User:Juliedobkin/sandbox

From Wikipedia, the free encyclopedia

All original content copied from Embryonic stem cell.

Direction with editing this article: The original article for Embryonic stem cell needs work on organization, general verification of information, and updates to outdated statements and claims. So far I have been working on stripping away a lot of what I believe to be "fillers" to bulk up sections, and fixing the organization of information, as well as updating outdated information and adding necessary references. I plan to continue to re-organize the article to make it easier to follow and for the flow of information to be more logical. I also plan to add far more information regarding ESC uses and published studies, as well as a small section discussing the controversy (though this shouldn't be too in-depth, as there is an entirely separate article for this topic).

Embryonic stem cell edits[edit]

Embryonic stem cells (ES cells) are pluripotent stem cells derived from the inner cell mass of a blastocyst, an early-stage pre-implantation embryo.[1][2] Human embryos reach the blastocyst stage 4–5 days post fertilization, at which time they consist of 50–150 cells. Isolating the embryoblast, or inner cell mass (ICM) results in destruction of the blastocyst, a process which raises ethical issues, including whether or not embryos at the pre-implantation stage should be have the same moral considerations as embryos in the post-implantation stage of development.[3][4] Researchers are currently focusing heavily on the therapeutic potential of embryonic stem cells, with clinical use being the goal for many labs. These cells are being studied to be used as clinical therapies, models of genetic disorders, and cellular/DNA repair. However, adverse effects in the research and clinical processes have also been reported.

This summary of ESCs was shortened, with information that was not pertinent removed. Minor edits revolving around sentence structure and grammar were also made.

Properties[edit]

The transcriptome of embryonic stem cells

Embryonic stem cells (ESCs), derived from the blastocyst stage of early mammalian embryos, are distinguished by their ability to differentiate into any cell type and by their ability to propagate. It is these traits that makes them valuable in the scientific/medical fields. ESC are also described as having a normal karyotype, maintaining high telomerase activity, and exhibiting remarkable long-term proliferative potential.[5]

Grammar/organization edits made, along with removal of unnecessary information removed. Links to relevant wiki pages added.

Pluripotent[edit]

Embryonic stem cells of the inner cell mass are pluripotent, meaning they are able to differentiate to generate primitive ectoderm, which ultimately differentiates during gastrulation into all derivatives of the three primary germ layers: ectoderm, endoderm, and mesoderm. These include each of the more than 220 cell types in the adult human body. Pluripotency distinguishes embryonic stem cells from adult stem cells, which are multipotent and can only produce a limited number of cell types.

Grammatical/structural edits made and links added. Confusing/repetitive language removed.

In 2012, the Nobel Prize for Medicine was attributed conjointed to John B. Gurdon and Shinya Yamanaka for the discovery that mature cells can be reprogrammed to become pluripotent.[6]


Propagation[edit]

Under defined conditions, embryonic stem cells are capable of propagating indefinitely in an undifferentiated state. Conditions must either prevent the cells from clumping, or maintain an environment that supports an unspecialized state. [7] While being able to remain undifferentiated, ESCs also have the capacity, when provided with the appropriate signals, to differentiate (presumably via the initial formation of precursor cells) into nearly all mature cell phenotypes.[8]

Information updated/added and relevant sources referenced. Incorrect/improperly sourced information removed

Uses[edit]

Due to their plasticity and potentially unlimited capacity for self-renewal, embryonic stem cell therapies have been proposed for regenerative medicine and tissue replacement after injury or disease. Pluripotent stem cells have shown potential in treating a number of varying conditions, including but not limited to: spinal cord injuries, age related macular degeneration, diabetes, neurodegenerative disorders (such as Parkinson's disease), AIDS, etc [9]. In addition to their potential in regenerative medicine, embryonic stem cells provide an alternative source of tissue/organs which serves as a possible solution to the donor shortage dilemma. Not only that, but tissue/organs derived from ESCs can be made immunocompatible with the recipient. Aside from these uses, embryonic stem cells can also serve as tools for the investigation of early human development, study of genetic disease and as in vitro systems for toxicology testing.[5]

Entire section re-write. Information that was not pertinent to the section was removed and relevant facts/sources added. Grammar/organization edits also made.

Usefulness (prior to rewrite)[edit]

Because of their plasticity and potentially unlimited capacity for self-renewal, embryonic stem cell therapies have been proposed for regenerative medicine and tissue replacement after injury or disease. Diseases that could potentially be treated by pluripotent stem cells include a number of blood and immune-system related genetic diseases, cancers, and disorders; juvenile diabetes; Parkinson's disease; blindness and spinal cord injuries. Besides the ethical concerns of stem cell therapy (see stem cell controversy), there is a technical problem of graft-versus-host disease associated with allogeneic stem cell transplantation. However, these problems associated with histocompatibility may be solved using autologous donor adult stem cells, therapeutic cloning. Stem cell banks or more recently by reprogramming of somatic cells with defined factors (e.g. induced pluripotent stem cells). Embryonic stem cells provide hope that it will be possible to overcome the problems of donor tissue shortage and also, by making the cells immunocompatible with the recipient. Other potential uses of embryonic stem cells include investigation of early human development, study of genetic disease and as in vitro systems for toxicology testing.[5]

Utilizations[edit]

According to a 2002 article in PNAS, "Human embryonic stem cells have the potential to differentiate into various cell types, and, thus, may be useful as a source of cells for transplantation or tissue engineering."[10]

Embryoid bodies 24 hours after formation.

However, embryonic stem cells are not limited to cell/tissue engineering.

Cell Replacement Therapies[edit]

Current research focuses on differentiating ESCs into a variety of cell types for eventual use as cell replacement therapies (CRTs). Some of the cell types that have or are currently being developed include cardiomyocytes (CM), neurons, hepatocytes, bone marrow cells, islet cells and endothelial cells.[11] However, the derivation of such cell types from ESCs is not without obstacles, therefore current research is focused on overcoming these barriers. For example, studies are underway to differentiate ESCs in to tissue specific CMs and to eradicate their immature properties that distinguish them from adult CMs.[12]

Clinical Potential[edit]

Researchers have differentiated ESCs into dopamine-producing cells with the hope that these neurons could be used in the treatment of Parkinson’s disease.[13][14] ESCs have also been differentiated to natural killer (NK) cells and bone tissue.[15] Studies involving ESCs are also underway to provide an alternative treatment for diabetes. For example, D’Amour et al. were able to differentiate ESCs into insulin producing cells[16] and researchers at Harvard University were able to produce large quantities of pancreatic beta cells from ES.[17]

An article published in the European Heart Journal describes a translational process of generating human embryonic stem cell-derived cardiac progenitor cells to be used in clinical trials of patients with severe heart failure. [18]

Drug Discovery[edit]

Besides becoming an important alternative to organ transplants, ESCs are also being used in field of toxicology and as cellular screens to uncover new chemical entities (NCEs) that can be developed as small molecule drugs. Studies have shown that cardiomyocytes derived from ESCs are validated in vitro models to test drug responses and predict toxicity profiles.[11] ES derived cardiomyocytes have been shown to respond to pharmacological stimuli and hence can be used to assess cardiotoxicity like Torsades de Pointes.[19]

ESC-derived hepatocytes are also useful models that could be used in the preclinical stages of drug discovery. However, the development of hepatocytes from ESCs has proven to be challenging and this hinders the ability to test drug metabolism. Therefore, current research is focusing on establishing fully functional ESC-derived hepatocytes with stable phase I and II enzyme activity.[20]

Models of Genetic Disorder[edit]

Several new studies have started to address this issue. This has been done either by genetically manipulating the cells, or more recently by deriving diseased cell lines identified by prenatal genetic diagnosis (PGD). This approach may very well prove invaluable at studying disorders such as Fragile-X syndrome, Cystic fibrosis, and other genetic maladies that have no reliable model system.

Yury Verlinsky, a Russian-American medical researcher who specialized in embryo and cellular genetics (genetic cytology), developed prenatal diagnosis testing methods to determine genetic and chromosomal disorders a month and a half earlier than standard amniocentesis. The techniques are now used by many pregnant women and prospective parents, especially those couples with a history of genetic abnormalities or where the woman is over the age of 35, when the risk of genetically related disorders is higher. In addition, by allowing parents to select an embryo without genetic disorders, they have the potential of saving the lives of siblings that already had similar disorders and diseases using cells from the disease free offspring.[21]

Scientists have discovered a new technique for deriving human embryonic stem cell (ESC). Normal ESC lines from different sources of embryonic material including morula and whole blastocysts have been established. These findings allows researchers to construct ESC lines from embryos that acquire different genetic abnormalities; therefore, allowing for recognition of mechanisms in the molecular level that are possibly blocked that could impede the disease progression. The ESC lines originating from embryos with genetic and chromosomal abnormalities provide the data necessary to understand the pathways of genetic defects.[22]

A donor patient acquires one defective gene copy and one normal, and only one of these two copies is used for reproduction. By selecting egg cell derived from embryonic stem cells that have two normal copies, researchers can find variety of treatments for various diseases. To test this theory Dr. McLaughlin and several of his colleagues looked at whether parthenogenesis תשרקץצףפעסןembryonic stem cells can be used in a mouse model that has thalassemia intermedia. This disease is described as an inherited blood disorder in which there is a lack of hemoglobin leading to anemia. The mouse model used, had one defective gene copy. Embryonic stem cells from an unfertilized egg of the diseased mice were gathered and those stem cells that contained only healthy hemoglobin genes were identified. The healthy embryonic stem cell lines were then converted into cells transplanted into the carrier mice. After five weeks, the test results from the transplant illustrated that these carrier mice now had a normal blood cell count and hemoglobin levels.[23]

Repair of DNA damage[edit]

Differentiated somatic cells and ES cells use different strategies for dealing with DNA damage. For instance, human foreskin fibroblasts, one type of somatic cell, use non-homologous end joining (NHEJ), an error prone DNA repair process, as the primary pathway for repairing double-strand breaks (DSBs) during all cell cycle stages.[24] Because of its error-prone nature, NHEJ tends to produce mutations in a cell’s clonal descendants.

ES cells use a different strategy to deal with DSBs.[25] Because ES cells give rise to all of the cell types of an organism including the cells of the germ line, mutations arising in ES cells due to faulty DNA repair are a more serious problem than in differentiated somatic cells. Consequently, robust mechanisms are needed in ES cells to repair DNA damages accurately, and if repair fails, to remove those cells with un-repaired DNA damages. Thus, mouse ES cells predominantly use high fidelity homologous recombinational repair (HRR) to repair DSBs.[25] This type of repair depends on the interaction of the two sister chromosomes formed during S phase and present together during the G2 phase of the cell cycle. HRR can accurately repair DSBs in one sister chromosome by using intact information from the other sister chromosome. Cells in the G1 phase of the cell cycle (i.e. after metaphase/cell division but prior the next round of replication) have only one copy of each chromosome (i.e. sister chromosomes aren’t present). Mouse ES cells lack a G1 checkpoint and do not undergo cell cycle arrest upon acquiring DNA damage.[26] Rather they undergo programmed cell death (apoptosis) in response to DNA damage.[27] Apoptosis can be used as a fail-safe strategy to remove cells with un-repaired DNA damages in order to avoid mutation and progression to cancer.[28] Consistent with this strategy, mouse ES stem cells have a mutation frequency about 100-fold lower than that of isogenic mouse somatic cells.[29]

Clinical trial[edit]

On January 23, 2009, Phase I clinical trials for transplantation of oligodendrocytes (a cell type of the brain and spinal cord) derived from human ES cells into spinal cord-injured individuals received approval from the U.S. Food and Drug Administration (FDA), marking it the world's first human ES cell human trial.[30] The study leading to this scientific advancement was conducted by Hans Keirstead and colleagues at the University of California, Irvine and supported by Geron Corporation of Menlo Park, CA, founded by Michael D. West, PhD. A previous experiment had shown an improvement in locomotor recovery in spinal cord-injured rats after a 7-day delayed transplantation of human ES cells that had been pushed into an oligodendrocytic lineage.[31] The phase I clinical study was designed to enroll about eight to ten paraplegics who have had their injuries no longer than two weeks before the trial begins, since the cells must be injected before scar tissue is able to form. The researchers emphasized that the injections were not expected to fully cure the patients and restore all mobility. Based on the results of the rodent trials, researchers speculated that restoration of myelin sheathes and an increase in mobility might occur. This first trial was primarily designed to test the safety of these procedures and if everything went well, it was hoped that it would lead to future studies that involve people with more severe disabilities.[32] The trial was put on hold in August 2009 due to FDA concerns regarding a small number of microscopic cysts found in several treated rat models but the hold was lifted on July 30, 2010.[33]

In October 2010 researchers enrolled and administered ESTs to the first patient at Shepherd Center in Atlanta.[34] The makers of the stem cell therapy, Geron Corporation, estimated that it would take several months for the stem cells to replicate and for the GRNOPC1 therapy to be evaluated for success or failure.

In November 2011 Geron announced it was halting the trial and dropping out of stem cell research for financial reasons, but would continue to monitor existing patients, and was attempting to find a partner that could continue their research.[35] In 2013 BioTime (AMEXBTX), led by CEO Dr. Michael D. West, acquired all of Geron's stem cell assets, with the stated intention of restarting Geron's embryonic stem cell-based clinical trial for spinal cord injury research.[36]

BioTime company Asterias Biotherapeutics (NYSE MKT: AST) was granted a $14.3 million Strategic Partnership Award by the California Institute for Regenerative Medicine (CIRM) to re-initiate the world’s first embryonic stem cell-based human clinical trial, for spinal cord injury. Supported by California public funds, CIRM is the largest funder of stem cell-related research and development in the world.[37]

The award provides funding for Asterias to reinitiate clinical development of AST-OPC1 in subjects with spinal cord injury and to expand clinical testing of escalating doses in the target population intended for future pivotal trials.[37]

AST-OPC1 is a population of cells derived from human embryonic stem cells (hESCs) that contains oligodendrocyte progenitor cells (OPCs). OPCs and their mature derivatives called oligodendrocytes provide critical functional support for nerve cells in the spinal cord and brain. Asterias recently presented the results from phase 1 clinical trial testing of a low dose of AST-OPC1 in patients with neurologically-complete thoracic spinal cord injury. The results showed that AST-OPC1 was successfully delivered to the injured spinal cord site. Patients followed 2–3 years after AST-OPC1 administration showed no evidence of serious adverse events associated with the cells in detailed follow-up assessments including frequent neurological exams and MRIs. Immune monitoring of subjects through one year post-transplantation showed no evidence of antibody-based or cellular immune responses to AST-OPC1. In four of the five subjects, serial MRI scans performed throughout the 2–3 year follow-up period indicate that reduced spinal cord cavitation may have occurred and that AST-OPC1 may have had some positive effects in reducing spinal cord tissue deterioration. There was no unexpected neurological degeneration or improvement in the five subjects in the trial as evaluated by the International Standards for Neurological Classification of Spinal Cord Injury (ISNCSCI) exam.[37]

The Strategic Partnership III grant from CIRM will provide funding to Asterias to support the next clinical trial of AST-OPC1 in subjects with spinal cord injury, and for Asterias’ product development efforts to refine and scale manufacturing methods to support later-stage trials and eventually commercialization. CIRM funding will be conditional on FDA approval for the trial, completion of a definitive agreement between Asterias and CIRM, and Asterias’ continued progress toward the achievement of certain pre-defined project milestones.[37]


History[edit]

  • 1964: Lewis Kleinsmith and G. Barry Pierce Jr. isolated a single type of cell from a teratocarcinoma, a tumor now known to be derived from a germ cell.[38] These cells isolated from the teratocarcinoma replicated and grew in cell culture as a stem cell and are now known as embryonal carcinoma (EC) cells.[39] Although similarities in morphology and differentiating potential (pluripotency) led to the use of EC cells as the in vitro model for early mouse development,[40] EC cells harbor genetic mutations and often abnormal karyotypes that accumulated during the development of the teratocarcinoma. These genetic aberrations further emphasized the need to be able to culture pluripotent cells directly from the inner cell mass.
Martin Evans revealed a new technique for culturing the mouse embryos in the uterus to allow for the derivation of ES cells from these embryos.
  • 1981: Embryonic stem cells (ES cells) were independently first derived from mouse embryos by two groups. Martin Evans and Matthew Kaufman from the Department of Genetics, University of Cambridge published first in July, revealing a new technique for culturing the mouse embryos in the uterus to allow for an increase in cell number, allowing for the derivation of ES cells from these embryos.[41] Gail R. Martin, from the Department of Anatomy, University of California, San Francisco, published her paper in December and coined the term “Embryonic Stem Cell”.[42] She showed that embryos could be cultured in vitro and that ES cells could be derived from these embryos. In 1998, a breakthrough occurred when researchers, led by James Thomson at the University of Wisconsin-Madison, first developed a technique to isolate and grow human embryonic stem cells in cell culture.[43]
  • 1989: Mario R. Cappechi, Martin J. Evans, and Oliver Smithies publish their research which details their isolation and genetic modifications of embryonic stem cells, creating the first "knockout mice". [44] In creating knockout mice, this publication provided scientists with an entirely new way to study disease.
  • 1998: A paper titled "Embryonic Stem Cell Lines Derived From Human Blastocysts" is published by a team from the University of Wisconsin, Madison. The researchers behind this study not only create the first embryonic stem cells, but recognize their pluripotency, as well as their capacity for self-renewal. The abstract of the paper notes the significance of the discovery with regards to the fields of developmental biology and drug discovery. [45]
  • 2001: President George W. Bush allows federal funding to support research on roughly 60—at this time, already existing—lines of embryonic stem cells. Seeing as the limited lines that Bush allowed research on had already been established, this law supported embryonic stem cell research without raising any ethical questions that could arise with the creation of new lines under federal budget. [46]
  • 2006: Japanese scientists Shinya Yamanaka and Kazutoshi Takashi publish a paper describing the induction of pluripotent stem cells from cultures of adult mouse fibroblasts. Induced pluripotent stem cells (iPSCs) are a huge discovery, as they are seemingly identical to embryonic stem cells and could be used without sparking the same moral controversy. [47]
  • January, 2009: The US Food and Drug Administration (FDA) provides approval for Geron Corporation's phase I trial of their human embryonic stem cell-derived treatment for spinal cord injuries. The announcement was met with excitement from the scientific community, but also with wariness from stem cell opposers. The treatment cells were, however, derived from the cell lines approved under George W. Bush's ESC policy.[48]
  • March, 2009: Executive Order 13505 is signed by President Barack Obama, removing the restrictions put in place on federal funding for human stem cells by the previous presidential administration. This would allow the National Institutes of Health (NIH) to provide funding for hESC research. The document also states that the NIH must provide revised federal funding guidelines within 120 days of the order's signing. [49]

References[edit]

  1. ^ Thomson; Itskovitz-Eldor, J; Shapiro, SS; Waknitz, MA; Swiergiel, JJ; Marshall, VS; Jones, JM (1998). "Blastocysts Embryonic Stem Cell Lines Derived from Human". Science. 282 (5391): 1145–1147. Bibcode:1998Sci...282.1145T. doi:10.1126/science.282.5391.1145. PMID 9804556.
  2. ^ "NIH Stem Cell Basics. What are embryonic stem cells?".
  3. ^ Baldwing A (2009). "Morality and human embryo research. Introduction to the Talking Point on morality and human embryo research". EMBO Reports. 10 (4): 299–300. doi:10.1038/embor.2009.37. PMC 2672902. PMID 19337297.
  4. ^ Nakaya, Andrea C. (August 1, 2011). Biomedical ethics. San Diego, CA: ReferencePoint Press. p. 96. ISBN 160152157X.
  5. ^ a b c Thomson, J. A.; Itskovitz-Eldor, J; Shapiro, S. S.; Waknitz, M. A.; Swiergiel, J. J.; Marshall, V. S.; Jones, J. M. (1998). "Embryonic Stem Cell Lines Derived from Human Blastocysts". Science. 282 (5391): 1145–7. Bibcode:1998Sci...282.1145T. doi:10.1126/science.282.5391.1145. PMID 9804556.
  6. ^ The Nobel Prize in Physiology or Medicine 2012. nobelprize.org
  7. ^ "NIH Stem Cell Basics. What are embryonic stem cells?".
  8. ^ Ying; Nichols, J; Chambers, I; Smith, A (2003). "BMP Induction of Id Proteins Suppresses Differentiation and Sustains Embryonic Stem Cell Self-Renewal in Collaboration with STAT3". Cell. 115 (3): 281–292. doi:10.1016/S0092-8674(03)00847-X. PMID 14636556.
  9. ^ Mahla, Ranjeet (July 19th, 2016). "Stem Cell Applications in Regenerative Medicine and Disease Therapeutics". International Journal of Cell Biology: 1–24. doi:10.1155/2016/6940283. PMID 27516776. {{cite journal}}: Check date values in: |date= (help)CS1 maint: unflagged free DOI (link)
  10. ^ Levenberg, S. (2002). "Endothelial cells derived from human embryonic stem cells". Proceedings of the National Academy of Sciences. 99 (7): 4391–4396. Bibcode:2002PNAS...99.4391L. doi:10.1073/pnas.032074999. PMC 123658.
  11. ^ a b Davila, JC; Cezar, GG; Thiede, M; Strom, S; Miki, T; Trosko, J (2004). "Use and application of stem cells in toxicology". Toxicological Sciences. 79 (2): 214–23. doi:10.1093/toxsci/kfh100. PMID 15014205.
  12. ^ Siu, CW; Moore, JC; Li, RA (2007). "Human embryonic stem cell-derived cardiomyocytes for heart therapies". Cardiovascular & Hematological Disorders Drug Targets. 7 (2): 145–52. doi:10.2174/187152907780830851. PMID 17584049.
  13. ^ Perrier, A. L. (2004). "Derivation of midbrain dopamine neurons from human embryonic stem cells". Proceedings of the National Academy of Sciences. 101 (34): 12543–12548. Bibcode:2004PNAS..10112543P. doi:10.1073/pnas.0404700101. PMC 515094.
  14. ^ Parish, CL; Arenas, E (2007). "Stem-cell-based strategies for the treatment of Parkinson's disease". Neuro-Degenerative Diseases. 4 (4): 339–47. doi:10.1159/000101892. PMID 17627139.
  15. ^ Waese, EY; Kandel, RA; Stanford, WL (2008). "Application of stem cells in bone repair". Skeletal Radiology. 37 (7): 601–8. doi:10.1007/s00256-007-0438-8. PMID 18193216.
  16. ^ d'Amour, KA; Bang, AG; Eliazer, S; Kelly, OG; Agulnick, AD; Smart, NG; Moorman, MA; Kroon, E; Carpenter, MK; Baetge, EE (2006). "Production of pancreatic hormone-expressing endocrine cells from human embryonic stem cells". Nature Biotechnology. 24 (11): 1392–401. doi:10.1038/nbt1259. PMID 17053790.
  17. ^ Colen, B.D. (9 October 2014) Giant leap against diabetes The Harvard Gazette, Retrieved 24 November 2014
  18. ^ Menasché, Phillip; Vanneaux, Valérie; Fabreguettes, Jean-Roch; Bel, Alain; Tosca, Lucie; Garcia, Sylvie (21 March 2015). "Towards a clinical use of human embryonic stem cell derived-cardiac progenitors: a translational experience". European Heart Journal. 36 (12): 743–750. doi:https://doi.org/10.1093/eurheartj/ehu192. {{cite journal}}: Check |doi= value (help); External link in |doi= (help)
  19. ^ Jensen, J; Hyllner, J; Björquist, P (2009). "Human embryonic stem cell technologies and drug discovery". Journal of Cellular Physiology. 219 (3): 513–9. doi:10.1002/jcp.21732. PMID 19277978.
  20. ^ Söderdahl, T; Küppers-Munther, B; Heins, N; Edsbagge, J; Björquist, P; Cotgreave, I; Jernström, B (2007). "Glutathione transferases in hepatocyte-like cells derived from human embryonic stem cells". Toxicology in Vitro. 21 (5): 929–37. doi:10.1016/j.tiv.2007.01.021. PMID 17346923.
  21. ^ "Dr. Yury Verlinsky, 1943–2009: Expert in reproductive technology" Chicago Tribune, July 20, 2009
  22. ^ Verlinsky, Y; Strelchenko, N; Kukharenko, V; Rechitsky, S; Verlinsky, O; Galat, V; Kuliev, A (2005). "Human embryonic stem cell lines with genetic disorders". Reproductive Biomedicine Online. 10 (1): 105–10. doi:10.1016/S1472-6483(10)60810-3. PMID 15705304.
  23. ^ Embryonic Stem Cells Help Deliver 'Good Genes' In A Model Of Inherited Blood Disorder, ScienceDaily (February 13, 2011).
  24. ^ Mao Z, Bozzella M, Seluanov A, Gorbunova V (September 2008). "DNA repair by nonhomologous end joining and homologous recombination during cell cycle in human cells". Cell Cycle. 7 (18): 2902–6. doi:10.4161/cc.7.18.6679. PMC 2754209. PMID 18769152.
  25. ^ a b Tichy ED, Pillai R, Deng L, et al. (November 2010). "Mouse embryonic stem cells, but not somatic cells, predominantly use homologous recombination to repair double-strand DNA breaks". Stem Cells Dev. 19 (11): 1699–711. doi:10.1089/scd.2010.0058. PMC 3128311. PMID 20446816.
  26. ^ Hong Y, Stambrook PJ (October 2004). "Restoration of an absent G1 arrest and protection from apoptosis in embryonic stem cells after ionizing radiation". Proc. Natl. Acad. Sci. U.S.A. 101 (40): 14443–8. Bibcode:2004PNAS..10114443H. doi:10.1073/pnas.0401346101. PMC 521944. PMID 15452351.
  27. ^ Aladjem MI, Spike BT, Rodewald LW, et al. (January 1998). "ES cells do not activate p53-dependent stress responses and undergo p53-independent apoptosis in response to DNA damage". Curr. Biol. 8 (3): 145–55. doi:10.1016/S0960-9822(98)70061-2. PMID 9443911.
  28. ^ Bernstein C, Bernstein H, Payne CM, Garewal H (June 2002). "DNA repair/pro-apoptotic dual-role proteins in five major DNA repair pathways: fail-safe protection against carcinogenesis". Mutat. Res. 511 (2): 145–78. doi:10.1016/S1383-5742(02)00009-1. PMID 12052432.
  29. ^ Cervantes RB, Stringer JR, Shao C, Tischfield JA, Stambrook PJ (March 2002). "Embryonic stem cells and somatic cells differ in mutation frequency and type". Proc. Natl. Acad. Sci. U.S.A. 99 (6): 3586–90. Bibcode:2002PNAS...99.3586C. doi:10.1073/pnas.062527199. PMC 122567. PMID 11891338.
  30. ^ "FDA approves human embryonic stem cell study - CNN.com". January 23, 2009. Retrieved May 1, 2010.
  31. ^ Keirstead HS, Nistor G, Bernal G, et al. (2005). "Human embryonic stem cell-derived oligodendrocyte progenitor cell transplants remyelinate and restore locomotion after spinal cord injury". J. Neurosci. 25 (19): 4694–705. doi:10.1523/JNEUROSCI.0311-05.2005. PMID 15888645.
  32. ^ Reinberg, Steven (2009-01-23) FDA OKs 1st Embryonic Stem Cell Trial. The Washington Post
  33. ^ Geron comments on FDA hold on spinal cord injury trial. geron.com (August 27, 2009)
  34. ^ Vergano, Dan (11 October 2010). "Embryonic stem cells used on patient for first time". USA Today. Retrieved 12 October 2010.
  35. ^ Brown, Eryn (November 15, 2011). "Geron exits stem cell research". LA Times. Retrieved 2011-11-15.
  36. ^ "Great news: BioTime Subsidiary Asterias Acquires Geron Embryonic Stem Cell Program". iPScell.com. October 1, 2013.
  37. ^ a b c d California Institute of Regenerative Medicine. BioTime, Inc.
  38. ^ Kleinsmith LJ, Pierce GB Jr (1964). "Multipotentiality of Single Embryoncal Carcinoma Cells". Cancer Res. 24: 1544–51. PMID 14234000.
  39. ^ Andrews P, Matin M, Bahrami A, Damjanov I, Gokhale P, Draper J (2005). "Embryonic stem (ES) cells and embryonal carcinoma (EC) cells: opposite sides of the same coin" (PDF). Biochem Soc Trans. 33 (Pt 6): 1526–30. doi:10.1042/BST20051526. PMID 16246161.
  40. ^ Martin GR (1980). "Teratocarcinomas and mammalian embryogenesis". Science. 209 (4458): 768–76. Bibcode:1980Sci...209..768M. doi:10.1126/science.6250214. PMID 6250214.
  41. ^ Evans M, Kaufman M (1981). "Establishment in culture of pluripotent cells from mouse embryos". Nature. 292 (5819): 154–6. Bibcode:1981Natur.292..154E. doi:10.1038/292154a0. PMID 7242681.
  42. ^ Martin G (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 (12): 7634–8. Bibcode:1981PNAS...78.7634M. doi:10.1073/pnas.78.12.7634. PMC 349323. PMID 6950406.
  43. ^ Thomson J, Itskovitz-Eldor J, Shapiro S, Waknitz M, Swiergiel J, Marshall V, Jones J (1998). "Embryonic stem cell lines derived from human blastocysts". Science. 282 (5391): 1145–7. Bibcode:1998Sci...282.1145T. doi:10.1126/science.282.5391.1145. PMID 9804556.
  44. ^ "The 2007 Nobel Prize in Physiology or Medicine - Advanced Information". Nobel Prize. Nobel Media.
  45. ^ Thompson, James A.; Itskovitz-Eldor, Joseph; Shapiro, Sander S.; Waknitz, Michelle A.; Swiergiel, Jennifer J.; Marshall, Vivienne S.; Jones, Jeffrey M. (06 Nov 1998). "Embryonic Stem Cell Lines Derived From Human Blastocyst". Science. 282 (5391): 1145–1147. doi:10.1126/science.282.5391.1145. {{cite journal}}: Check date values in: |date= (help)
  46. ^ "President George W. Bush's address on stem cell research". CNN Inside Politics. CNN. Aug 9th 2001. {{cite news}}: Check date values in: |date= (help)
  47. ^ Yamanaka, Shinya; Takahashi, Kazutoshi (25 Aug 2006). "Induction of Pluripotent Stem Cells From Mouse Embryonic and Adult Fibroblast Cultures by Defined Factors". Cell. 126 (4): 663–676. doi:https://doi.org/10.1016/j.cell.2006.07.024. {{cite journal}}: Check |doi= value (help); External link in |doi= (help)
  48. ^ Wadman, Meredith (27 Jan. 2009). "Stem cells ready for primetime". Nature News. Nature. doi:10.1038/457516a. {{cite web}}: Check date values in: |date= (help)
  49. ^ "Executive Order 13505—Removing Barriers To Responsible Scientific Research Involving Human Stem Cells" (PDF). Federal Register: Presidential Documents. 74 (46). 11 March, 2009. {{cite journal}}: Check date values in: |date= (help)