.2013.09.011
  94. ^ NAKAUCHI Hiromitsu, KAMIYA Akihide, SUZUKI Nao, ITO Keiichi, YAMAZAKI Satoshi (2011) METHOD FOR PRODUCING CELLS INDUCED TO DIFFERENTIATE FROM PLURIPOTENT STEM CELLS PATENT COOPERATION TREATY APPLICATION, patno: WO2011071085 (A1) ― 2011-06-16 (C12N5/07)
  95. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1182/blood-2012-06-434407, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1182/blood-2012-06-434407 instead.
  96. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1038/mt.2013.71, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1038/mt.2013.71 instead.
  97. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1038/mt.2013.129, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1038/mt.2013.129 instead.
  98. ^ Attention: This template ({{cite pmid}}) is deprecated. To cite the publication identified by PMID 23293079, please use {{cite journal}} with |pmid=23293079 instead.
  99. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1016/j.stem.2013.11.014, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1016/j.stem.2013.11.014 instead.
  100. ^ Template:Cite 10.1016/j.neulet.2009.04.035
  101. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1002/stem.189, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1002/stem.189 instead.
  102. ^ Jin Yang, Eva Nong, Stephen H Tsang (2013) Induced pluripotent stem cells and retinal degeneration treatment. Expert Rev. Ophthalmol. 8(1), 5–8 doi: 10.1586/EOP.12.75
  103. ^ Fields,, Mark A.; Hwang,, John; Gong,, Jie; Cai,, Hui; Del Priore, Lucian (9 December 2012). Stephen Tsang (ed.). The Eye as a Target Organ for Stem Cell Therapy. Springer. pp. 1–30. ISBN 978-1-4614-5493-9. {{cite book}}: |work= ignored (help); Unknown parameter |Chapter= ignored (|chapter= suggested) (help)CS1 maint: extra punctuation (link)
  104. ^ Li Y, Tsai YT, Hsu CW et al. (2012) Long-term safety and efficacy of human induced pluripotent stem cell (iPS) grafts in a preclinical model of retinitis pigmentosa Mol. Med. 18(1), 1312–1319 doi: 10.2119/molmed.2012.00242
  105. ^ Stem cell therapy for RP is now offered at St. Luke’s Medical Center.
  106. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1159/000346525, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1159/000346525 instead.
  107. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1111/resp.12102<, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1111/resp.12102< instead.
  108. ^ Wong, A. P., & Rossant, J. (2013) Generation of Lung Epithelium from Pluripotent Stem Cells. Current pathobiology reports, 1(2), 137-145, DOI: 10.1007/s40139-013-0016-9
  109. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1016/j.stem.2012.01.018, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1016/j.stem.2012.01.018 instead.
  110. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1172/JCI68793, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1172/JCI68793 instead.
  111. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1016/j.biomaterials.2013.10.018, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1016/j.biomaterials.2013.10.018 instead.
  112. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1038/nbt.2754, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1038/nbt.2754 instead.
  113. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1002/cbf.2924, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1002/cbf.2924 instead.
  114. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1111/j.1365-2184.2012.00811.x, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1111/j.1365-2184.2012.00811.x instead.
  115. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:{{{1}}}, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi={{{1}}} instead.
  116. ^ Antibody that Transforms Bone Marrow Stem Cells Directly into Brain Cells
  117. ^ Jia Xie, Hongkai Zhang, Kyungmoo Yea, and Richard A. Lerner (2013) Autocrine signaling based selection of combinatorial antibodies that transdifferentiate human stem cells PNAS; doi:10.1073/pnas.1306263110
  118. ^ a b c d Xuefeng Liu, Virginie Ory, Sandra Chapman, et al. & Richard Schlegel ( 2012) ROCK Inhibitor and Feeder Cells Induce the Conditional Reprogramming of Epithelial Cells. The American Journal of Pathology, 180(2), 599-607 http://dx.doi.org/10.1016/j.ajpath.2011.10.036 Cite error: The named reference "Xuefeng" was defined multiple times with different content (see the help page).
  119. ^ Rheinwald JG, Green H. (1975) Serial cultivation of strains of human epidermal keratinocytes: the formation of keratinizing colonies from single cells. Cell. 1975 Nov;6(3):331-43
  120. ^ Hiew, Y.-L. (2011) Examining the biological consequences of DNA damage caused by irradiated J2-3T3 fibroblast feeder cells and HPV16: characterisation of the biological functions of Mll. Doctoral thesis, UCL (University College London)
  121. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.3109/09553002.2012.699698, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.3109/09553002.2012.699698 instead.
  122. ^ Hiroshi Kurosawa (2012) Application of Rho-associated protein kinase (ROCK) inhibitor to human pluripotent stem cells. Journal of Bioscience and Bioengineering, 114(6), 577–581 http://dx.doi.org/10.1016/j.jbiosc.2012.07.013
  123. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1089/ten.tea.2009.0339, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1089/ten.tea.2009.0339 instead.
  124. ^ Sandra Chapman, Xuefeng Liu, Craig Meyers, Richard Schlegel, and Alison A. McBride. ( 2010) Human keratinocytes are efficiently immortalized by a Rho kinase inhibitor http://www.jci.org/articles/view/42297
  125. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1073/pnas.1213241109, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1073/pnas.1213241109 instead.
  126. ^ a b Seema Agarwal, David L. Rimm (2012) Making Every Cell Like HeLa: A Giant Step For Cell Culture. The American Journal of Pathology, 180( 2), 443-445 http://dx.doi.org/10.1016/j.ajpath.2011.12.001
  127. ^ a b Lisanti MP, Tanowitz HB. (2012) Translational discoveries, personalized medicine, and living biobanks of the future. The American Journal of Pathology, 2012 Apr;180(4):1334-6 http://www.sciencedirect.com/science/article/pii/S0002944012000971
  128. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1056/NEJMoa1203055, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1056/NEJMoa1203055 instead.
  129. ^ a b Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1016/j.ajpath.2013.08.009, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1016/j.ajpath.2013.08.009 instead.
  130. ^ Sukhbir Kaur, David R. Soto-Pantoja, Erica V. Stein et al. & David D. Roberts.( 2013) Thrombospondin-1 Signaling through CD47 Inhibits Self-renewal by Regulating c-Myc and Other Stem Cell Transcription Factors. Scientific Reports; 3, Article number: 1673 DOI:10.1038/srep01673
  131. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1038/srep01038, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1038/srep01038 instead.
  132. ^ a b Leo Kurian, Ignacio Sancho-Martinez, Emmanuel Nivet, et al. & Juan Carlos Izpisua Belmonte (2012) Conversion of human fibroblasts to angioblast-like progenitor cells. Nature Methods. doi:10.1038/nmeth.2255
  133. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1038/cr.2011.148, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1038/cr.2011.148 instead.
  134. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1074/mcp.M112.020586, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1074/mcp.M112.020586 instead.
  135. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1074/jbc.M202416200, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1074/jbc.M202416200 instead.
  136. ^ ZABecker-Kojič, J . Ureña-Peralta, R.Saffrich et al. & M.Stojkovič ( 2013 ) A new glycoprotein ACA - the main regulator of human hematopoiesis . CELL TECHNOLOGIES IN BIOLOGY AND MEDICINE , 9 (2 ) , 69-84 (rus.)
  137. ^ ZABecker-Kojič, JRUreña-Peralta, I.Zipančić, et al. & M.Stojkovič ( 2013 ) Activation of surface glycoprotein ACA induced pluripotent hematopoietic progenitor cells. CELL TECHNOLOGIES IN BIOLOGY AND MEDICINE , 9 (2 ) , 85-101 (rus.)
  138. ^ Mikkola, M. ( 2013 ) Human pluripotent stem cells: glycomic approaches for culturing and characterization. http://urn.fi/URN:ISBN 978-952-10-8444-7
  139. ^ Obokata, Haruko (30 January 2014). "Stimulus-triggered fate conversion of somatic cells into pluripotency". Nature. 505: 641–647. doi:10.1038/nature12968. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  140. ^ Obokata, Haruko (30 January 2014). "Bidirectional developmental potential in reprogrammed cells with acquired pluripotency". Nature. 505: 676–680. doi:10.1038/nature12969. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  141. ^ Cyranoski, David (30 January 2014). "Acid bath offers easy path to stem cells". Nature. 505. doi:10.1038/505596a.
  142. ^ Stress turns ordinary cells pluripotent. RIKEN news
  143. ^ Wakao, S., Kitada, M. and Dezawa, M. (2013),The elite and stochastic model for iPS cell generation: Multilineage-differentiating stress enduring (Muse) cells are readily reprogrammable into iPS cells. Cytometry, 83A: 18–26. doi: 10.1002/cyto.a.22069
  144. ^ Wakao S,Kitada M,Kuroda Y,Shigemoto T,Matsuse D, et al. Multilineage-differentiating stress-enduring (Muse) cells are a primary source of induced pluripotent stem cells in human fibroblasts. Proc Natl Acad Sci USA 2011; 108: 9875–9880
  145. ^ Stress gives cells a ‘second childhood’. RIKEN RESEARCH,10 February 2014
  146. ^ "STAP NEW DATA | Knoepfler Lab Stem Cell Blog". Ipscell.com. Retrieved 2014-02-19.
  147. ^ jobs. "Acid-bath stem-cell study under investigation : Nature News & Comment". Nature.com. Retrieved 2014-02-19.
  148. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1038/embor.2011.88, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1038/embor.2011.88 instead.
  149. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1016/j.scr.2013.08.009, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1016/j.scr.2013.08.009 instead.
  150. ^ Guannan Su, Yannan Zhao, Jianshu Wei, et al. & Jianwu Dai (2013) Direct conversion of fibroblasts into neural progenitor-like cells by forced growth into 3D spheres on low attachment surfaces. Biomaterials, 34(24), 5897–5906 http://dx.doi.org/10.1016/j.biomaterials.2013.04.040
  151. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1038/nmat3777, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1038/nmat3777 instead.
  152. ^ Patricia Murray, Marina Prewitz, Isabel Hopp, et al. (2013) The self-renewal of mouse embryonic stem cells is regulated by cell–substratum adhesion and cell spreading. The International Journal of Biochemistry & Cell Biology, 45(11), 2698–2705 http://dx.doi.org/10.1016/j.biocel.2013.07.001
  153. ^ Guilak, F., Cohen, D. M., Estes, B. T., et al. & Chen, C. S. (2009) Control of stem cell fate by physical interactions with the extracellular matrix. Cell stem cell, 5(1), 17-26. doi: 10.1016/j.stem.2009.06.016
  154. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1007/s12668-012-0067-0, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1007/s12668-012-0067-0 instead.
  155. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1007/s13238-012-2029-2, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1007/s13238-012-2029-2 instead.
  156. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1016/j.stem.2011.11.015, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1016/j.stem.2011.11.015 instead.
  157. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1038/cr.2012.32, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1038/cr.2012.32 instead.
  158. ^ a b Maucksch, C., E. Firmin, et al. (2012). "Non-viral generation of neural precursor-like cells from adult human fibroblasts." J Stem Cells Regen Med 8(3): 1-9.
  159. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1016/j.stem.2012.05.018, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1016/j.stem.2012.05.018 instead.
  160. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1038/cr.2012.73, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1038/cr.2012.73 instead.
  161. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1073/pnas.1303829110, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1073/pnas.1303829110 instead.
  162. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1038/nbt.2561, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1038/nbt.2561 instead.
  163. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1038/nbt.2564, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1038/nbt.2564 instead.
  164. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1016/j.molmed.2012.06.009, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1016/j.molmed.2012.06.009 instead.
  165. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1016/j.stemcr.2013.07.005, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1016/j.stemcr.2013.07.005 instead.
  166. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1161/CIRCRESAHA.112.270264, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1161/CIRCRESAHA.112.270264 instead.
  167. ^ Paul W. Burridge, Gordon Keller, Joseph D. Gold, Joseph C. Wu (2012) Production of De Novo Cardiomyocytes: Human Pluripotent Stem Cell Differentiation and Direct Reprogramming. Review Article. Cell Stem Cell, 10 (1), 16-28
  168. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1089/scd.2011.0075, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1089/scd.2011.0075 instead.
  169. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1113/jphysiol.2013.252288, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1113/jphysiol.2013.252288 instead.
  170. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1073/pnas.1200250109, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1073/pnas.1200250109 instead.
  171. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1016/j.stem.2012.04.025, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1016/j.stem.2012.04.025 instead.
  172. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1038/ncomms3307, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1038/ncomms3307 instead.
  173. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1681/ASN.2012121143, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1681/ASN.2012121143 instead.
  174. ^ Xinaris C, Benedetti V, Rizzo P, et al. and Giuseppe Remuzzi (2012) In vivo maturation of functional renal organoids formed from embryonic cell suspensions. J Am Soc Nephrol 23: 1857–1868, doi: 10.1681/ASN.2012050505
  175. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1161/CIRCRESAHA.111.250126, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1161/CIRCRESAHA.111.250126 instead.
  176. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1161/CIRCRESAHA.112.265207, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1161/CIRCRESAHA.112.265207 instead.
  177. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1016/j.jacc.2012.04.047, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1016/j.jacc.2012.04.047 instead.
  178. ^ American Heart Association (2012, July 25). Adult stem cells from liposuction used to create blood vessels in the lab. ScienceDaily.
  179. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1038/nbt1287, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1038/nbt1287 instead.
  180. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1073/pnas.1310675110, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1073/pnas.1310675110 instead.
  181. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1038/nbt.2682, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1038/nbt.2682 instead.
  182. ^ Carlos-Filipe Pereira, Betty Chang, Jiajing Qiu et al & Kateri Moore (2013) Induction of a Hemogenic Program in Mouse Fibroblasts. Cell Stem Cell, http://dx.doi.org/10.1016/j.stem.2013.05.024
  183. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1002/stem.1136, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1002/stem.1136 instead.
  184. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1016/j.stemcr.2013.10.010, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1016/j.stemcr.2013.10.010 instead.
  185. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi: 10.1182/blood-2011-06-362038, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi= 10.1182/blood-2011-06-362038 instead.
  186. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi: 10.3324/haematol.2011.055566, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi= 10.3324/haematol.2011.055566 instead.
  187. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi: 10.4061/2011/139851, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi= 10.4061/2011/139851 instead.
  188. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.5966/sctm.2012-0059, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.5966/sctm.2012-0059 instead.
  189. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1182/blood-2012-11-466722, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1182/blood-2012-11-466722 instead.
  190. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1111/j.1537-2995.2010.02644.x, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1111/j.1537-2995.2010.02644.x instead.
  191. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1016/j.stem.2014.01.011, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1016/j.stem.2014.01.011 instead.
  192. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1146/annurev.iy.13.040195.002553, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1146/annurev.iy.13.040195.002553 instead.
  193. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1016/j.stem.2012.11.002, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1016/j.stem.2012.11.002 instead.
  194. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1016/j.stem.2012.12.006, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1016/j.stem.2012.12.006 instead.
  195. ^ a b c Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.3791/3986, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.3791/3986 instead. Cite error: The named reference "Crompton" was defined multiple times with different content (see the help page).
  196. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1158/2159-8290.CD-12-054, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1158/2159-8290.CD-12-054 instead.
  197. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1038/nbt.2678, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1038/nbt.2678 instead.
  198. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1155/2012/720803, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1155/2012/720803 instead.
  199. ^ a b Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1007/s12185-012-1091-0, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1007/s12185-012-1091-0 instead. Cite error: The named reference "Koseki" was defined multiple times with different content (see the help page).
  200. ^ a b Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1038/gt.2012.59, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1038/gt.2012.59 instead.
  201. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1016/j.stem.2009.10.004, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1016/j.stem.2009.10.004 instead.
  202. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1016/j.cell.2008.03.028, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1016/j.cell.2008.03.028 instead.
  203. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1038/nature12885, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1038/nature12885 instead.
  204. ^ Rapino et al., (2013) C/EBPaInduces Highly Efficient Macrophage Transdifferentiation of B Lymphoma and Leukemia Cell Lines and Impairs Their Tumorigenicity, Cell Reports, http://dx.doi.org/10.1016/j.celrep.2013.03.003
  205. ^ a b c Template:CITE DOI
  206. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.5966/sctm.2013-0014, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.5966/sctm.2013-0014 instead.
  207. ^ a b Stolzing A, Jones E, McGonagle D et al. (2008) Age-related changes in human bone marrow derived mesenchymal stem cells: Consequences for cell therapies. Mech Ageing Dev;129:163–173
  208. ^ a b Irina Eberle, Mohsen Moslem, Reinhard Henschler, Tobias Cantz (2012) Engineered MSCs from Patient-Specific iPS Cells. Advances in Biochemical Engineering Biotechnology
  209. ^ a b Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.5966/sctm.2011-0022, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.5966/sctm.2011-0022 instead.
  210. ^ Ruenn Chai Lai, Ronne Wee Yeh Yeo, Soon Sim Tan, Bin Zhang, et al. and Sai Kiang Lim (2013) Mesenchymal Stem Cell Exosomes: The Future MSC-Based Therapy? In: Mesenchymal Stem Cell Therapy. Chase, Lucas G.; Vemuri, Mohan C. (Eds.). 39-61 DOI 10.1007/978-1-62703-200-1_3
  211. ^ Ruenn Chai Lai, Ronne Wee Yeh Yeo, Kok Hian Tan, Sai Kiang Lim (2013) Exosomes for drug delivery — a novel application for the mesenchymal stem cell. Biotechnology Advances.http://dx.doi.org/10.1016/j.biotechadv.2012.08.008
  212. ^ Ronne Wee Yeh Yeoa, Ruenn Chai Laia, Bin Zhanga, et al. & Sai Kiang Lim (2012)Mesenchymal stem cell: An efficient mass producer of exosomes for drug delivery. Advanced Drug Delivery Reviewshttp://dx.doi.org/10.1016/j.addr.2012.07.001
  213. ^ Nobuyoshi Kosaka, Fumitaka Takeshita, Yusuke Yoshioka, et al. & Takahiro Ochiya (2012) Exosomal tumor-suppressive microRNAs as novel cancer therapy: «Exocure» is another choice for cancer treatment. Advanced Drug Delivery Reviewshttp://dx.doi.org/10.1016/j.addr.2012.07.011
  214. ^ a b c Bieback K, Kern S, Kocaomer A et al. (2008) Comparing mesenchymal stromal cells from different human tissues: Bone marrow, adipose tissue and umbilical cord blood. Biomed Mater Eng; 18:S71–S76
  215. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.5966/sctm.2013-0107, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.5966/sctm.2013-0107 instead.
  216. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1002/stem.1067, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1002/stem.1067 instead.
  217. ^ a b Shen JF, Sugawara A, Yamashita J, Ogura H, Sato S. (2011) Dedifferentiated fat cells: an alternative source of adult multipotent cells from the adipose tissues. Int J Oral Sci.;3(3):117-24
  218. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.5966/sctm.2012-0184, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.5966/sctm.2012-0184 instead.
  219. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1089/ten.teb.2012.0757, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1089/ten.teb.2012.0757 instead.
  220. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1371/journal.pone.0077365, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1371/journal.pone.0077365 instead.
  221. ^ Lei F, Haque R, Xiong X, Song J. (2012) Directed differentiation of induced pluripotent stem cells towards T lymphocytes. J Vis Exp. ;(63):e3986. >Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.3791/3986, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.3791/3986 instead.
  222. ^ Sadelain, M., Brentjens, R. & Riviere, I. (2013). The basic principles of chimeric antigen receptor design. Cancer Discov. 3, 388–398 >Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1158/2159-8290.CD-12-054, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1158/2159-8290.CD-12-054 instead.
  223. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1038/nbt.2678, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1038/nbt.2678 instead.
  224. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1155/2012/720803, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1155/2012/720803 instead.
  225. ^ Xie, H., Ye, M., Feng, R. & Graf, T (2004) Stepwise reprogramming of B cells into macrophages. Cell 117(5), 663–676 >Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1016/S0092-8674(04)00419-2, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1016/S0092-8674(04)00419-2 instead.
  226. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1016/j.stem.2009.10.004, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1016/j.stem.2009.10.004 instead.ref>Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1016/j.cell.2008.03.028, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1016/j.cell.2008.03.028 instead.
  227. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1038/nature12885, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1038/nature12885 instead.
  228. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1016/j.celrep.2013.03.003, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1016/j.celrep.2013.03.003 instead.
  229. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.5966/sctm.2013-0014, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.5966/sctm.2013-0014 instead.
  230. ^ Ruenn Chai Lai, Ronne Wee Yeh Yeo, Soon Sim Tan, Bin Zhang, et al. and Sai Kiang Lim (2013) Mesenchymal Stem Cell Exosomes: The Future MSC-Based Therapy? In: Mesenchymal Stem Cell Therapy. Chase, Lucas G.; Vemuri, Mohan C. (Eds.). 39-61 DOI 10.1007/978-1-62703-200-1_3
  231. ^ Ruenn Chai Lai, Ronne Wee Yeh Yeo, Kok Hian Tan, Sai Kiang Lim (2013) Exosomes for drug delivery — a novel application for the mesenchymal stem cell. Biotechnology Advances.http://dx.doi.org/10.1016/j.biotechadv.2012.08.008
  232. ^ Ronne Wee Yeh Yeoa, Ruenn Chai Laia, Bin Zhanga, et al. & Sai Kiang Lim (2012)Mesenchymal stem cell: An efficient mass producer of exosomes for drug delivery. Advanced Drug Delivery Reviewshttp://dx.doi.org/10.1016/j.addr.2012.07.001
  233. ^ Nobuyoshi Kosaka, Fumitaka Takeshita, Yusuke Yoshioka, et al. & Takahiro Ochiya (2012) Exosomal tumor-suppressive microRNAs as novel cancer therapy: «Exocure» is another choice for cancer treatment. Advanced Drug Delivery Reviewshttp://dx.doi.org/10.1016/j.addr.2012.07.011
  234. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.5966/sctm.2012-0184, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.5966/sctm.2012-0184 instead.
  235. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1089/ten.teb.2012.0757, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1089/ten.teb.2012.0757 instead.
  236. ^ Attention: This template ({{cite doi}}) is deprecated. To cite the publication identified by doi:10.1371/journal.pone.0077365, please use {{cite journal}} (if it was published in a bona fide academic journal, otherwise {{cite report}} with |doi=10.1371/journal.pone.0077365 instead.
  237. ^ Keisuke Okita, Tatsuya Yamakawa, Yasuko Matsumura, et al. & Shinya Yamanaka (2013) An Efficient Non-viral Method to Generate Integration-Free Human iPS Cells from Cord Blood and Peripheral Blood Cells. STEM CELLS, DOI: 10.1002/stem.1293
  238. ^ Imbisaat Geti, Mark L. Ormiston, Foad Rouhani, et al & Nicholas W. Morrell (2012) A Practical and Efficient Cellular Substrate for the Generation of Induced Pluripotent Stem Cells from Adults: Blood-Derived Endothelial Progenitor Cells. Stem Cells Trans Med. sctm.2012-0093 doi:10.5966/sctm.2012-0093
  239. ^ Judith Staerk, Meelad M. Dawlaty, Qing Gaoet al. and Rudolf Jaenisch (2010) Reprogramming of Human Peripheral Blood Cells to Induced Pluripotent Stem Cells. Cell Stem Cell, 7(1), 20-24 doi:10.1016/j.stem.2010.06.002
  240. ^ Park TS, Huo JS, Peters A, Talbot CC Jr, Verma K, et al. (2012) Growth Factor-Activated Stem Cell Circuits and Stromal Signals Cooperatively Accelerate Non-Integrated iPSC Reprogramming of Human Myeloid Progenitors. PLoS ONE 7(8): e42838. doi:10.1371/journal.pone.0042838
  241. ^ Katsuhiro Yoshikawa, Motoko Naitoh, Hiroshi Kubota, et al. (2013) Multipotent stem cells are effectively collected from adult human cheek skin. Biochemical and Biophysical Research Communications, 431(1), 104–110 http://dx.doi.org/10.1016/j.bbrc.2012.12.069
  242. ^ Zhou T, Benda C, Duzinger S, Et al & Esteban MA(2011) Generation of induced pluripotent stem cells from urine. J Am Soc Nephrol 22: 1221—1228
  243. ^ Ting Zhou, Christina Benda, Sarah Dunzinger, et al. & Miguel A Esteban (2012) Generation of human induced pluripotent stem cells from urine samples. Nature Protocols. 7(12), 2080–2089 doi:10.1038/nprot.2012.115
  244. ^ Lihui Wang, Linli Wang, Wenhao Huang, & Duanqing Pei (2012) Generation of integration-free neural progenitor cells from cells in human urine. Nature Methods, doi:10.1038/nmeth.2283
  245. ^ . Cai J, Zhang Y, Liu P, Chen S, Wu X, Sun Y, Li A, Huang K et al (2013)Generation of tooth-like structures from integration-free human urine induced pluripotent stem cells. Cell Regeneration , 2:6 doi:10.1186/2045-9769-2-6
  246. ^ Shantaram Bharadwaj, Guihua Liu, Yingai Shi, et al. & Yuanyuan Zhang (2013) Multi-Potential Differentiation of Human Urine-Derived Stem Cells: Potential for Therapeutic Applications in Urology. STEM CELLS, DOI: 10.1002/stem.1424
  247. ^ Yimei Wang1, Jinyu Liu1, Xiaohua Tan1, et al. and Yulin Li (2012) Induced Pluripotent Stem Cells from Human Hair Follicle Mesenchymal Stem Cells. Stem Cell Reviews and Reports, doi:10.1007/s12015-012-9420-5
  248. ^ Schnabel L. V, Abratte C. M., Schimenti J. C, et al. and Fortier L. A. (2012) Genetic background affects induced pluripotent stem cell generation. Stem Cell Research & Therapy 2012, 3:30 doi:10.1186/scrt121
  249. ^ Panopoulos AD, Ruiz S, Yi F, Herrerías A, Batchelder EM, Izpisua Belmonte JC.(2011) Rapid and highly efficient generation of induced pluripotent stem cells from human umbilical vein endothelial cells. PLoS One;6:e19743
  250. ^ a b J.M. Polo, S. Liu, M.E. Figueroa, et al. & Konrad Hochedlinger (2010) Cell type of origin influences the molecular and functional properties of mouse induced pluripotent stem cells. Nat Biotechnol, 28, 848–855 doi:10.1038/nbt.1667
  251. ^ Miura K, Okada Y, Aoi T, Okada A, et al & Yamanaka S.(2009) Variation in the safety of induced pluripotent stem cell lines. Nat Biotechnol.;27:743-745
  252. ^ Liang Y, Zhang H, Feng QS et al. (2012) The Propensity for Tumorigenesis in Human Induced Pluripotent Stem Cells is Correlated with Genomic Instability. Chin J Cancer. doi: 10.5732/cjc.012.10065. [Epub ahead of print]
  253. ^ K. Kim, A. Doi, B. Wen, K. Ng, R. Zhao, P. Cahan, J. Kim, M.J. Aryee, H. Ji, L.I. Ehrlich et al. (2010) Epigenetic memory in induced pluripotent stem cells. Nature, 467, 285–290 doi:10.1038/nature09342
  254. ^ K. Kim, R. Zhao, A. Doi,et al. (2011) Donor cell type can influence the epigenome and differentiation potential of human induced pluripotent stem cells. Nat Biotechnol, 29, pp. 1117–1119
  255. ^ O. Bar-Nur, H.A. Russ, S. Efrat, N. Benvenisty (2011) Epigenetic memory and preferential lineage-specific differentiation in induced pluripotent stem cells derived from human pancreatic islet beta cells. Cell Stem Cell, 9 , 17–23 http://dx.doi.org/10.1016/j.stem.2011.06.007
  256. ^ Denker H-W.( 2012) Time to Reconsider Stem Cell Induction Strategies. Cells.; 1(4):1293-1312. doi:10.3390/cells1041293

Notes