fbpx
Wikipedia

Embryonic stem cell

Embryonic stem cells (ESCs) 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 inner cell mass (embryoblast) using immunosurgery results in destruction of the blastocyst, a process which raises ethical issues, including whether or not embryos at the pre-implantation stage have the same moral considerations as embryos in the post-implantation stage of development.[3][4]

Human embryonic stem cells in cell culture
Pluripotent: Embryonic stem cells are able to develop into any type of cell, excepting those of the placenta. Only embryonic stem cells of the morula are totipotent: able to develop into any type of cell, including those of the placenta.

Researchers are currently focusing heavily on the therapeutic potential of embryonic stem cells, with clinical use being the goal for many laboratories.[2] Potential uses include the treatment of diabetes and heart disease.[2] The 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 such as tumors and unwanted immune responses have also been reported.[5]

Properties

 
IPS Cell
 
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 embryonic cell type and by their ability to self-renew. It is these traits that makes them valuable in the scientific and medical fields. ESCs have a normal karyotype, maintain high telomerase activity, and exhibit remarkable long-term proliferative potential.[6]

Pluripotent

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 germ layers generate each of the more than 220 cell types in the adult human body. When provided with the appropriate signals, ESCs initially form precursor cells that in subsequently differentiate into the desired cell types. Pluripotency distinguishes embryonic stem cells from adult stem cells, which are multipotent and can only produce a limited number of cell types.

Self renewal and repair of structure

Under defined conditions, embryonic stem cells are capable of self-renewing indefinitely in an undifferentiated state. Self-renewal conditions must prevent the cells from clumping and maintain an environment that supports an unspecialized state.[7] Typically this is done in the lab with media containing serum and leukemia inhibitory factor or serum-free media supplements with two inhibitory drugs ("2i"), the MEK inhibitor PD03259010 and GSK-3 inhibitor CHIR99021.[8]

Growth

ESCs divide very frequently due to a shortened G1 phase in their cell cycle. Rapid cell division allows the cells to quickly grow in number, but not size, which is important for early embryo development. In ESCs, cyclin A and cyclin E proteins involved in the G1/S transition are always expressed at high levels.[9] Cyclin-dependent kinases such as CDK2 that promote cell cycle progression are overactive, in part due to downregulation of their inhibitors.[10] Retinoblastoma proteins that inhibit the transcription factor E2F until the cell is ready to enter S phase are hyperphosphorylated and inactivated in ESCs, leading to continual expression of proliferation genes.[9] These changes result in accelerated cycles of cell division. Although high expression levels of pro-proliferative proteins and a shortened G1 phase have been linked to maintenance of pluripotency,[11][12] ESCs grown in serum-free 2i conditions do express hypo-phosphorylated active Retinoblastoma proteins and have an elongated G1 phase.[13] Despite this difference in the cell cycle when compared to ESCs grown in media containing serum these cells have similar pluripotent characteristics.[14] Pluripotency factors Oct4 and Nanog play a role in transcriptionally regulating the embryonic stem cell cycle.[15][16]

Uses

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 promise 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.[17] In addition to their potential in regenerative medicine, embryonic stem cells provide a possible alternative source of tissue/organs which serves as a possible solution to the donor shortage dilemma. There are some ethical controversies surrounding this though (see Ethical debate section below). Aside from these uses, ESCs can also be used for research on early human development, certain genetic disease, and in vitro toxicology testing.[6]

Utilizations

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."[18]

Tissue engineering

 
Embryoid bodies 24 hours after formation.

In tissue engineering, the use of stem cells are known to be of importance. In order to successfully engineer a tissue, the cells used must be able to perform specific biological functions such as secretion of cytokines, signaling molecules, interacting with neighboring cells, and producing an extracellular matrix in the correct organization. Stem cells demonstrates these specific biological functions along with being able to self-renew and differentiate into one or more types of specialized cells. Embryonic stem cells is one of the sources that are being considered for the use of tissue engineering.[19] The use of human embryonic stem cells have opened many new possibilities for tissue engineering, however, there are many hurdles that must be made before human embryonic stem cell can even be utilized. It is theorized that if embryonic stem cells can be altered to not evoke the immune response when implanted into the patient then this would be a revolutionary step in tissue engineering.[20] Embryonic stem cells are not limited to tissue engineering.

Cell replacement therapies

Research has focused on differentiating ESCs into a variety of cell types for eventual use as cell replacement therapies. Some of the cell types that have or are currently being developed include cardiomyocytes, neurons, hepatocytes, bone marrow cells, islet cells and endothelial cells.[21] However, the derivation of such cell types from ESCs is not without obstacles, therefore research has focused on overcoming these barriers. For example, studies are underway to differentiate ESCs into tissue specific cardiomyocytes and to eradicate their immature properties that distinguish them from adult cardiomyocytes.[22]

Clinical potential

  • Researchers have differentiated ESCs into dopamine-producing cells with the hope that these neurons could be used in the treatment of Parkinson's disease.[23][24]
  • ESCs have been differentiated to natural killer cells and bone tissue.[25]
  • Studies involving ESCs are underway to provide an alternative treatment for diabetes. For example ESCs have been differentiated into insulin-producing cells,[26] and researchers at Harvard University were able to produce large quantities of pancreatic beta cells from ESCs.[27]
  • 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.[28]

Drug discovery

Besides becoming an important alternative to organ transplants, ESCs are also being used in the field of toxicology, and as cellular screens to uncover new chemical entities 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.[21] ESC derived cardiomyocytes have been shown to respond to pharmacological stimuli and hence can be used to assess cardiotoxicity such as torsades de pointes.[29]

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, research has focused on establishing fully functional ESC-derived hepatocytes with stable phase I and II enzyme activity.[30]

Models of genetic disorder

Several new studies have started to address the concept of modeling genetic disorders with embryonic stem cells. Either by genetically manipulating the cells, or more recently, by deriving diseased cell lines identified by prenatal genetic diagnosis (PGD), modeling genetic disorders is something that has been accomplished with stem cells. This approach may very well prove valuable 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 couples who have 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.[31]

Repair of DNA damage

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.[32] 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.[33] 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.[33] This type of repair depends on the interaction of the two sister chromosomes[verification needed] 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.[34] Rather they undergo programmed cell death (apoptosis) in response to DNA damage.[35] 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.[36] Consistent with this strategy, mouse ES stem cells have a mutation frequency about 100-fold lower than that of isogenic mouse somatic cells.[37]

Clinical trial

On January 23, 2009, Phase I clinical trials for transplantation of oligodendrocytes (a cell type of the brain and spinal cord) derived from human ESCs into spinal cord-injured individuals received approval from the U.S. Food and Drug Administration (FDA), marking it the world's first human ESC human trial.[38] 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 ESCs that had been pushed into an oligodendrocytic lineage.[39] 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.[40] 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.[41]

In October 2010 researchers enrolled and administered ESCs to the first patient at Shepherd Center in Atlanta.[42] 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.[43] In 2013 BioTime, 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.[44]

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.[45]

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.[45]

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.[45]

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.[45]

Concern and controversy

Adverse effects

The major concern with the possible transplantation of ESCs into patients as therapies is their ability to form tumors including teratomas.[46] Safety issues prompted the FDA to place a hold on the first ESC clinical trial, however no tumors were observed.

The main strategy to enhance the safety of ESCs for potential clinical use is to differentiate the ESCs into specific cell types (e.g. neurons, muscle, liver cells) that have reduced or eliminated ability to cause tumors. Following differentiation, the cells are subjected to sorting by flow cytometry for further purification. ESCs are predicted to be inherently safer than iPS cells created with genetically integrating viral vectors because they are not genetically modified with genes such as c-Myc that are linked to cancer. Nonetheless, ESCs express very high levels of the iPS inducing genes and these genes including Myc are essential for ESC self-renewal and pluripotency,[47] and potential strategies to improve safety by eliminating c-Myc expression are unlikely to preserve the cells' "stemness". However, N-myc and L-myc have been identified to induce iPS cells instead of c-myc with similar efficiency.[48] Later protocols to induce pluripotency bypass these problems completely by using non-integrating RNA viral vectors such as sendai virus or mRNA transfection.

Ethical debate

Due to the nature of embryonic stem cell research, there are a lot of controversial opinions on the topic. Since harvesting embryonic stem cells usually necessitates destroying the embryo from which those cells are obtained, the moral status of the embryo comes into question. Some people claim that the 5-day-old mass of cells is too young to achieve personhood or that the embryo, if donated from an IVF clinic (where labs typically acquire embryos), would otherwise go to medical waste anyway. Opponents of ESC research claim that an embryo is a human life, therefore destroying it is murder and the embryo must be protected under the same ethical view as a more developed human being.[49]

History

  • 1964: Lewis Kleinsmith and G. Barry Pierce Jr. isolated a single type of cell from a teratocarcinoma, a tumor now known from a germ cell.[50] These cells were isolated from the teratocarcinoma replicated and grew in cell culture as a stem cell and are now known as embryonal carcinoma (EC) cells.[citation needed] Although similarities in morphology and differentiating potential (pluripotency) led to the use of EC cells as the in vitro model for early mouse development,[51] 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 a 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 cell from these embryos.[52] 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".[53] She showed that embryos could be cultured in vitro and that ES cells could be derived from these embryos.
  • 1989: Mario R. Cappechi, Martin J. Evans, and Oliver Smithies publish their research that details their isolation and genetic modifications of embryonic stem cells, creating the first "knockout mice".[54] In creating knockout mice, this publication provided scientists with an entirely new way to study disease.
  •  
    Dolly the sheep cell differentiation
    1996: Dolly, was the first mammal cloned from an adult cell by the Roslin Institute of the University of Edinburgh.[55] This experiment instituted the proposition that specialized adult cells obtain the genetic makeup to perform a specific task; which established a basis for further research within a variety of cloning techniques. The Dolly experiment was performed by obtaining the mammalian udder cells from a sheep (Dolly) and differentiating these cells until division was concluded. An egg cell was then procured from a different sheep host and the nucleus was removed. An udder cell was placed next to the egg cell and connected by electricity causing this cell to share DNA. This egg cell differentiated into an embryo and the embryo was inserted into a third sheep which gave birth to the clone version of Dolly.[56]
  • 1998: A team from the University of Wisconsin, Madison (James A. Thomson, Joseph Itskovitz-Eldor, Sander S. Shapiro, Michelle A. Waknitz, Jennifer J. Swiergiel, Vivienne S. Marshall, and Jeffrey M. Jones) publish a paper titled "Embryonic Stem Cell Lines Derived From Human Blastocysts". The researchers behind this study not only created the first embryonic stem cells, but recognized 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.[57]
  • 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.[58]
  • 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.[59]
  • 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.[60]
  • 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.[61]

Techniques and conditions for derivation and culture

Derivation from humans

In vitro fertilization generates multiple embryos. The surplus of embryos is not clinically used or is unsuitable for implantation into the patient, and therefore may be donated by the donor with consent. Human embryonic stem cells can be derived from these donated embryos or additionally they can also be extracted from cloned embryos created using a cell from a patient and a donated egg through the process of somatic cell nuclear transfer.[62] The inner cell mass (cells of interest), from the blastocyst stage of the embryo, is separated from the trophectoderm, the cells that would differentiate into extra-embryonic tissue. Immunosurgery, the process in which antibodies are bound to the trophectoderm and removed by another solution, and mechanical dissection are performed to achieve separation. The resulting inner cell mass cells are plated onto cells that will supply support. The inner cell mass cells attach and expand further to form a human embryonic cell line, which are undifferentiated. These cells are fed daily and are enzymatically or mechanically separated every four to seven days. For differentiation to occur, the human embryonic stem cell line is removed from the supporting cells to form embryoid bodies, is co-cultured with a serum containing necessary signals, or is grafted in a three-dimensional scaffold to result.[63]

Derivation from other animals

Embryonic stem cells are derived from the inner cell mass of the early embryo, which are harvested from the donor mother animal. Martin Evans and Matthew Kaufman reported a technique that delays embryo implantation, allowing the inner cell mass to increase. This process includes removing the donor mother's ovaries and dosing her with progesterone, changing the hormone environment, which causes the embryos to remain free in the uterus. After 4–6 days of this intrauterine culture, the embryos are harvested and grown in in vitro culture until the inner cell mass forms “egg cylinder-like structures,” which are dissociated into single cells, and plated on fibroblasts treated with mitomycin-c (to prevent fibroblast mitosis). Clonal cell lines are created by growing up a single cell. Evans and Kaufman showed that the cells grown out from these cultures could form teratomas and embryoid bodies, and differentiate in vitro, all of which indicating that the cells are pluripotent.[52]

Gail Martin derived and cultured her ES cells differently. She removed the embryos from the donor mother at approximately 76 hours after copulation and cultured them overnight in a medium containing serum. The following day, she removed the inner cell mass from the late blastocyst using microsurgery. The extracted inner cell mass was cultured on fibroblasts treated with mitomycin-c in a medium containing serum and conditioned by ES cells. After approximately one week, colonies of cells grew out. These cells grew in culture and demonstrated pluripotent characteristics, as demonstrated by the ability to form teratomas, differentiate in vitro, and form embryoid bodies. Martin referred to these cells as ES cells.[53]

It is now known that the feeder cells provide leukemia inhibitory factor (LIF) and serum provides bone morphogenetic proteins (BMPs) that are necessary to prevent ES cells from differentiating.[64][65] These factors are extremely important for the efficiency of deriving ES cells. Furthermore, it has been demonstrated that different mouse strains have different efficiencies for isolating ES cells.[66] Current uses for mouse ES cells include the generation of transgenic mice, including knockout mice. For human treatment, there is a need for patient specific pluripotent cells. Generation of human ES cells is more difficult and faces ethical issues. So, in addition to human ES cell research, many groups are focused on the generation of induced pluripotent stem cells (iPS cells).[67]

Potential methods for new cell line derivation

On August 23, 2006, the online edition of Nature scientific journal published a letter by Dr. Robert Lanza (medical director of Advanced Cell Technology in Worcester, MA) stating that his team had found a way to extract embryonic stem cells without destroying the actual embryo.[68] This technical achievement would potentially enable scientists to work with new lines of embryonic stem cells derived using public funding in the US, where federal funding was at the time limited to research using embryonic stem cell lines derived prior to August 2001. In March, 2009, the limitation was lifted.[69]

Human embryonic stem cells have also been derived by somatic cell nuclear transfer (SCNT).[70][71] This approach has also sometimes been referred to as "therapeutic cloning" because SCNT bears similarity to other kinds of cloning in that nuclei are transferred from a somatic cell into an enucleated zygote. However, in this case SCNT was used to produce embryonic stem cell lines in a lab, not living organisms via a pregnancy. The "therapeutic" part of the name is included because of the hope that SCNT produced embryonic stem cells could have clinical utility.

Induced pluripotent stem cells

The iPS cell technology was pioneered by Shinya Yamanaka's lab in Kyoto, Japan, who showed in 2006 that the introduction of four specific genes encoding transcription factors could convert adult cells into pluripotent stem cells.[72] He was awarded the 2012 Nobel Prize along with Sir John Gurdon "for the discovery that mature cells can be reprogrammed to become pluripotent."[73]

In 2007, it was shown that pluripotent stem cells, highly similar to embryonic stem cells, can be induced by the delivery of four factors (Oct3/4, Sox2, c-Myc, and Klf4) to differentiated cells.[74] Utilizing the four genes previously listed, the differentiated cells are "reprogrammed" into pluripotent stem cells, allowing for the generation of pluripotent/embryonic stem cells without the embryo. The morphology and growth factors of these lab induced pluripotent cells, are equivalent to embryonic stem cells, leading these cells to be known as induced pluripotent stem cells (iPS cells).[75] This observation was observed in mouse pluripotent stem cells, originally, but now can be performed in human adult fibroblasts using the same four genes. [76]

Because ethical concerns regarding embryonic stem cells typically are about their derivation from terminated embryos, it is believed that reprogramming to these iPS cells may be less controversial.

This may enable the generation of patient specific ES cell lines that could potentially be used for cell replacement therapies. In addition, this will allow the generation of ES cell lines from patients with a variety of genetic diseases and will provide invaluable models to study those diseases.

However, as a first indication that the iPS cell technology can in rapid succession lead to new cures, it was used by a research team headed by Rudolf Jaenisch of the Whitehead Institute for Biomedical Research in Cambridge, Massachusetts, to cure mice of sickle cell anemia, as reported by Science journal's online edition on December 6, 2007.[77][78]

On January 16, 2008, a California-based company, Stemagen, announced that they had created the first mature cloned human embryos from single skin cells taken from adults. These embryos can be harvested for patient matching embryonic stem cells.[79]

Contamination by reagents used in cell culture

The online edition of Nature Medicine published a study on January 24, 2005, which stated that the human embryonic stem cells available for federally funded research are contaminated with non-human molecules from the culture medium used to grow the cells.[80] It is a common technique to use mouse cells and other animal cells to maintain the pluripotency of actively dividing stem cells. The problem was discovered when non-human sialic acid in the growth medium was found to compromise the potential uses of the embryonic stem cells in humans, according to scientists at the University of California, San Diego.[81]

However, a study published in the online edition of Lancet Medical Journal on March 8, 2005, detailed information about a new stem cell line that was derived from human embryos under completely cell- and serum-free conditions. After more than 6 months of undifferentiated proliferation, these cells demonstrated the potential to form derivatives of all three embryonic germ layers both in vitro and in teratomas. These properties were also successfully maintained (for more than 30 passages) with the established stem cell lines.[82]

Muse cells

Muse cells (Multi-lineage differentiating stress enduring cell) are non-cancerous pluripotent stem cell found in adults.[83][84] They were discovered in 2010 by Mari Dezawa and her research group.[83] Muse cells reside in the connective tissue of nearly every organ including the umbilical cord, bone marrow and peripheral blood.[85][83][86][87][88] They are collectable from commercially obtainable mesenchymal cells such as human fibroblasts, bone marrow-mesenchymal stem cells and adipose-derived stem cells.[89][90][91] Muse cells are able to generate cells representative of all three germ layers from a single cell both spontaneously and under cytokine induction. Expression of pluripotency genes and triploblastic differentiation are self-renewable over generations. Muse cells do not undergo teratoma formation when transplanted into a host environment in vivo, eradicating the risk of tumorigenesis through unbridled cell proliferation.[83]

See also

References

  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. ^ a b c "Stem Cell Basics | STEM Cell Information". stemcells.nih.gov. Retrieved 5 June 2022.
  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. pp. 96. ISBN 978-1601521576.
  5. ^ Carla A Herberts; Marcel SG Kwa; Harm PH Hermsen (2011). "Risk factors in the development of stem cell therapy". Journal of Translational Medicine. 9 (29): 29. doi:10.1186/1479-5876-9-29. PMC 3070641. PMID 21418664.
  6. ^ a b 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.
  7. ^ 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. S2CID 7201396.
  8. ^ Martello, G.; Smith, A. (2014). "The nature of embryonic stem cells". Annual Review of Cell and Developmental Biology. 30: 647–75. doi:10.1146/annurev-cellbio-100913-013116. PMID 25288119.
  9. ^ a b Boward, B.; Wu, T.; Dalton, S. (2016). "Concise Review: Control of Cell Fate Through Cell Cycle and Pluripotency Networks". Stem Cells. 34 (6): 1427–36. doi:10.1002/stem.2345. PMC 5201256. PMID 26889666.
  10. ^ White, J.; Stead, E.; Faast, R.; Conn, S.; Cartwright, P.; Dalton, S. (2005). "Developmental activation of the Rb-E2F pathway and establishment of cell cycle-regulated cyclin-dependent kinase activity during embryonic stem cell differentiation". Molecular Biology of the Cell. 16 (4): 2018–27. doi:10.1091/mbc.e04-12-1056. PMC 1073679. PMID 15703208.
  11. ^ Ter Huurne, Menno; Stunnenberg, Hendrik G. (21 April 2021). "G1-phase progression in pluripotent stem cells". Cellular and Molecular Life Sciences. 21 (10): 4507–4519. doi:10.1007/s00018-021-03797-8. ISSN 1875-9777. PMC 8195903. PMID 33884444.
  12. ^ Singh, Amar M.; Dalton, Stephen (2009-08-07). "The cell cycle and Myc intersect with mechanisms that regulate pluripotency and reprogramming". Cell Stem Cell. 5 (2): 141–149. doi:10.1016/j.stem.2009.07.003. ISSN 1875-9777. PMC 2909475. PMID 19664987.
  13. ^ Ter Huurne, Menno; Chappell, James; Dalton, Stephen; Stunnenberg, Hendrik G. (5 October 2017). "Distinct Cell-Cycle Control in Two Different States of Mouse Pluripotency". Cell Stem Cell. 21 (4): 449–455.e4. doi:10.1016/j.stem.2017.09.004. ISSN 1875-9777. PMC 5658514. PMID 28985526.
  14. ^ Ying, Qi-Long; Wray, Jason; Nichols, Jennifer; Batlle-Morera, Laura; Doble, Bradley; Woodgett, James; Cohen, Philip; Smith, Austin (2008-05-22). "The ground state of embryonic stem cell self-renewal". Nature. 453 (7194): 519–523. Bibcode:2008Natur.453..519Y. doi:10.1038/nature06968. ISSN 1476-4687. PMC 5328678. PMID 18497825.
  15. ^ Lee, J.; Go, Y.; Kang, I.; Han, Y. M.; Kim, J. (2010). "Oct-4 controls cell-cycle progression of embryonic stem cells". The Biochemical Journal. 426 (2): 171–81. doi:10.1042/BJ20091439. PMC 2825734. PMID 19968627.
  16. ^ Zhang, X.; Neganova, I.; Przyborski, S.; Yang, C.; Cooke, M.; Atkinson, S. P.; Anyfantis, G.; Fenyk, S.; Keith, W. N.; Hoare, S. F.; Hughes, O.; Strachan, T.; Stojkovic, M.; Hinds, P. W.; Armstrong, L.; Lako, M. (2009). "A role for NANOG in G1 to S transition in human embryonic stem cells through direct binding of CDK6 and CDC25A". The Journal of Cell Biology. 184 (1): 67–82. doi:10.1083/jcb.200801009. PMC 2615089. PMID 19139263.
  17. ^ Mahla, Ranjeet (July 19, 2016). "Stem Cell Applications in Regenerative Medicine and Disease Therapeutics". International Journal of Cell Biology. 2016: 6940283. doi:10.1155/2016/6940283. PMC 4969512. PMID 27516776.
  18. ^ 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. PMID 11917100.
  19. ^ Vats, A; Tolley, N S; Bishop, A E; Polak, J M (2005-08-01). "Embryonic Stem Cells and Tissue Engineering: Delivering Stem Cells to the Clinic". Journal of the Royal Society of Medicine. 98 (8): 346–350. doi:10.1177/014107680509800804. ISSN 0141-0768. PMC 1181832. PMID 16055897.
  20. ^ Heath, Carole A. (2000-01-01). "Cells for tissue engineering". Trends in Biotechnology. 18 (1): 17–19. doi:10.1016/S0167-7799(99)01396-7. ISSN 0167-7799. PMID 10631775.
  21. ^ 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): 214–223. doi:10.1093/toxsci/kfh100. PMID 15014205.
  22. ^ Siu, CW; Moore, JC; Li, RA (2007). "Human embryonic stem cell-derived cardiomyocytes for heart therapies". Cardiovascular & Hematological Disorders Drug Targets. 7 (2): 145–152. doi:10.2174/187152907780830851. PMID 17584049.
  23. ^ 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. PMID 15310843.
  24. ^ Parish, CL; Arenas, E (2007). "Stem-cell-based strategies for the treatment of Parkinson's disease". Neuro-Degenerative Diseases. 4 (4): 339–347. doi:10.1159/000101892. PMID 17627139. S2CID 37229348.
  25. ^ Waese, EY; Kandel, RA; Stanford, WL (2008). "Application of stem cells in bone repair". Skeletal Radiology. 37 (7): 601–608. doi:10.1007/s00256-007-0438-8. PMID 18193216. S2CID 12401889.
  26. ^ 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): 1392–1401. doi:10.1038/nbt1259. PMID 17053790. S2CID 11040949.
  27. ^ Colen, B.D. (9 October 2014) Giant leap against diabetes The Harvard Gazette, Retrieved 24 November 2014
  28. ^ 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:10.1093/eurheartj/ehu192. PMID 24835485.
  29. ^ Jensen, J; Hyllner, J; Björquist, P (2009). "Human embryonic stem cell technologies and drug discovery". Journal of Cellular Physiology. 219 (3): 513–519. doi:10.1002/jcp.21732. PMID 19277978. S2CID 36354049.
  30. ^ 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): 929–937. doi:10.1016/j.tiv.2007.01.021. PMID 17346923.
  31. ^ "Dr. Yury Verlinsky, 1943–2009: Expert in reproductive technology" 2009-08-08 at the Wayback Machine Chicago Tribune, July 20, 2009
  32. ^ 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): 2902–2906. doi:10.4161/cc.7.18.6679. PMC 2754209. PMID 18769152.
  33. ^ 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): 1699–1711. doi:10.1089/scd.2010.0058. PMC 3128311. PMID 20446816.
  34. ^ 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): 14443–14448. Bibcode:2004PNAS..10114443H. doi:10.1073/pnas.0401346101. PMC 521944. PMID 15452351.
  35. ^ 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): 145–155. doi:10.1016/S0960-9822(98)70061-2. PMID 9443911. S2CID 13938854.
  36. ^ 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): 145–178. doi:10.1016/S1383-5742(02)00009-1. PMID 12052432.
  37. ^ 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): 3586–3590. Bibcode:2002PNAS...99.3586C. doi:10.1073/pnas.062527199. PMC 122567. PMID 11891338.
  38. ^ "FDA approves human embryonic stem cell study – CNN.com". January 23, 2009. Retrieved May 1, 2010.
  39. ^ 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" (PDF). J. Neurosci. 25 (19): 4694–4705. doi:10.1523/JNEUROSCI.0311-05.2005. PMC 6724772. PMID 15888645.
  40. ^ Reinberg, Steven (2009-01-23) FDA OKs 1st Embryonic Stem Cell Trial. The Washington Post
  41. ^ . geron.com (August 27, 2009)
  42. ^ Vergano, Dan (11 October 2010). "Embryonic stem cells used on patient for first time". USA Today. Retrieved 12 October 2010.
  43. ^ Brown, Eryn (November 15, 2011). "Geron exits stem cell research". LA Times. Retrieved 2011-11-15.
  44. ^ "Great news: BioTime Subsidiary Asterias Acquires Geron Embryonic Stem Cell Program". iPScell.com. October 1, 2013.
  45. ^ a b c d California Institute of Regenerative Medicine 2017-10-24 at the Wayback Machine. BioTime, Inc.
  46. ^ Knoepfler, Paul S. (2009). "Deconstructing Stem Cell Tumorigenicity: A Roadmap to Safe Regenerative Medicine". Stem Cells. 27 (5): 1050–1056. doi:10.1002/stem.37. PMC 2733374. PMID 19415771.
  47. ^ Varlakhanova, Natalia V.; Cotterman, Rebecca F.; Devries, Wilhelmine N.; Morgan, Judy; Donahue, Leah Rae; Murray, Stephen; Knowles, Barbara B.; Knoepfler, Paul S. (2010). "Myc maintains embryonic stem cell pluripotency and self-renewal". Differentiation. 80 (1): 9–19. doi:10.1016/j.diff.2010.05.001. PMC 2916696. PMID 20537458.
  48. ^ Wernig, Marius; Meissner, Alexander; Cassady, John P; Jaenisch, Rudolf (2008). "C-Myc is Dispensable for Direct Reprogramming of Mouse Fibroblasts". Cell Stem Cell. 2 (1): 10–12. doi:10.1016/j.stem.2007.12.001. PMID 18371415.
  49. ^ King, Nancy; Perrin, Jacob (July 7, 2014). "Ethical issues in stem cell research and therapy". Stem Cell Research & Therapy. 5 (4): 85. doi:10.1186/scrt474. PMC 4097842. PMID 25157428.
  50. ^ Kleinsmith LJ, Pierce GB Jr (1964). "Multipotentiality of Single Embryoncal Carcinoma Cells". Cancer Res. 24: 1544–1551. PMID 14234000.
  51. ^ Martin GR (1980). "Teratocarcinomas and mammalian embryogenesis". Science. 209 (4458): 768–776. Bibcode:1980Sci...209..768M. doi:10.1126/science.6250214. PMID 6250214.
  52. ^ a b Evans M, Kaufman M (1981). "Establishment in culture of pluripotent cells from mouse embryos". Nature. 292 (5819): 154–156. Bibcode:1981Natur.292..154E. doi:10.1038/292154a0. PMID 7242681. S2CID 4256553.
  53. ^ a b 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): 7634–7638. Bibcode:1981PNAS...78.7634M. doi:10.1073/pnas.78.12.7634. PMC 349323. PMID 6950406.
  54. ^ "The 2007 Nobel Prize in Physiology or Medicine – Advanced Information". Nobel Prize. Nobel Media.
  55. ^ "The Life of Dolly | Dolly the Sheep". Retrieved 2022-02-21.
  56. ^ Klotzko, Arlene Judith; Klotzko, Visiting Scholar Royal Free and University College Medical School Arlene Judith (2006). A Clone of Your Own?. Cambridge University Press. ISBN 978-0-521-85294-4.
  57. ^ Thompson, James A.; Itskovitz-Eldor, Joseph; Shapiro, Sander S.; Waknitz, Michelle A.; Swiergiel, Jennifer J.; Marshall, Vivienne S.; Jones, Jeffrey M. (6 November 1998). "Embryonic Stem Cell Lines Derived From Human Blastocyst". Science. 282 (5391): 1145–1147. Bibcode:1998Sci...282.1145T. doi:10.1126/science.282.5391.1145. PMID 9804556.
  58. ^ "President George W. Bush's address on stem cell research". CNN Inside Politics. Aug 9, 2001.
  59. ^ 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:10.1016/j.cell.2006.07.024. hdl:2433/159777. PMID 16904174. S2CID 1565219.
  60. ^ Wadman, Meredith (27 January 2009). "Stem cells ready for primetime". Nature. 457 (7229): 516. doi:10.1038/457516a. PMID 19177087.
  61. ^ "Executive Order 13505—Removing Barriers To Responsible Scientific Research Involving Human Stem Cells" (PDF). Federal Register: Presidential Documents. 74 (46). 11 March 2009.
  62. ^ Mountford, JC (2008). "Human embryonic stem cells: origins, characteristics and potential for regenerative therapy". Transfus Med. 18 (1): 1–12. doi:10.1111/j.1365-3148.2007.00807.x. PMID 18279188. S2CID 20874633.
  63. ^ Thomson JA, Itskovitz-Eldor J, Shapiro SS, Waknitz MA, Swiergiel JJ, Marshall VS, Jones JM (1998). "Embryonic stem cell lines derived from human blastocysts". Science. 282 (5391): 1145–1147. Bibcode:1998Sci...282.1145T. doi:10.1126/science.282.5391.1145. PMID 9804556.
  64. ^ Smith AG, Heath JK, Donaldson DD, Wong GG, Moreau J, Stahl M, Rogers D (1988). "Inhibition of pluripotential embryonic stem cell differentiation by purified polypeptides". Nature. 336 (6200): 688–690. Bibcode:1988Natur.336..688S. doi:10.1038/336688a0. PMID 3143917. S2CID 4325137.
  65. ^ Williams RL, Hilton DJ, Pease S, Willson TA, Stewart CL, Gearing DP, Wagner EF, Metcalf D, Nicola NA, Gough NM (1988). "Myeloid leukaemia inhibitory factor maintains the developmental potential of embryonic stem cells". Nature. 336 (6200): 684–687. Bibcode:1988Natur.336..684W. doi:10.1038/336684a0. PMID 3143916. S2CID 4346252.
  66. ^ Ledermann B, Bürki K (1991). "Establishment of a germ-line competent C57BL/6 embryonic stem cell line". Exp Cell Res. 197 (2): 254–258. doi:10.1016/0014-4827(91)90430-3. PMID 1959560.
  67. ^ Takahashi K, Tanabe K, Ohnuki M, Narita M, Ichisaka T, Tomoda K, Yamanaka S (2007). "Induction of pluripotent stem cells from adult human fibroblasts by defined factors". Cell. 131 (5): 861–872. doi:10.1016/j.cell.2007.11.019. hdl:2433/49782. PMID 18035408. S2CID 8531539.
  68. ^ Klimanskaya I, Chung Y, Becker S, Lu SJ, Lanza R (2006). "Human embryonic stem cell lines derived from single blastomeres". Nature. 444 (7118): 481–485. Bibcode:2006Natur.444..481K. doi:10.1038/nature05142. PMID 16929302. S2CID 84792371.
  69. ^ US scientists relieved as Obama lifts ban on stem cell research, The Guardian, 10 March 2009
  70. ^ Tachibana, Masahito; Amato, Paula; Sparman, Michelle; Gutierrez, Nuria Marti; Tippner-Hedges, Rebecca; Ma, Hong; Kang, Eunju; Fulati, Alimujiang; Lee, Hyo-Sang; Sritanaudomchai, Hathaitip; Masterson, Keith (2013-06-06). "Human Embryonic Stem Cells Derived by Somatic Cell Nuclear Transfer". Cell. 153 (6): 1228–1238. doi:10.1016/j.cell.2013.05.006. ISSN 0092-8674. PMC 3772789. PMID 23683578.
  71. ^ Chung, Young Gie; Eum, Jin Hee; Lee, Jeoung Eun; Shim, Sung Han; Sepilian, Vicken; Hong, Seung Wook; Lee, Yumie; Treff, Nathan R.; Choi, Young Ho; Kimbrel, Erin A.; Dittman, Ralph E. (2014-06-05). "Human Somatic Cell Nuclear Transfer Using Adult Cells". Cell Stem Cell. 14 (6): 777–780. doi:10.1016/j.stem.2014.03.015. ISSN 1934-5909. PMID 24746675.
  72. ^ Takahashi, K; Yamanaka, S (2006). "Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors". Cell. 126 (4): 663–676. doi:10.1016/j.cell.2006.07.024. hdl:2433/159777. PMID 16904174. S2CID 1565219. 
  73. ^ "The Nobel Prize in Physiology or Medicine – 2012 Press Release". Nobel Media AB. 8 October 2012.
  74. ^ Wernig, Marius; Meissner, Alexander; Foreman, Ruth; Brambrink, Tobias; Ku, Manching; Hochedlinger, Konrad; Bernstein, Bradley E.; Jaenisch, Rudolf (2007-07-19). "In vitro reprogramming of fibroblasts into a pluripotent ES-cell-like state". Nature. 448 (7151): 318–324. Bibcode:2007Natur.448..318W. doi:10.1038/nature05944. ISSN 1476-4687. PMID 17554336. S2CID 4377572.
  75. ^ Takahashi, Kazutoshi; Yamanaka, Shinya (2006-08-25). "Induction of Pluripotent Stem Cells from Mouse Embryonic and Adult Fibroblast Cultures by Defined Factors". Cell. 126 (4): 663–676. doi:10.1016/j.cell.2006.07.024. ISSN 0092-8674. PMID 16904174. S2CID 1565219.
  76. ^ Takahashi, Kazutoshi; Tanabe, Koji; Ohnuki, Mari; Narita, Megumi; Ichisaka, Tomoko; Tomoda, Kiichiro; Yamanaka, Shinya (2007-11-30). "Induction of Pluripotent Stem Cells from Adult Human Fibroblasts by Defined Factors". Cell. 131 (5): 861–872. doi:10.1016/j.cell.2007.11.019. ISSN 0092-8674. PMID 18035408. S2CID 8531539.
  77. ^ Weiss, Rick (2007-12-07). "Scientists Cure Mice Of Sickle Cell Using Stem Cell Technique: New Approach Is From Skin, Not Embryos". The Washington Post. pp. A02.
  78. ^ Hanna, J.; Wernig, M.; Markoulaki, S.; Sun, C.-W.; Meissner, A.; Cassady, J. P.; Beard, C.; Brambrink, T.; Wu, L.-C.; Townes, T. M.; Jaenisch, R. (2007). "Treatment of Sickle Cell Anemia Mouse Model with iPS Cells Generated from Autologous Skin". Science. 318 (5858): 1920–1923. Bibcode:2007Sci...318.1920H. doi:10.1126/science.1152092. PMID 18063756. S2CID 657569.
  79. ^ Helen Briggs (2008-01-17). "US team makes embryo clone of men". BBC. pp. A01.
  80. ^ Ebert, Jessica (24 January 2005). . Nature News. London: Nature Publishing Group. doi:10.1038/news050124-1. Archived from the original on 2010-09-24. Retrieved 2009-02-27.
  81. ^ Martin MJ, Muotri A, Gage F, Varki A (2005). "Human embryonic stem cells express an immunogenic nonhuman sialic acid". Nat. Med. 11 (2): 228–232. doi:10.1038/nm1181. PMID 15685172. S2CID 13739919.
  82. ^ Klimanskaya I, Chung Y, Meisner L, Johnson J, West MD, Lanza R (2005). "Human embryonic stem cells derived without feeder cells". Lancet. 365 (9471): 1636–1641. doi:10.1016/S0140-6736(05)66473-2. PMID 15885296. S2CID 17139951.
  83. ^ a b c d Kuroda, Y.; Kitada, M.; Wakao, S.; Nishikawa, K.; Tanimura, Y.; Makinoshima, H.; Goda, M.; Akashi, H.; Inutsuka, A.; Niwa, A.; Shigemoto, T.; Nabeshima, Y.; Nakahata, T.; Nabeshima, Y.-i.; Fujiyoshi, Y.; Dezawa, M. (2010). "Unique multipotent cells in adult human mesenchymal cell populations". Proceedings of the National Academy of Sciences. 107 (19): 8639–8643. Bibcode:2010PNAS..107.8639K. doi:10.1073/pnas.0911647107. PMC 2889306. PMID 20421459.
  84. ^ Muse Cells | SpringerLink.
  85. ^ Zikuan Leng 1 2, Dongming Sun 2, Zihao Huang 3, Iman Tadmori 2, Ning Chiang 2, Nikhit Kethidi 2, Ahmed Sabra 2, Yoshihiro Kushida 4, Yu-Show Fu 3, Mari Dezawa 4, Xijing He 1, Wise Young 2Quantitative Analysis of SSEA3+ Cells from Human Umbilical Cord after Magnetic SortingCell Transplant . 2019 Jul;28(7):907–923.
  86. ^ Wakao, S.; Kitada, M.; Kuroda, Y.; Shigemoto, T.; Matsuse, D.; Akashi, H.; Tanimura, Y.; Tsuchiyama, K.; Kikuchi, T.; Goda, M.; Nakahata, T.; Fujiyoshi, Y.; Dezawa, M. (2011). "Multilineage-differentiating stress-enduring (Muse) cells are a primary source of induced pluripotent stem cells in human fibroblasts". Proceedings of the National Academy of Sciences. 108 (24): 9875–9880. Bibcode:2011PNAS..108.9875W. doi:10.1073/pnas.1100816108. PMC 3116385. PMID 21628574.
  87. ^ Dezawa, Mari (2016). "Muse Cells Provide the Pluripotency of Mesenchymal Stem Cells: Direct Contribution of Muse Cells to Tissue Regeneration". Cell Transplantation. 25 (5): 849–861. doi:10.3727/096368916X690881. PMID 26884346.
  88. ^ Hori, Emiko; Hayakawa, Yumiko; Hayashi, Tomohide; Hori, Satoshi; Okamoto, Soushi; Shibata, Takashi; Kubo, Michiya; Horie, Yukio; Sasahara, Masakiyo; Kuroda, Satoshi (2016). "Mobilization of Pluripotent Multilineage-Differentiating Stress-Enduring Cells in Ischemic Stroke". Journal of Stroke and Cerebrovascular Diseases. 25 (6): 1473–1481. doi:10.1016/j.jstrokecerebrovasdis.2015.12.033. PMID 27019988.
  89. ^ Kuroda, Yasumasa; Wakao, Shohei; Kitada, Masaaki; Murakami, Toru; Nojima, Makoto; Dezawa, Mari (2013). "Isolation, culture and evaluation of multilineage-differentiating stress-enduring (Muse) cells". Nature Protocols. 8 (7): 1391–1415. doi:10.1038/nprot.2013.076. PMID 23787896. S2CID 28597290.[unreliable medical source?]
  90. ^ Ogura, Fumitaka; Wakao, Shohei; Kuroda, Yasumasa; Tsuchiyama, Kenichiro; Bagheri, Mozhdeh; Heneidi, Saleh; Chazenbalk, Gregorio; Aiba, Setsuya; Dezawa, Mari (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 and Development. 23 (7): 717–728. doi:10.1089/scd.2013.0473. PMID 24256547.
  91. ^ Heneidi, Saleh; Simerman, Ariel A.; Keller, Erica; Singh, Prapti; Li, Xinmin; Dumesic, Daniel A.; Chazenbalk, Gregorio (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. Bibcode:2013PLoSO...864752H. doi:10.1371/journal.pone.0064752. PMC 3673968. PMID 23755141.

External links

  • Understanding Stem Cells: A View of the Science and Issues from the National Academies
  • National Institutes of Health
  • University of Oxford practical workshop on pluripotent stem cell technology 2016-04-08 at the Wayback Machine
  • Fact sheet on embryonic stem cells
  • Fact sheet on ethical issues in embryonic stem cell research
  • Information & Alternatives to Embryonic Stem Cell Research
  • A blog focusing specifically on ES cells and iPS cells including research, biotech, and patient-oriented issues

embryonic, stem, cell, escs, pluripotent, stem, cells, derived, from, inner, cell, mass, blastocyst, early, stage, implantation, embryo, human, embryos, reach, blastocyst, stage, days, post, fertilization, which, time, they, consist, cells, isolating, inner, c. Embryonic stem cells ESCs 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 inner cell mass embryoblast using immunosurgery results in destruction of the blastocyst a process which raises ethical issues including whether or not embryos at the pre implantation stage have the same moral considerations as embryos in the post implantation stage of development 3 4 Human embryonic stem cells in cell culture Pluripotent Embryonic stem cells are able to develop into any type of cell excepting those of the placenta Only embryonic stem cells of the morula are totipotent able to develop into any type of cell including those of the placenta Researchers are currently focusing heavily on the therapeutic potential of embryonic stem cells with clinical use being the goal for many laboratories 2 Potential uses include the treatment of diabetes and heart disease 2 The 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 such as tumors and unwanted immune responses have also been reported 5 Contents 1 Properties 1 1 Pluripotent 1 2 Self renewal and repair of structure 1 3 Growth 1 4 Uses 2 Utilizations 2 1 Tissue engineering 2 2 Cell replacement therapies 2 3 Clinical potential 2 4 Drug discovery 2 5 Models of genetic disorder 2 6 Repair of DNA damage 2 7 Clinical trial 3 Concern and controversy 3 1 Adverse effects 3 2 Ethical debate 4 History 5 Techniques and conditions for derivation and culture 5 1 Derivation from humans 5 2 Derivation from other animals 5 3 Potential methods for new cell line derivation 5 4 Induced pluripotent stem cells 5 5 Contamination by reagents used in cell culture 5 6 Muse cells 6 See also 7 References 8 External linksProperties Edit IPS Cell 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 embryonic cell type and by their ability to self renew It is these traits that makes them valuable in the scientific and medical fields ESCs have a normal karyotype maintain high telomerase activity and exhibit remarkable long term proliferative potential 6 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 germ layers generate each of the more than 220 cell types in the adult human body When provided with the appropriate signals ESCs initially form precursor cells that in subsequently differentiate into the desired cell types Pluripotency distinguishes embryonic stem cells from adult stem cells which are multipotent and can only produce a limited number of cell types Self renewal and repair of structure Edit Under defined conditions embryonic stem cells are capable of self renewing indefinitely in an undifferentiated state Self renewal conditions must prevent the cells from clumping and maintain an environment that supports an unspecialized state 7 Typically this is done in the lab with media containing serum and leukemia inhibitory factor or serum free media supplements with two inhibitory drugs 2i the MEK inhibitor PD03259010 and GSK 3 inhibitor CHIR99021 8 Growth Edit ESCs divide very frequently due to a shortened G1 phase in their cell cycle Rapid cell division allows the cells to quickly grow in number but not size which is important for early embryo development In ESCs cyclin A and cyclin E proteins involved in the G1 S transition are always expressed at high levels 9 Cyclin dependent kinases such as CDK2 that promote cell cycle progression are overactive in part due to downregulation of their inhibitors 10 Retinoblastoma proteins that inhibit the transcription factor E2F until the cell is ready to enter S phase are hyperphosphorylated and inactivated in ESCs leading to continual expression of proliferation genes 9 These changes result in accelerated cycles of cell division Although high expression levels of pro proliferative proteins and a shortened G1 phase have been linked to maintenance of pluripotency 11 12 ESCs grown in serum free 2i conditions do express hypo phosphorylated active Retinoblastoma proteins and have an elongated G1 phase 13 Despite this difference in the cell cycle when compared to ESCs grown in media containing serum these cells have similar pluripotent characteristics 14 Pluripotency factors Oct4 and Nanog play a role in transcriptionally regulating the embryonic stem cell cycle 15 16 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 promise 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 17 In addition to their potential in regenerative medicine embryonic stem cells provide a possible alternative source of tissue organs which serves as a possible solution to the donor shortage dilemma There are some ethical controversies surrounding this though see Ethical debate section below Aside from these uses ESCs can also be used for research on early human development certain genetic disease and in vitro toxicology testing 6 Utilizations EditAccording 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 18 Tissue engineering Edit Embryoid bodies 24 hours after formation In tissue engineering the use of stem cells are known to be of importance In order to successfully engineer a tissue the cells used must be able to perform specific biological functions such as secretion of cytokines signaling molecules interacting with neighboring cells and producing an extracellular matrix in the correct organization Stem cells demonstrates these specific biological functions along with being able to self renew and differentiate into one or more types of specialized cells Embryonic stem cells is one of the sources that are being considered for the use of tissue engineering 19 The use of human embryonic stem cells have opened many new possibilities for tissue engineering however there are many hurdles that must be made before human embryonic stem cell can even be utilized It is theorized that if embryonic stem cells can be altered to not evoke the immune response when implanted into the patient then this would be a revolutionary step in tissue engineering 20 Embryonic stem cells are not limited to tissue engineering Cell replacement therapies Edit Research has focused on differentiating ESCs into a variety of cell types for eventual use as cell replacement therapies Some of the cell types that have or are currently being developed include cardiomyocytes neurons hepatocytes bone marrow cells islet cells and endothelial cells 21 However the derivation of such cell types from ESCs is not without obstacles therefore research has focused on overcoming these barriers For example studies are underway to differentiate ESCs into tissue specific cardiomyocytes and to eradicate their immature properties that distinguish them from adult cardiomyocytes 22 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 23 24 ESCs have been differentiated to natural killer cells and bone tissue 25 Studies involving ESCs are underway to provide an alternative treatment for diabetes For example ESCs have been differentiated into insulin producing cells 26 and researchers at Harvard University were able to produce large quantities of pancreatic beta cells from ESCs 27 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 28 Drug discovery Edit Besides becoming an important alternative to organ transplants ESCs are also being used in the field of toxicology and as cellular screens to uncover new chemical entities 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 21 ESC derived cardiomyocytes have been shown to respond to pharmacological stimuli and hence can be used to assess cardiotoxicity such as torsades de pointes 29 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 research has focused on establishing fully functional ESC derived hepatocytes with stable phase I and II enzyme activity 30 Models of genetic disorder Edit Several new studies have started to address the concept of modeling genetic disorders with embryonic stem cells Either by genetically manipulating the cells or more recently by deriving diseased cell lines identified by prenatal genetic diagnosis PGD modeling genetic disorders is something that has been accomplished with stem cells This approach may very well prove valuable 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 couples who have 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 31 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 32 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 33 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 33 This type of repair depends on the interaction of the two sister chromosomes verification needed 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 34 Rather they undergo programmed cell death apoptosis in response to DNA damage 35 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 36 Consistent with this strategy mouse ES stem cells have a mutation frequency about 100 fold lower than that of isogenic mouse somatic cells 37 Clinical trial Edit Main article Human embryonic stem cells clinical trials On January 23 2009 Phase I clinical trials for transplantation of oligodendrocytes a cell type of the brain and spinal cord derived from human ESCs into spinal cord injured individuals received approval from the U S Food and Drug Administration FDA marking it the world s first human ESC human trial 38 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 ESCs that had been pushed into an oligodendrocytic lineage 39 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 40 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 41 In October 2010 researchers enrolled and administered ESCs to the first patient at Shepherd Center in Atlanta 42 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 43 In 2013 BioTime 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 44 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 45 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 45 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 45 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 45 Concern and controversy EditAdverse effects Edit The major concern with the possible transplantation of ESCs into patients as therapies is their ability to form tumors including teratomas 46 Safety issues prompted the FDA to place a hold on the first ESC clinical trial however no tumors were observed The main strategy to enhance the safety of ESCs for potential clinical use is to differentiate the ESCs into specific cell types e g neurons muscle liver cells that have reduced or eliminated ability to cause tumors Following differentiation the cells are subjected to sorting by flow cytometry for further purification ESCs are predicted to be inherently safer than iPS cells created with genetically integrating viral vectors because they are not genetically modified with genes such as c Myc that are linked to cancer Nonetheless ESCs express very high levels of the iPS inducing genes and these genes including Myc are essential for ESC self renewal and pluripotency 47 and potential strategies to improve safety by eliminating c Myc expression are unlikely to preserve the cells stemness However N myc and L myc have been identified to induce iPS cells instead of c myc with similar efficiency 48 Later protocols to induce pluripotency bypass these problems completely by using non integrating RNA viral vectors such as sendai virus or mRNA transfection Ethical debate Edit Main article Stem cell controversy Due to the nature of embryonic stem cell research there are a lot of controversial opinions on the topic Since harvesting embryonic stem cells usually necessitates destroying the embryo from which those cells are obtained the moral status of the embryo comes into question Some people claim that the 5 day old mass of cells is too young to achieve personhood or that the embryo if donated from an IVF clinic where labs typically acquire embryos would otherwise go to medical waste anyway Opponents of ESC research claim that an embryo is a human life therefore destroying it is murder and the embryo must be protected under the same ethical view as a more developed human being 49 History Edit1964 Lewis Kleinsmith and G Barry Pierce Jr isolated a single type of cell from a teratocarcinoma a tumor now known from a germ cell 50 These cells were isolated from the teratocarcinoma replicated and grew in cell culture as a stem cell and are now known as embryonal carcinoma EC cells citation needed Although similarities in morphology and differentiating potential pluripotency led to the use of EC cells as the in vitro model for early mouse development 51 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 a 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 cell from these embryos 52 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 53 She showed that embryos could be cultured in vitro and that ES cells could be derived from these embryos 1989 Mario R Cappechi Martin J Evans and Oliver Smithies publish their research that details their isolation and genetic modifications of embryonic stem cells creating the first knockout mice 54 In creating knockout mice this publication provided scientists with an entirely new way to study disease Dolly the sheep cell differentiation1996 Dolly was the first mammal cloned from an adult cell by the Roslin Institute of the University of Edinburgh 55 This experiment instituted the proposition that specialized adult cells obtain the genetic makeup to perform a specific task which established a basis for further research within a variety of cloning techniques The Dolly experiment was performed by obtaining the mammalian udder cells from a sheep Dolly and differentiating these cells until division was concluded An egg cell was then procured from a different sheep host and the nucleus was removed An udder cell was placed next to the egg cell and connected by electricity causing this cell to share DNA This egg cell differentiated into an embryo and the embryo was inserted into a third sheep which gave birth to the clone version of Dolly 56 1998 A team from the University of Wisconsin Madison James A Thomson Joseph Itskovitz Eldor Sander S Shapiro Michelle A Waknitz Jennifer J Swiergiel Vivienne S Marshall and Jeffrey M Jones publish a paper titled Embryonic Stem Cell Lines Derived From Human Blastocysts The researchers behind this study not only created the first embryonic stem cells but recognized 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 57 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 58 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 59 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 60 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 61 Techniques and conditions for derivation and culture EditDerivation from humans Edit In vitro fertilization generates multiple embryos The surplus of embryos is not clinically used or is unsuitable for implantation into the patient and therefore may be donated by the donor with consent Human embryonic stem cells can be derived from these donated embryos or additionally they can also be extracted from cloned embryos created using a cell from a patient and a donated egg through the process of somatic cell nuclear transfer 62 The inner cell mass cells of interest from the blastocyst stage of the embryo is separated from the trophectoderm the cells that would differentiate into extra embryonic tissue Immunosurgery the process in which antibodies are bound to the trophectoderm and removed by another solution and mechanical dissection are performed to achieve separation The resulting inner cell mass cells are plated onto cells that will supply support The inner cell mass cells attach and expand further to form a human embryonic cell line which are undifferentiated These cells are fed daily and are enzymatically or mechanically separated every four to seven days For differentiation to occur the human embryonic stem cell line is removed from the supporting cells to form embryoid bodies is co cultured with a serum containing necessary signals or is grafted in a three dimensional scaffold to result 63 Derivation from other animals Edit Embryonic stem cells are derived from the inner cell mass of the early embryo which are harvested from the donor mother animal Martin Evans and Matthew Kaufman reported a technique that delays embryo implantation allowing the inner cell mass to increase This process includes removing the donor mother s ovaries and dosing her with progesterone changing the hormone environment which causes the embryos to remain free in the uterus After 4 6 days of this intrauterine culture the embryos are harvested and grown in in vitro culture until the inner cell mass forms egg cylinder like structures which are dissociated into single cells and plated on fibroblasts treated with mitomycin c to prevent fibroblast mitosis Clonal cell lines are created by growing up a single cell Evans and Kaufman showed that the cells grown out from these cultures could form teratomas and embryoid bodies and differentiate in vitro all of which indicating that the cells are pluripotent 52 Gail Martin derived and cultured her ES cells differently She removed the embryos from the donor mother at approximately 76 hours after copulation and cultured them overnight in a medium containing serum The following day she removed the inner cell mass from the late blastocyst using microsurgery The extracted inner cell mass was cultured on fibroblasts treated with mitomycin c in a medium containing serum and conditioned by ES cells After approximately one week colonies of cells grew out These cells grew in culture and demonstrated pluripotent characteristics as demonstrated by the ability to form teratomas differentiate in vitro and form embryoid bodies Martin referred to these cells as ES cells 53 It is now known that the feeder cells provide leukemia inhibitory factor LIF and serum provides bone morphogenetic proteins BMPs that are necessary to prevent ES cells from differentiating 64 65 These factors are extremely important for the efficiency of deriving ES cells Furthermore it has been demonstrated that different mouse strains have different efficiencies for isolating ES cells 66 Current uses for mouse ES cells include the generation of transgenic mice including knockout mice For human treatment there is a need for patient specific pluripotent cells Generation of human ES cells is more difficult and faces ethical issues So in addition to human ES cell research many groups are focused on the generation of induced pluripotent stem cells iPS cells 67 Potential methods for new cell line derivation Edit On August 23 2006 the online edition of Nature scientific journal published a letter by Dr Robert Lanza medical director of Advanced Cell Technology in Worcester MA stating that his team had found a way to extract embryonic stem cells without destroying the actual embryo 68 This technical achievement would potentially enable scientists to work with new lines of embryonic stem cells derived using public funding in the US where federal funding was at the time limited to research using embryonic stem cell lines derived prior to August 2001 In March 2009 the limitation was lifted 69 Human embryonic stem cells have also been derived by somatic cell nuclear transfer SCNT 70 71 This approach has also sometimes been referred to as therapeutic cloning because SCNT bears similarity to other kinds of cloning in that nuclei are transferred from a somatic cell into an enucleated zygote However in this case SCNT was used to produce embryonic stem cell lines in a lab not living organisms via a pregnancy The therapeutic part of the name is included because of the hope that SCNT produced embryonic stem cells could have clinical utility Induced pluripotent stem cells Edit Main article Induced pluripotent stem cell The iPS cell technology was pioneered by Shinya Yamanaka s lab in Kyoto Japan who showed in 2006 that the introduction of four specific genes encoding transcription factors could convert adult cells into pluripotent stem cells 72 He was awarded the 2012 Nobel Prize along with Sir John Gurdon for the discovery that mature cells can be reprogrammed to become pluripotent 73 In 2007 it was shown that pluripotent stem cells highly similar to embryonic stem cells can be induced by the delivery of four factors Oct3 4 Sox2 c Myc and Klf4 to differentiated cells 74 Utilizing the four genes previously listed the differentiated cells are reprogrammed into pluripotent stem cells allowing for the generation of pluripotent embryonic stem cells without the embryo The morphology and growth factors of these lab induced pluripotent cells are equivalent to embryonic stem cells leading these cells to be known as induced pluripotent stem cells iPS cells 75 This observation was observed in mouse pluripotent stem cells originally but now can be performed in human adult fibroblasts using the same four genes 76 Because ethical concerns regarding embryonic stem cells typically are about their derivation from terminated embryos it is believed that reprogramming to these iPS cells may be less controversial This may enable the generation of patient specific ES cell lines that could potentially be used for cell replacement therapies In addition this will allow the generation of ES cell lines from patients with a variety of genetic diseases and will provide invaluable models to study those diseases However as a first indication that the iPS cell technology can in rapid succession lead to new cures it was used by a research team headed by Rudolf Jaenisch of the Whitehead Institute for Biomedical Research in Cambridge Massachusetts to cure mice of sickle cell anemia as reported by Science journal s online edition on December 6 2007 77 78 On January 16 2008 a California based company Stemagen announced that they had created the first mature cloned human embryos from single skin cells taken from adults These embryos can be harvested for patient matching embryonic stem cells 79 Contamination by reagents used in cell culture Edit The online edition of Nature Medicine published a study on January 24 2005 which stated that the human embryonic stem cells available for federally funded research are contaminated with non human molecules from the culture medium used to grow the cells 80 It is a common technique to use mouse cells and other animal cells to maintain the pluripotency of actively dividing stem cells The problem was discovered when non human sialic acid in the growth medium was found to compromise the potential uses of the embryonic stem cells in humans according to scientists at the University of California San Diego 81 However a study published in the online edition of Lancet Medical Journal on March 8 2005 detailed information about a new stem cell line that was derived from human embryos under completely cell and serum free conditions After more than 6 months of undifferentiated proliferation these cells demonstrated the potential to form derivatives of all three embryonic germ layers both in vitro and in teratomas These properties were also successfully maintained for more than 30 passages with the established stem cell lines 82 Muse cells Edit Main article Muse cell Muse cells Multi lineage differentiating stress enduring cell are non cancerous pluripotent stem cell found in adults 83 84 They were discovered in 2010 by Mari Dezawa and her research group 83 Muse cells reside in the connective tissue of nearly every organ including the umbilical cord bone marrow and peripheral blood 85 83 86 87 88 They are collectable from commercially obtainable mesenchymal cells such as human fibroblasts bone marrow mesenchymal stem cells and adipose derived stem cells 89 90 91 Muse cells are able to generate cells representative of all three germ layers from a single cell both spontaneously and under cytokine induction Expression of pluripotency genes and triploblastic differentiation are self renewable over generations Muse cells do not undergo teratoma formation when transplanted into a host environment in vivo eradicating the risk of tumorigenesis through unbridled cell proliferation 83 See also EditEmbryoid body Embryonic Stem Cell Research Oversight Committees Fetal tissue implant Induced stem cells KOSR KnockOut Serum Replacement Stem cell controversyReferences Edit 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 a b c Stem Cell Basics STEM Cell Information stemcells nih gov Retrieved 5 June 2022 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 Nakaya Andrea C August 1 2011 Biomedical ethics San Diego CA ReferencePoint Press pp 96 ISBN 978 1601521576 Carla A Herberts Marcel SG Kwa Harm PH Hermsen 2011 Risk factors in the development of stem cell therapy Journal of Translational Medicine 9 29 29 doi 10 1186 1479 5876 9 29 PMC 3070641 PMID 21418664 a b 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 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 S2CID 7201396 Martello G Smith A 2014 The nature of embryonic stem cells Annual Review of Cell and Developmental Biology 30 647 75 doi 10 1146 annurev cellbio 100913 013116 PMID 25288119 a b Boward B Wu T Dalton S 2016 Concise Review Control of Cell Fate Through Cell Cycle and Pluripotency Networks Stem Cells 34 6 1427 36 doi 10 1002 stem 2345 PMC 5201256 PMID 26889666 White J Stead E Faast R Conn S Cartwright P Dalton S 2005 Developmental activation of the Rb E2F pathway and establishment of cell cycle regulated cyclin dependent kinase activity during embryonic stem cell differentiation Molecular Biology of the Cell 16 4 2018 27 doi 10 1091 mbc e04 12 1056 PMC 1073679 PMID 15703208 Ter Huurne Menno Stunnenberg Hendrik G 21 April 2021 G1 phase progression in pluripotent stem cells Cellular and Molecular Life Sciences 21 10 4507 4519 doi 10 1007 s00018 021 03797 8 ISSN 1875 9777 PMC 8195903 PMID 33884444 Singh Amar M Dalton Stephen 2009 08 07 The cell cycle and Myc intersect with mechanisms that regulate pluripotency and reprogramming Cell Stem Cell 5 2 141 149 doi 10 1016 j stem 2009 07 003 ISSN 1875 9777 PMC 2909475 PMID 19664987 Ter Huurne Menno Chappell James Dalton Stephen Stunnenberg Hendrik G 5 October 2017 Distinct Cell Cycle Control in Two Different States of Mouse Pluripotency Cell Stem Cell 21 4 449 455 e4 doi 10 1016 j stem 2017 09 004 ISSN 1875 9777 PMC 5658514 PMID 28985526 Ying Qi Long Wray Jason Nichols Jennifer Batlle Morera Laura Doble Bradley Woodgett James Cohen Philip Smith Austin 2008 05 22 The ground state of embryonic stem cell self renewal Nature 453 7194 519 523 Bibcode 2008Natur 453 519Y doi 10 1038 nature06968 ISSN 1476 4687 PMC 5328678 PMID 18497825 Lee J Go Y Kang I Han Y M Kim J 2010 Oct 4 controls cell cycle progression of embryonic stem cells The Biochemical Journal 426 2 171 81 doi 10 1042 BJ20091439 PMC 2825734 PMID 19968627 Zhang X Neganova I Przyborski S Yang C Cooke M Atkinson S P Anyfantis G Fenyk S Keith W N Hoare S F Hughes O Strachan T Stojkovic M Hinds P W Armstrong L Lako M 2009 A role for NANOG in G1 to S transition in human embryonic stem cells through direct binding of CDK6 and CDC25A The Journal of Cell Biology 184 1 67 82 doi 10 1083 jcb 200801009 PMC 2615089 PMID 19139263 Mahla Ranjeet July 19 2016 Stem Cell Applications in Regenerative Medicine and Disease Therapeutics International Journal of Cell Biology 2016 6940283 doi 10 1155 2016 6940283 PMC 4969512 PMID 27516776 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 PMID 11917100 Vats A Tolley N S Bishop A E Polak J M 2005 08 01 Embryonic Stem Cells and Tissue Engineering Delivering Stem Cells to the Clinic Journal of the Royal Society of Medicine 98 8 346 350 doi 10 1177 014107680509800804 ISSN 0141 0768 PMC 1181832 PMID 16055897 Heath Carole A 2000 01 01 Cells for tissue engineering Trends in Biotechnology 18 1 17 19 doi 10 1016 S0167 7799 99 01396 7 ISSN 0167 7799 PMID 10631775 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 2 214 223 doi 10 1093 toxsci kfh100 PMID 15014205 Siu CW Moore JC Li RA 2007 Human embryonic stem cell derived cardiomyocytes for heart therapies Cardiovascular amp Hematological Disorders Drug Targets 7 2 145 152 doi 10 2174 187152907780830851 PMID 17584049 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 PMID 15310843 Parish CL Arenas E 2007 Stem cell based strategies for the treatment of Parkinson s disease Neuro Degenerative Diseases 4 4 339 347 doi 10 1159 000101892 PMID 17627139 S2CID 37229348 Waese EY Kandel RA Stanford WL 2008 Application of stem cells in bone repair Skeletal Radiology 37 7 601 608 doi 10 1007 s00256 007 0438 8 PMID 18193216 S2CID 12401889 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 1401 doi 10 1038 nbt1259 PMID 17053790 S2CID 11040949 Colen B D 9 October 2014 Giant leap against diabetes The Harvard Gazette Retrieved 24 November 2014 Menasche Phillip Vanneaux Valerie 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 10 1093 eurheartj ehu192 PMID 24835485 Jensen J Hyllner J Bjorquist P 2009 Human embryonic stem cell technologies and drug discovery Journal of Cellular Physiology 219 3 513 519 doi 10 1002 jcp 21732 PMID 19277978 S2CID 36354049 Soderdahl T Kuppers Munther B Heins N Edsbagge J Bjorquist P Cotgreave I Jernstrom B 2007 Glutathione transferases in hepatocyte like cells derived from human embryonic stem cells Toxicology in Vitro 21 5 929 937 doi 10 1016 j tiv 2007 01 021 PMID 17346923 Dr Yury Verlinsky 1943 2009 Expert in reproductive technology Archived 2009 08 08 at the Wayback Machine Chicago Tribune July 20 2009 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 2906 doi 10 4161 cc 7 18 6679 PMC 2754209 PMID 18769152 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 1711 doi 10 1089 scd 2010 0058 PMC 3128311 PMID 20446816 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 14448 Bibcode 2004PNAS 10114443H doi 10 1073 pnas 0401346101 PMC 521944 PMID 15452351 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 3 145 155 doi 10 1016 S0960 9822 98 70061 2 PMID 9443911 S2CID 13938854 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 2 145 178 doi 10 1016 S1383 5742 02 00009 1 PMID 12052432 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 3590 Bibcode 2002PNAS 99 3586C doi 10 1073 pnas 062527199 PMC 122567 PMID 11891338 FDA approves human embryonic stem cell study CNN com January 23 2009 Retrieved May 1 2010 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 PDF J Neurosci 25 19 4694 4705 doi 10 1523 JNEUROSCI 0311 05 2005 PMC 6724772 PMID 15888645 Reinberg Steven 2009 01 23 FDA OKs 1st Embryonic Stem Cell Trial The Washington Post Geron comments on FDA hold on spinal cord injury trial geron com August 27 2009 Vergano Dan 11 October 2010 Embryonic stem cells used on patient for first time USA Today Retrieved 12 October 2010 Brown Eryn November 15 2011 Geron exits stem cell research LA Times Retrieved 2011 11 15 Great news BioTime Subsidiary Asterias Acquires Geron Embryonic Stem Cell Program iPScell com October 1 2013 a b c d California Institute of Regenerative Medicine Archived 2017 10 24 at the Wayback Machine BioTime Inc Knoepfler Paul S 2009 Deconstructing Stem Cell Tumorigenicity A Roadmap to Safe Regenerative Medicine Stem Cells 27 5 1050 1056 doi 10 1002 stem 37 PMC 2733374 PMID 19415771 Varlakhanova Natalia V Cotterman Rebecca F Devries Wilhelmine N Morgan Judy Donahue Leah Rae Murray Stephen Knowles Barbara B Knoepfler Paul S 2010 Myc maintains embryonic stem cell pluripotency and self renewal Differentiation 80 1 9 19 doi 10 1016 j diff 2010 05 001 PMC 2916696 PMID 20537458 Wernig Marius Meissner Alexander Cassady John P Jaenisch Rudolf 2008 C Myc is Dispensable for Direct Reprogramming of Mouse Fibroblasts Cell Stem Cell 2 1 10 12 doi 10 1016 j stem 2007 12 001 PMID 18371415 King Nancy Perrin Jacob July 7 2014 Ethical issues in stem cell research and therapy Stem Cell Research amp Therapy 5 4 85 doi 10 1186 scrt474 PMC 4097842 PMID 25157428 Kleinsmith LJ Pierce GB Jr 1964 Multipotentiality of Single Embryoncal Carcinoma Cells Cancer Res 24 1544 1551 PMID 14234000 Martin GR 1980 Teratocarcinomas and mammalian embryogenesis Science 209 4458 768 776 Bibcode 1980Sci 209 768M doi 10 1126 science 6250214 PMID 6250214 a b Evans M Kaufman M 1981 Establishment in culture of pluripotent cells from mouse embryos Nature 292 5819 154 156 Bibcode 1981Natur 292 154E doi 10 1038 292154a0 PMID 7242681 S2CID 4256553 a b 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 7638 Bibcode 1981PNAS 78 7634M doi 10 1073 pnas 78 12 7634 PMC 349323 PMID 6950406 The 2007 Nobel Prize in Physiology or Medicine Advanced Information Nobel Prize Nobel Media The Life of Dolly Dolly the Sheep Retrieved 2022 02 21 Klotzko Arlene Judith Klotzko Visiting Scholar Royal Free and University College Medical School Arlene Judith 2006 A Clone of Your Own Cambridge University Press ISBN 978 0 521 85294 4 Thompson James A Itskovitz Eldor Joseph Shapiro Sander S Waknitz Michelle A Swiergiel Jennifer J Marshall Vivienne S Jones Jeffrey M 6 November 1998 Embryonic Stem Cell Lines Derived From Human Blastocyst Science 282 5391 1145 1147 Bibcode 1998Sci 282 1145T doi 10 1126 science 282 5391 1145 PMID 9804556 President George W Bush s address on stem cell research CNN Inside Politics Aug 9 2001 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 10 1016 j cell 2006 07 024 hdl 2433 159777 PMID 16904174 S2CID 1565219 Wadman Meredith 27 January 2009 Stem cells ready for primetime Nature 457 7229 516 doi 10 1038 457516a PMID 19177087 Executive Order 13505 Removing Barriers To Responsible Scientific Research Involving Human Stem Cells PDF Federal Register Presidential Documents 74 46 11 March 2009 Mountford JC 2008 Human embryonic stem cells origins characteristics and potential for regenerative therapy Transfus Med 18 1 1 12 doi 10 1111 j 1365 3148 2007 00807 x PMID 18279188 S2CID 20874633 Thomson JA Itskovitz Eldor J Shapiro SS Waknitz MA Swiergiel JJ Marshall VS Jones JM 1998 Embryonic stem cell lines derived from human blastocysts Science 282 5391 1145 1147 Bibcode 1998Sci 282 1145T doi 10 1126 science 282 5391 1145 PMID 9804556 Smith AG Heath JK Donaldson DD Wong GG Moreau J Stahl M Rogers D 1988 Inhibition of pluripotential embryonic stem cell differentiation by purified polypeptides Nature 336 6200 688 690 Bibcode 1988Natur 336 688S doi 10 1038 336688a0 PMID 3143917 S2CID 4325137 Williams RL Hilton DJ Pease S Willson TA Stewart CL Gearing DP Wagner EF Metcalf D Nicola NA Gough NM 1988 Myeloid leukaemia inhibitory factor maintains the developmental potential of embryonic stem cells Nature 336 6200 684 687 Bibcode 1988Natur 336 684W doi 10 1038 336684a0 PMID 3143916 S2CID 4346252 Ledermann B Burki K 1991 Establishment of a germ line competent C57BL 6 embryonic stem cell line Exp Cell Res 197 2 254 258 doi 10 1016 0014 4827 91 90430 3 PMID 1959560 Takahashi K Tanabe K Ohnuki M Narita M Ichisaka T Tomoda K Yamanaka S 2007 Induction of pluripotent stem cells from adult human fibroblasts by defined factors Cell 131 5 861 872 doi 10 1016 j cell 2007 11 019 hdl 2433 49782 PMID 18035408 S2CID 8531539 Klimanskaya I Chung Y Becker S Lu SJ Lanza R 2006 Human embryonic stem cell lines derived from single blastomeres Nature 444 7118 481 485 Bibcode 2006Natur 444 481K doi 10 1038 nature05142 PMID 16929302 S2CID 84792371 US scientists relieved as Obama lifts ban on stem cell research The Guardian 10 March 2009 Tachibana Masahito Amato Paula Sparman Michelle Gutierrez Nuria Marti Tippner Hedges Rebecca Ma Hong Kang Eunju Fulati Alimujiang Lee Hyo Sang Sritanaudomchai Hathaitip Masterson Keith 2013 06 06 Human Embryonic Stem Cells Derived by Somatic Cell Nuclear Transfer Cell 153 6 1228 1238 doi 10 1016 j cell 2013 05 006 ISSN 0092 8674 PMC 3772789 PMID 23683578 Chung Young Gie Eum Jin Hee Lee Jeoung Eun Shim Sung Han Sepilian Vicken Hong Seung Wook Lee Yumie Treff Nathan R Choi Young Ho Kimbrel Erin A Dittman Ralph E 2014 06 05 Human Somatic Cell Nuclear Transfer Using Adult Cells Cell Stem Cell 14 6 777 780 doi 10 1016 j stem 2014 03 015 ISSN 1934 5909 PMID 24746675 Takahashi K Yamanaka S 2006 Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors Cell 126 4 663 676 doi 10 1016 j cell 2006 07 024 hdl 2433 159777 PMID 16904174 S2CID 1565219 The Nobel Prize in Physiology or Medicine 2012 Press Release Nobel Media AB 8 October 2012 Wernig Marius Meissner Alexander Foreman Ruth Brambrink Tobias Ku Manching Hochedlinger Konrad Bernstein Bradley E Jaenisch Rudolf 2007 07 19 In vitro reprogramming of fibroblasts into a pluripotent ES cell like state Nature 448 7151 318 324 Bibcode 2007Natur 448 318W doi 10 1038 nature05944 ISSN 1476 4687 PMID 17554336 S2CID 4377572 Takahashi Kazutoshi Yamanaka Shinya 2006 08 25 Induction of Pluripotent Stem Cells from Mouse Embryonic and Adult Fibroblast Cultures by Defined Factors Cell 126 4 663 676 doi 10 1016 j cell 2006 07 024 ISSN 0092 8674 PMID 16904174 S2CID 1565219 Takahashi Kazutoshi Tanabe Koji Ohnuki Mari Narita Megumi Ichisaka Tomoko Tomoda Kiichiro Yamanaka Shinya 2007 11 30 Induction of Pluripotent Stem Cells from Adult Human Fibroblasts by Defined Factors Cell 131 5 861 872 doi 10 1016 j cell 2007 11 019 ISSN 0092 8674 PMID 18035408 S2CID 8531539 Weiss Rick 2007 12 07 Scientists Cure Mice Of Sickle Cell Using Stem Cell Technique New Approach Is From Skin Not Embryos The Washington Post pp A02 Hanna J Wernig M Markoulaki S Sun C W Meissner A Cassady J P Beard C Brambrink T Wu L C Townes T M Jaenisch R 2007 Treatment of Sickle Cell Anemia Mouse Model with iPS Cells Generated from Autologous Skin Science 318 5858 1920 1923 Bibcode 2007Sci 318 1920H doi 10 1126 science 1152092 PMID 18063756 S2CID 657569 Helen Briggs 2008 01 17 US team makes embryo clone of men BBC pp A01 Ebert Jessica 24 January 2005 Human stem cells trigger immune attack Nature News London Nature Publishing Group doi 10 1038 news050124 1 Archived from the original on 2010 09 24 Retrieved 2009 02 27 Martin MJ Muotri A Gage F Varki A 2005 Human embryonic stem cells express an immunogenic nonhuman sialic acid Nat Med 11 2 228 232 doi 10 1038 nm1181 PMID 15685172 S2CID 13739919 Klimanskaya I Chung Y Meisner L Johnson J West MD Lanza R 2005 Human embryonic stem cells derived without feeder cells Lancet 365 9471 1636 1641 doi 10 1016 S0140 6736 05 66473 2 PMID 15885296 S2CID 17139951 a b c d Kuroda Y Kitada M Wakao S Nishikawa K Tanimura Y Makinoshima H Goda M Akashi H Inutsuka A Niwa A Shigemoto T Nabeshima Y Nakahata T Nabeshima Y i Fujiyoshi Y Dezawa M 2010 Unique multipotent cells in adult human mesenchymal cell populations Proceedings of the National Academy of Sciences 107 19 8639 8643 Bibcode 2010PNAS 107 8639K doi 10 1073 pnas 0911647107 PMC 2889306 PMID 20421459 Muse Cells SpringerLink Zikuan Leng 1 2 Dongming Sun 2 Zihao Huang 3 Iman Tadmori 2 Ning Chiang 2 Nikhit Kethidi 2 Ahmed Sabra 2 Yoshihiro Kushida 4 Yu Show Fu 3 Mari Dezawa 4 Xijing He 1 Wise Young 2Quantitative Analysis of SSEA3 Cells from Human Umbilical Cord after Magnetic SortingCell Transplant 2019 Jul 28 7 907 923 Wakao S Kitada M Kuroda Y Shigemoto T Matsuse D Akashi H Tanimura Y Tsuchiyama K Kikuchi T Goda M Nakahata T Fujiyoshi Y Dezawa M 2011 Multilineage differentiating stress enduring Muse cells are a primary source of induced pluripotent stem cells in human fibroblasts Proceedings of the National Academy of Sciences 108 24 9875 9880 Bibcode 2011PNAS 108 9875W doi 10 1073 pnas 1100816108 PMC 3116385 PMID 21628574 Dezawa Mari 2016 Muse Cells Provide the Pluripotency of Mesenchymal Stem Cells Direct Contribution of Muse Cells to Tissue Regeneration Cell Transplantation 25 5 849 861 doi 10 3727 096368916X690881 PMID 26884346 Hori Emiko Hayakawa Yumiko Hayashi Tomohide Hori Satoshi Okamoto Soushi Shibata Takashi Kubo Michiya Horie Yukio Sasahara Masakiyo Kuroda Satoshi 2016 Mobilization of Pluripotent Multilineage Differentiating Stress Enduring Cells in Ischemic Stroke Journal of Stroke and Cerebrovascular Diseases 25 6 1473 1481 doi 10 1016 j jstrokecerebrovasdis 2015 12 033 PMID 27019988 Kuroda Yasumasa Wakao Shohei Kitada Masaaki Murakami Toru Nojima Makoto Dezawa Mari 2013 Isolation culture and evaluation of multilineage differentiating stress enduring Muse cells Nature Protocols 8 7 1391 1415 doi 10 1038 nprot 2013 076 PMID 23787896 S2CID 28597290 unreliable medical source Ogura Fumitaka Wakao Shohei Kuroda Yasumasa Tsuchiyama Kenichiro Bagheri Mozhdeh Heneidi Saleh Chazenbalk Gregorio Aiba Setsuya Dezawa Mari 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 and Development 23 7 717 728 doi 10 1089 scd 2013 0473 PMID 24256547 Heneidi Saleh Simerman Ariel A Keller Erica Singh Prapti Li Xinmin Dumesic Daniel A Chazenbalk Gregorio 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 Bibcode 2013PLoSO 864752H doi 10 1371 journal pone 0064752 PMC 3673968 PMID 23755141 External links Edit Wikimedia Commons has media related to Embryonic stem cells Understanding Stem Cells A View of the Science and Issues from the National Academies National Institutes of Health University of Oxford practical workshop on pluripotent stem cell technology Archived 2016 04 08 at the Wayback Machine Fact sheet on embryonic stem cells Fact sheet on ethical issues in embryonic stem cell research Information amp Alternatives to Embryonic Stem Cell Research A blog focusing specifically on ES cells and iPS cells including research biotech and patient oriented issues Portals Biology Medicine Retrieved from https en wikipedia org w index php title Embryonic stem cell amp oldid 1118859889, wikipedia, wiki, book, books, library,

article

, read, download, free, free download, mp3, video, mp4, 3gp, jpg, jpeg, gif, png, picture, music, song, movie, book, game, games.