Page 20«..10..19202122..»

Okyanos Heart Institute CEO Matt Feshbach Congratulates Japan’s Legislators On Stem Cell Bill And Global Regulatory …

By daniellenierenberg

FREEPORT, The Bahamas (PRWEB) December 06, 2013

December 6, 2013 Matt Feshbach, CEO of Okyanos Heart Institute whose mission it is to bring a new standard of care and better quality of life to patients with coronary artery disease using cardiac stem cell therapy, acknowledges the Japanese legislature for its recent approval of a bill aimed at the treatment of certain chronic diseases using regenerative medicine strategies.

The legislation was passed in Japan on November 20th, 2013. The new regenerative medicine law emphasizes the importance of establishing patient safety in the use of adult stem cell therapies prior to being offered commercially. It also serves to support innovation in stem cell and regenerative medicine therapies by providing a framework by which such technologies may be granted new, limited approval paths for some biologics.

Japan has taken a leadership position globally for its passage of enlightened legislation for stem cell therapy, said Feshbach, who recognizes this development as an important milestone in its potential to benefit patients and the field of healthcare.

We applaud Japan as well as other countries including but not limited to Australia, Singapore, and New Zealand for approving stem cell processing devices and/or biologics (such as stem cells) for use in clinics today, he added. This legislation in Japan says that if a stem cell therapy protocol can demonstrate a strong safety profile, physicians have the option to offer it to patients, generally when other standard-of-care interventions have not proven effective and the patients have no other options available to them. Patients will have the choice to use their own stem cells to treat the condition. By tracking the progress of the patients over time, efficacy can be determined and the treatment may become another standard-of-care treatment option available to patients.

While this research is important over the long term, adult stem cell therapy is unique in that it takes advantage of the natural mechanisms of a persons own stem cells to repair the cells, tissues or organs damaged by disease or injury, stated Feshbach. The dawn of a new phase in the evolution of medicine has begun.

Additional countries such as The Bahamas, Panama, Argentina and Jordan have established regulations and legislation designed to both protect patient safety and give access to treatments which have the potential to help unmet needs such as heart failure and other diseases.

Japan represents the second-largest medical market in the world and remains a global leader in both adult stem cell and gene therapy trials. Dr. Shinya Yamanaka, professor and director for the Center for iPS Cell Research and Application (CiRA) at Kyoto University, was awarded a Nobel Prize in 2012 for the discovery of induced pluripotent stem cells (iPS). Click here to read more about the Japanese legislatures recent stem cell measures.

About Okyanos Heart Institute: (Oh key AH nos) Based in Freeport, The Bahamas, Okyanos Heart Institutes mission is to bring a new standard of care and a better quality of life to patients with coronary artery disease using cardiac stem cell therapy. Okyanos adheres to U.S. surgical center standards and is led by Chief Medical Officer Howard T. Walpole Jr., M.D., M.B.A., F.A.C.C., F.S.C.A.I. Okyanos Treatment utilizes a unique blend of stem and regenerative cells derived from ones own adipose (fat) tissue. The cells, when placed into the heart via a minimally-invasive catheterization, stimulate the growth of new blood vessels, a process known as angiogenesis. The treatment facilitates blood flow in the heart and supports intake and use of oxygen (as demonstrated in rigorous clinical trials such as the PRECISE trial). The literary name Okyanos (Oceanos) symbolizes flow. For more information, go to http://www.okyanos.com

View original post here:
Okyanos Heart Institute CEO Matt Feshbach Congratulates Japan’s Legislators On Stem Cell Bill And Global Regulatory ...

To Read More: Okyanos Heart Institute CEO Matt Feshbach Congratulates Japan’s Legislators On Stem Cell Bill And Global Regulatory …
categoriaIPS Cell Therapy commentoComments Off on Okyanos Heart Institute CEO Matt Feshbach Congratulates Japan’s Legislators On Stem Cell Bill And Global Regulatory … | dataDecember 13th, 2013
Read All

‘Something positive for humankind’: Girls lend cells to genetic study

By raymumme

NBC News - At 15, Hayley Mogul lacks the fine motor skills needed to write. Her sister Bari is 9 and still eating baby food.

There's no cure for their rare disorders, caused by unique genetic mutations. But for once, there's an advantage to having conditions so rare that drug companies cannot even think of looking for a cure. The sisters are taking part in a whole new kind of experiment in which scientists are literally turning back the clock on their cells.

They're using an experimental technique to transform the cells into embryonic form, and then growing these baby cells in lab dishes.

The goal is the get the cells to misfire in the lab in just the same way they are in Hayley's and Bari's bodies. It's a new marriage of genetics and stem cell research, and represents one of the most promising applications of so-called pluripotent stem cells.

"One day these two girls will probably change the face of medicine as we know it," said their father, Steven Mogul.

Steven and Robyn Mogul don't understand why both their daughters ended up with the rare mutations, which cause a range of neurological and metabolic problems.

"We have been tested," said Mogul, a 45-year-old wealth manager living in Chicago. "We don't have any mutations, and there are no developmental issues. We have no idea how it happened. "

The girls need special schooling and physical therapy. They must wear diapers, and when they get a cold or the flu, they can develop dangerously low blood sugar. "When the kids get sick, get colds or flu, we have to get them to the hospital," Mogul said.

Hayley, 15, has a mutation in a gene called RAI1, which can cause Smith-Magenis syndrome. The syndrome affects 1 in 25,000 people and can disturb sleep patterns, cause obesity and behavioral issues. But Hayley's mutation is unique and puzzling. Bari, 9, has an RAI1 mutation and a similarly unique mutation in the GRIN2B gene, which can cause learning disabilities.

"Bari doesn't talk," Mogul said. "She walks around, she gets around and lets you know what she wants. She is eating baby food and she is drinking from bottles."

See the original post here:
'Something positive for humankind': Girls lend cells to genetic study

To Read More: ‘Something positive for humankind’: Girls lend cells to genetic study
categoriaIPS Cell Therapy commentoComments Off on ‘Something positive for humankind’: Girls lend cells to genetic study | dataDecember 13th, 2013
Read All

Induced Pluripotent Stem Cells (iPS) from Human Skin: Probable …

By daniellenierenberg

Introduction

The announcement of the ability to produce embryonic cell-like lines from ordinary skin cells has the news media scrambling to get feedback about the possible efficacy of such lines in stem cell therapies. Many politicians have landed on one side or the other, with liberals saying that embryonic stem cell research is still necessary1 and conservatives claiming that all embryonic research should be halted. The marketplace of science will eventually weigh-in on which method(s) are used in real therapies.

Embryonic stem cell (ESC) research has been a hot topic, with conservatives saying that such research is morally unacceptable and liberals saying that conservatives value a clump of cells more than people who have serious disabling diseases. Several groups of medical researchers (including James Thomson, the first person to culture ESC) recently showed that normal skin cells can be reprogrammed to an embryonic state, producing what are now called induced pluripotent stem (iPS) cells. Originally performed in mice in June, 2007,2 researchers took four genes OCT3/4, SOX2, KLF4, and c-MYC and incorporated those genes into the nucleus of cells to induce pluripotency. Such lines could be expanded indefinitely and could differentiate to form numerous kinds of different tissues.

Just five months after the mouse study was published, the feat was repeated by three separate laboratories using human skin cells.3 One research group used the same genes as those used in the mouse study, whereas a second group used OCT3, SOX2, NANOG and LIN28. The techniques were efficient enough to generate one cell line for every 5-10 thousand cells treated. Although not extremely efficient, it is quite usable, since it is possible to obtain hundreds of thousands to millions of cells to carry out these kinds of studies. The technique was recently replicated for adult human skin cells,4 instead of skin cell lines, demonstrating that it could be used to generate patient-specific cell lines.

Studies using iPS cell lines have shown that those cells undergo similar changes compared to what is observed with embryonic stem cells. Cell populations grew at the same rate, telomerase (which preserves the ends of chromosomes) was present in both iPS and ESC. Severalgenes that are silenced in fibroblasts, but active in ESC, were also active in the iPS cells. The iPS cell lines could be differentiated into heart muscle and neuronal cells, in addition to basic cell types (ectoderm, mesoderm, and endoderm). Gene expression assays showed that 5,000 genes from iPS cells showed a five-fold difference in expression compared to those in fibroblasts, although 1,267 genes had a five-fold difference in expression between ESC and iPS cells. According to the James Thomson study, "The human iPS cells described here meet the defining criteria we originally proposed for human ES cells (14), with the significant exception that the iPS cells are not derived from embryos."3

Originally, the new technique is not without its own set of problems, although within two years, virtually all had been resolved. One of the original genes used for reprogramming (c-MYC) has been shown to produce tumors and cancers. Obviously, it would not be a good choice for patient therapy. However, this gene was eliminated in some of the later techniques.5 The second problem was that the genes were originally introduced through the use of a retrovirus that incorporates into the host cell DNA. Depending upon where the gene sequence inserts, it may cause trouble (including mutations and cancers). Those who watched the I am Legend movie will remember that a retrovirus-derived cancer treatment was responsible for turning the surviving members of the human race into an army of grotesque monsters. Although such a transformation is not possible, the initiation of cancer in even a small number of treated patients would make such treatments unusable for human therapy. Two years later the problem of using a retroviral system for reprogramming was solved by switching to a simple lentivirus reprogramming system.6 Within weeks, other researchers went a step further, eliminating viral reprogramming altogether by using reprogramming genes (OCT4, SOX2, NANOG, LIN28, c-Myc, and KLF4) cloned into a circular piece of DNA called a plasmid.7 Subsequent culture of of the iPS over a period of weeks resulted in the complete loss of the plasmid, but with continued pluripotency. The potential of iPS cells is so great that the researcher who first grew ESC in culture is now one of the leading proponents of iPS stem cell research.

A more recent, but somewhat uncertain potential problem has been identified more recently. Since iPS cells are derived from adult tissues, they tend to harbor some of the same epigenetic profiles as those adult tissues from which they are derived. As cells age or differentiate, certain genes are turned on or off through methylation of those gene's promoters. The process prevents those cells from undergoing additional changes that might cause the cells to lose their differentiated properties. When adults cells are induced to pluripotency, some of those epigenetic profiles are retained in the iPS cells.8 How will these vestiges of adult cells affect iPS ability to differentiate into cells that are useful for disease models or therapy? At this point, we don't know for sure. However, my guess is that different ESC lines will exhibit different epigenetic profiles, as will specific isolates of iPS cells. Although researchers have found no problems in producing differentiated iPS lines, some of these epigenetic changes might interfere with the ultimate function of these cells as differentiated cell lines.

Even with these issues, research institutes are beginning to focus their stem cell research on iPS cells. Cedars-Sinai Medical Center recently opened its Induced Pluripotent Stem Cell Core Production Facility in late 2011, according to their press release.9

Induction of pluripotency to produce embryonic-like stem cells is the hot topic in stem cell research. The fact that human iPS cells have been produced in many different laboratories after the initial animal studies shows that the technique is robust and easily reproducible. In contrast, the competing technique, human somatic cell nuclear transfer (cloning), has never been transferred from animal studies to human application, despite years of attempts. At this point, it seems pretty certain that the iPS technique will soon replace ESC as the preferred means of generating human stem cell lines. However, the disadvantage of iPS cells is that the cell lines produced would be patient specific (only useful for the intended patient), whereas the establishment of ESC lines allows biotech companies to patent the lines in order to make lots of money.

http://www.godandscience.org/doctrine/reprogrammed_stem_cells.html Last Modified October 6, 2011

Link:
Induced Pluripotent Stem Cells (iPS) from Human Skin: Probable ...

To Read More: Induced Pluripotent Stem Cells (iPS) from Human Skin: Probable …
categoriaIPS Cell Therapy commentoComments Off on Induced Pluripotent Stem Cells (iPS) from Human Skin: Probable … | dataDecember 6th, 2013
Read All

Stem Cell Quick Reference – Learn Genetics

By JoanneRUSSELL25

Somatic stem cells (also called adult stem cells) exist naturally in the body. They are important for growth, healing, and replacing cells that are lost through daily wear and tear.

Potential as therapy Stem cells from the blood and bone marrow are routinely used as a treatment for blood-related diseases. However, under natural circumstances somatic stem cells can become only a subset of related cell types. Bone marrow stem cells, for example, differentiate primarily into blood cells. This partial differentiation can be an advantage when you want to produce blood cells; but it is a disadvantage if you're interested in producing an unrelated cell type.

Special considerations Most types of somatic stem cells are present in low abundance and are difficult to isolate and grow in culture. Isolation of some types could cause considerable tissue or organ damage, as in the heart or brain. Somatic stem cells can be transplanted from donor to patient, but without drugs that suppress the immune system, a patient's immune system will recognize transplanted cells as foreign and attack them.

Ethical considerations Therapy involving somatic stem cells is not controversial; however, it is subject to the same ethical considerations that apply to all medical procedures.

View post:
Stem Cell Quick Reference - Learn Genetics

To Read More: Stem Cell Quick Reference – Learn Genetics
categoriaIPS Cell Therapy commentoComments Off on Stem Cell Quick Reference – Learn Genetics | dataDecember 6th, 2013
Read All

Biomanufacturing center takes central role in developing stem …

By NEVAGiles23

Oct. 17, 2013

A Waisman Biomanufacturing specialist examines cells in a culture in the cell therapy clean room. The UW-Madison Waisman Center opened Waisman Biomanufacturing to ease the research and development of biological products and drugs.

Photo: Waisman Biomanufacturing

Developing a new drug takes enormous amounts of time, money and skill, but the bar is even higher for a promising stem-cell therapy. Many types of cells derived from these ultra-flexible parent cells are moving toward the market, but the very quality that makes stem cells so valuable also makes them a difficult source of therapeutics.

"The ability to form many types of specialized cells is at the essence of why we are so interested in stem cells, but this pluripotency is not always good," says Derek Hei, director of Waisman Biomanufacturing, a facility in the Waisman Center at UW-Madison.

"The cells we can make from stem cells cells for the heart, brain and liver have amazing potential, but you can also end up with the wrong type of cell. If the cells are not fully differentiated, they can end up differentiating into the wrong cell type," Hei says.

Derek Hei

Just like drugs, stem cells for clinical trials must be produced under a demanding regulatory regime called "good manufacturing practice," he says. That capacity is rare in labs in private business and universities, and this is the only one at UWMadison.

Read the rest here:
Biomanufacturing center takes central role in developing stem ...

To Read More: Biomanufacturing center takes central role in developing stem …
categoriaIPS Cell Therapy commentoComments Off on Biomanufacturing center takes central role in developing stem … | dataDecember 4th, 2013
Read All

Combining Stem Cell Therapy with Gene Therapy | Boston …

By LizaAVILA

When pluripotent stem cells are made from a patients own cells, it may be also be possible to replace the faulty gene that caused their disease with a normal, healthy copy. The repaired stem cells could then be directed to form the tissue type needed, introduced into the body, allowed to divide, and used to reconstitute the diseased tissue. It's a treatment that should last a lifetime.

Boston Childrens Hospital researcher George Q. Daley, MD, PhD, then at the Whitehead Institute, was the first to demonstrate, in 2002, that pluripotent stem cells could successfully treat a disease. Working with mice that possess a genetic defect caused by an immune deficiency, the research team created genetically-matched embryonic stem cells through nuclear transfer, introduced corrective genes, then derived healthy blood stem cells and infused them into the mice, partially restoring their immune function. Daley, Director of Stem Cell Transplantation at Childrens, would like to do the same for his patients with blood diseases.

See the rest here:
Combining Stem Cell Therapy with Gene Therapy | Boston ...

To Read More: Combining Stem Cell Therapy with Gene Therapy | Boston …
categoriaIPS Cell Therapy commentoComments Off on Combining Stem Cell Therapy with Gene Therapy | Boston … | dataNovember 25th, 2013
Read All

Stem Cell Therapy for Neuromuscular Diseases | InTechOpen

By NEVAGiles23

1. Introduction

Neuromuscular disease is a very broad term that encompasses many diseases and aliments that either directly, via intrinsic muscle pathology, or indirectly, via nerve pathology, impair the functioning of the muscles. Neuromuscular diseases affect the muscles and/or their nervous control and lead to problems with movement. Many are genetic; sometimes, an immune system disorder can cause them. As they have no cure, the aim of clinical treatment is to improve symptoms, increase mobility and lengthen life. Some of them affect the anterior horn cell, and are classified as acquired (e.g. poliomyelitis) and hereditary (e.g. spinal muscular atrophy) diseases. SMA is a genetic disease that attacks nerve cells, called motor neurons, in the spinal cord. As a consequence of the lost of the neurons, muscles weakness becomes to be evident, affecting walking, crawling, breathing, swallowing and head and neck control. Neuropathies affect the peripheral nerve and are divided into demyelinating (e.g. leucodystrophies) and axonal (e.g. porphyria) diseases. Charcot-Marie-Tooth (CMT) is the most frequent hereditary form among the neuropathies and its characterized by a wide range of symptoms so that CMT-1a is classified as demyelinating and CMT-2 as axonal (Marchesi & Pareyson, 2010). Defects in neuromuscular junctions cause infantile and non-infantile Botulism and Myasthenia Gravis (MG). MG is a antibody-mediated autoimmune disorder of the neuromuscular junction (NMJ) (Drachman, 1994; Meriggioli & Sanders, 2009). In most cases, it is caused by pathogenic autoantibodies directed towards the skeletal muscle acetylcholine receptor (AChR) (Patrick & Lindstrom, 1973) while in others, non-AChR components of the postsynaptic muscle endplate, such as the muscle-specific receptor tyrosine kinase (MUSK), might serve as targets for the autoimmune attack (Hoch et al., 2001). Although the precise origin of the autoimmune response in MG is not known, genetic predisposition and abnormalities of the thymus gland such as hyperplasia and neoplasia could have an important role in the onset of the disease (Berrih et al., 1984; Roxanis et al., 2001).

Several diseases affect muscles: they are classified as acquired (e.g. dermatomyositis and polymyositis) and hereditary (e.g. myotonic disorders and myopaties) forms. Among the myopaties, muscular dystrophies are characterized by the primary wasting of skeletal muscle, caused by mutations in the proteins that form the link between the cytoskeleton and the basal lamina (Cossu & Sampaolesi, 2007). Mutations in the dystrophin gene cause severe form of hereditary muscular diseases; the most common are Duchenne Muscular Dystrophy (DMD) and Becker Muscular Dystrophy (BMD). DMD patients suffer for complete lack of dystrophin that causes progressive degeneration, muscle wasting and death into the second/third decade of life. Beside, BMD patients show a very mild phenotype, often asymptomatic primarily due to the expression of shorter dystrophin mRNA transcripts that maintain the coding reading frame. DMD patients muscles show absence of dystrophin and presence of endomysial fibrosis, small fibers rounded and muscle fiber degeneration/regeneration. Untreated, boys with DMD become progressively weak during their childhood and stop ambulation at a mean age of 9 years, later with corticosteroid treatment (12/13 yrs). Proximal weakness affects symmetrically the lower (such as quadriceps and gluteus) before the upper extremities, with progression to the point of wheelchair dependence. Eventually distal lower and then upper limb weakness occurs. Weakness of neck flexors is often present at the beginning, and most patients with DMD have never been able to jump. Wrist and hand muscles are involved later, allowing the patients to keep their autonomy in transfers using a joystick to guide their wheelchair. Musculoskeletal contractures (ankle, knees and hips) and learning difficulties can complicate the clinical expression of the disease. Besides this weakness distribution in the same patient, a deep variability among patients does exist. They could express a mild phenotype, between Becker and Duchenne dystrophy, or a really severe form, with the loss of deambulation at 7-8 years. Confinement to a wheelchair is followed by the development of scoliosis, respiratory failure and cardiomyopathy. In 90% of people death is directly related to chronic respiratory insufficiency (Rideau et al., 1983). The identification and characterization of dystrophin gene led to the development of potential treatments for this disorder (Bertoni, 2008). Even if only corticosteroids were proven to be effective on DMD patient (Hyser and Mendell, 1988), different therapeutic approaches were attempted, as described in detail below (see section 7).

The identification and characterization of the genes whose mutations caused the most common neuromuscular diseases led to the development of potential treatments for those disorders. Gene therapy for neuromuscular disorders embraced several concepts, including replacing and repairing a defective gene or modifying or enhancing cellular performance, using gene that is not directly related to the underlying defect (Shavlakadze et al., 2004). As an example, the finding that DMD pathology was caused by mutations in the dystrophin gene allowed the rising of different therapeutic approaches including growth-modulating agents that increase muscle regeneration and delay muscle fibrosis (Tinsley et al., 1998), powerful antisense oligonucleotides with exon-skipping capacity (Mc Clorey et al., 2006), anti-inflammatory or second-messenger signal-modulating agents that affect immune responses (Biggar et al., 2006), agents designed to suppress stop codon mutations (Hamed, 2006). Viral and non-viral vectors were used to deliver the full-length - or restricted versions - of the dystrophin gene into stem cells; alternatively, specific antisense oligonucleotides were designed to mask the putative splicing sites of exons in the mutated region of the primary RNA transcript whose removal would re-establish a correct reading frame. In parallel, the biology of stem cells and their role in regeneration were the subject of intensive and extensive research in many laboratories around the world because of the promise of stem cells as therapeutic agents to regenerate tissues damaged by disease or injury (Fuchs and Segre, 2000; Weissman, 2000). This research constituted a significant part of the rapidly developing field of regenerative biology and medicine, and the combination of gene and cell therapy arose as one of the most suitable possibility to treat degenerative disorders. Several works were published in which stem cell were genetically modified by ex vivo introduction of corrective genes and then transplanted in donor dystrophic animal models.

Stem cells received much attention because of their potential use in cell-based therapies for human disease such as leukaemia (Owonikoko et al., 2007), Parkinsons disease (Singh et al., 2007), and neuromuscular disorders (Endo, 2007; Nowak and Davies, 2004). The main advantage of stem cells rather than the other cells of the body is that they can replenish their numbers for long periods through cell division and, they can produce a progeny that can differentiate into multiple cell lineages with specific functions (Bertoni, 2008). The candidate stem cell had to be easy to extract, maintaining the capacity of myogenic conversion when transplanted into the host muscle and also the survival and the subsequent migration from the site of injection to the compromise muscles of the body (Price et al., 2007). With the advent of more sensitive markers, stem cell populations suitable for clinical experiments were found to derive from multiple region of the body at various stage of development. Numerous studies showed that the regenerative capacity of stem cells resided in the environmental microniche and its regulation. This way, it could be important to better elucidate the molecular composition cytokines, growth factors, cell adhesion molecules and extracellular matrix molecules - and interactions of the different microniches that regulate stem cell development (Stocum, 2001).

Several groups published different works concerning adult stem cells such as muscle-derived stem cells (Qu-Petersen et al., 2002), mesoangioblasts (Cossu and Bianco, 2003), blood- (Gavina et al., 2006) and muscle (Benchaouir et al., 2007)-derived CD133+ stem cells. Although some of them are able to migrate through the vasculature (Benchaouir et al., 2007; Galvez et al., 2006; Gavina et al., 2006) and efforts were done to increase their migratory ability (Lafreniere et al., 2006; Torrente et al., 2003a), poor results were obtained.

Embryonic and adult stem cells differ significantly in regard to their differentiation potential and in vitro expansion capability. While adult stem cells constitute a reservoir for tissue regeneration throughout the adult life, they are tissue-specific and possess limited capacity to be expanded ex vivo. Embryonic Stem (ES) cells are derived from the inner cell mass of blastocyst embryos and, by definition, are capable of unlimited in vitro self-renewal and have the ability to differentiate into any cell type of the body (Darabi et al., 2008b). ES cells, together with recently identified iPS cells, are now broadly and extensively studied for their applications in clinical studies.

Embryonic stem cells are pluripotent cells derived from the early embryo that are characterized by the ability to proliferate over prolonged periods of culture remaining undifferentiated and maintaining a stable karyotype (Amit and Itskovitz-Eldor, 2002; Carpenter et al., 2003; Hoffman and Carpenter, 2005). They are capable of differentiating into cells present in all 3 embryonic germ layers, namely ectoderm, mesoderm, and endoderm, and are characterized by self-renewal, immortality, and pluripotency (Strulovici et al., 2007).

hESCs are derived by microsurgical removal of cells from the inner cell mass of a blastocyst stage embryo (Fig. 1). The ES cells can be also obtained from single blastomeres. This technique creates ES cells from a single blastomere directly removed from the embryo bypassing the ethical issue of embryo destruction (Klimanskaya et al., 2006). Although maintaining the viability of the embryo, it has to be determined whether embryonic stem cell lines derived from a single blastomere that does not compromise the embryo can be considered for clinical studies. Cell Nuclear Transfer (SCNT): Nuclear transfer, also referred to as nuclear cloning, denotes the introduction of a nucleus from an adult donor cell into an enucleated oocyte to generate a cloned embryo (Wilmut et al., 2002).

ESCs differentiation. Differentiation potentiality of human embryonic stem cell lines. Human embryonic stem cell pluripotency is evaluated by the ability of the cells to differentiate into different cell types.

Original post:
Stem Cell Therapy for Neuromuscular Diseases | InTechOpen

To Read More: Stem Cell Therapy for Neuromuscular Diseases | InTechOpen
categoriaIPS Cell Therapy commentoComments Off on Stem Cell Therapy for Neuromuscular Diseases | InTechOpen | dataNovember 23rd, 2013
Read All

MD Supervised Stem Cell Therapy

By daniellenierenberg

Regenerative medicine is the process of replacing or regenerating human cells, tissues or organs to restore or establish normal function.[1] This field holds the promise of regenerating damaged tissues and organs in the body by replacing damaged tissue and/or by stimulating the bodys own repair mechanisms to heal previously irreparable tissues or organs.

Regenerative medicine also includes the possibility of growing tissues and organs in the laboratory and safely implant them when the body cannot heal itself This can potentially solves the problem of the shortage of organs available for donation, and the problem of organ transplant rejection if the organs cells are derived from the patients own tissue or cells.[2][3][4]

Widely attributed to having first been coined by William Haseltine (founder of Human Genome Sciences),[5] the term Regenerative Medicine was first found in a 1992 article on hospital administration by Leland Kaiser. Kaisers paper closes with a series of short paragraphs on future technologies that will impact hospitals. One such paragraph had Regenerative Medicine as a bold print title and went on to state, A new branch of medicine will develop that attempts to change the course of chronic disease and in many instances will regenerate tired and failing organ systems.[6][7]

Regenerative medicine refers to a group of biomedical approaches to clinical therapies that may involve the use of stem cells.[8] Examples include the injection of stem cells or progenitor cells (cell therapies); the induction of regeneration by biologically active molecules administered alone or as a secretion by infused cells (immunomodulation therapy); and transplantation of in vitro grown organs and tissues (Tissue engineering).[9][10]

A form of regenerative medicine that recently made it into clinical practice, is the use of heparan sulfate analogues on (chronic) wound healing. Heparan sulfate analogues replace degraded heparan sulfate at the wound site. They assist the damaged tissue to heal itself by repositioning growth factors and cytokines back into the damaged extracellular matrix.[11][12][13] For example, in abdominal wall reconstruction (like inguinal hernia repair), biologic meshes are being used with some success.

At the Wake Forest Institute for Regenerative Medicine, in North Carolina, Dr. Anthony Atala and his colleagues have successfully extracted muscle and bladder cells from several patients bodies, cultivated these cells in petri dishes, and then layered the cells in three-dimensional molds that resembled the shapes of the bladders. Within weeks, the cells in the molds began functioning as regular bladders which were then implanted back into the patients bodies.[14] The team is currently[when?] working on re-growing over 22 other different organs including the liver, heart, kidneys and testicles.[15]

From 1995 to 1998 Michael D. West, PhD, organized and managed the research between Geron Corporation and its academic collaborators James Thomson at the University of Wisconsin-Madison and John Gearhart of Johns Hopkins University that led to the first isolation of human embryonic stem and human embryonic germ cells.[16]

Dr. Stephen Badylak, a Research Professor in the Department of Surgery and director of Tissue Engineering at the McGowan Institute for Regenerative Medicine at the University of Pittsburgh, developed a process for scraping cells from the lining of a pigs bladder, decellularizing (removing cells to leave a clean extracellular structure) the tissue and then drying it to become a sheet or a powder. This cellular matrix powder was used to regrow the finger of Lee Spievak, who had severed half an inch of his finger after getting it caught in a propeller of a model plane.[17][18][19][dubious discuss] As of 2011, this new technology is being employed by the military to U.S. war veterans in Texas, as well as to some civilian patients. Nicknamed pixie-dust, the powdered extracellular matrix is being used success to regenerate tissue lost and damaged due to traumatic injuries.

In June 2008, at the Hospital Clnic de Barcelona, Professor Paolo Macchiarini and his team, of the University of Barcelona, performed the first tissue engineered trachea (wind pipe) transplantation. Adult stem cells were extracted from the patients bone marrow, grown into a large population, and matured into cartilage cells, or chondrocytes, using an adaptive method originally devised for treating osteoarthritis. The team then seeded the newly grown chondrocytes, as well as epithileal cells, into a decellularised (free of donor cells) tracheal segment that was donated from a 51 year old transplant donor who had died of cerebral hemorrhage. After four days of seeding, the graft was used to replace the patients left main bronchus. After one month, a biopsy elicited local bleeding, indicating that the blood vessels had already grown back successfully.[20][21]

In 2009 the SENS Foundation was launched, with its stated aim as the application of regenerative medicine defined to include the repair of living cells and extracellular material in situ to the diseases and disabilities of ageing. [22]

View post:
MD Supervised Stem Cell Therapy

To Read More: MD Supervised Stem Cell Therapy
categoriaIPS Cell Therapy commentoComments Off on MD Supervised Stem Cell Therapy | dataNovember 9th, 2013
Read All

IPS Cell Therapy – Genetherapy

By LizaAVILA

In 2006, Shinya Yamanaka of Kyoto University in Japan was the first to disprove the previous notion that reversible cell differentiation of mammals was impossible. He reprogrammed a fully differentiated mouse cell into a pluripotent stem cell by introducing four genes, Oct-4, SOX2, KLF4, and Myc, into the mouse fibroblast through gene-carrying viruses. With this method, he and his coworkers created induced pluripotent stem cells (iPS cells), the key component in this experiment.[1] Scientists have been able to conduct experiments that show the ability of iPS cells to treat and even cure diseases. In this experiment, tests were run on mice with inherited sickle cell anemia.Skin cells were turned into cells containing genes that transformed the cells into iPS cells. These replaced the diseased sickled cells, curing the test mice. The reprogramming of the pluripotent stem cells in mice was successfully duplicated with human pluripotent stem cells within about a year of the experiment on the mice.

Sickle cell anemia is a disease in which the body produces abnormally shaped red blood cells. Red blood cells are flexible and round, moving easily through the blood vessels. Infected cells are shaped like a crescent or sickle (the namesake of the disease). As a result of this disorder the hemoglobin protein in red blood cells is faulty. Normal hemoglobin bonds to oxygen, then releases it into cells that need it. The blood cell retains its original form and is cycled back to the lungs and re-oxygenated.

Sickle cell hemoglobin, however, after giving up oxygen, cling together and make the red blood cell stiff. The sickle shape also makes it difficult for the red blood cell to navigate arteries and causes blockages.[2] This can cause intense pain and organ damage. The sickled red blood cells are fragile and prone to rupture. When the number of red blood cells decreases from rupture (hemolysis), anemia is the result. Sickle cells also die in 1020 days as opposed to the traditional 120-day lifespan of a normal red blood cell.

Sickle cell anemia is inherited as an autosomal (meaning that the gene is not linked to a sex chromosome) recessive condition.[2] This means that the gene can be passed on from a carrier to his or her children. In order for sickle cell anemia to affect a person, the gene must be inherited from both the mother and the father, so that the child has two recessive sickle cell genes (a homozygous inheritance). People who inherit one sickle cell gene from one parent and one normal gene from the other parent, i.e. heterozygous patients, have a condition called sickle cell trait. Their bodies make both sickle hemoglobin and normal hemoglobin. They may pass the trait on to their children.

The effects of sickle cell anemia vary from person to person. People who have the disease suffer from varying degrees of chronic pain and fatigue. With proper care and treatment, the quality of health of most patients will improve. Doctors have learned a great deal about sickle cell anemia since its discovery in 1979. They know its causes, its effects on the body, and possible treatments for complications. Sickle cell anemia has no widely available cure. A bone marrow transplant is the only treatment method currently recognized to be able to cure the disease, though it does not work for every patient. Finding a donor is difficult and the procedure could potentially do more harm than good. Treatments for sickle cell anemia are generally aimed at avoiding crises, relieving symptoms, and preventing complications. Such treatments may include medications, blood transfusions, and supplemental oxygen.

During the first step of the experiment, skin cells (also known as fibroblasts) were collected from infected test mice and put in a culture. The fibroblasts were reprogrammed by infecting them with retroviruses that contained genes common to embryonic stem cells. These genes were the same four used by Yamanaka (Oct-4, SOX2, KLF4, and Myc) in his earlier study. The investigators were trying to produce cells with the potential to differentiate into any type of cell needed (i.e. pluripotent stem cells). As the experiment continued, the fibroblasts multiplied into identical copies of iPS cells. The cells were then treated to form the mutation needed to reverse the anemia in the mice. This was accomplished by restructuring the DNA containing the defective globin gene into DNA with the normal gene through the process of homologous recombination. The iPS cells then differentiated into blood stem cells, or hematopoietic stem cells. The hematopoietic cells were injected back into the infected mice, where they proliferate and differentiate into normal blood cells, curing the mice of the disease.[3][4][verification needed]

To determine whether the mice were cured from the disease, the scientists checked for the usual symptoms of sickle cell disease. They examined the blood for mean corpuscular volume (MCV) and red cell distribution width (RDW) and urine concentration defects. They also checked for sickled red blood cells. They examined the DNA through gel electrophoresis, checking for bands that display an allele that causes sickling. Compared to the untreated mice with the disease, which they used as a control, the treated animals had marked increases in RBC counts, healthy hemoglobin, and packed cell volume levels.[5]

Researchers examined the urine concentration defect, which results from RBC sickling in renal tubules and consequent reduction in renal medullary blood flow, and the general deteriorated systemic condition reflected by lower body weight and increased breathing.[5] They were able to see that these parts of the body of the mice had healed or improved. This indicated that all hematological and systemic parameters of sickle cell anemia improved substantially and were comparable to those in control mice.[5] They cannot say if this will work in humans because a safe way to inject the genes for the induced pluripotent cells is still needed.[citation needed]

The reprogramming of the induced pluripotent stem cells in mice was successfully duplicated in humans within a year of the successful experiment on the mice. This reprogramming was done in several labs and it was shown that the iPS cells in humans were almost identical to original embryonic stem cells (ES cells) that are responsible for the creation of all structures in a fetus.[1] An important feature of iPS cells is that they can be generated with cells taken from an adult, which would circumvent many of the ethical problems associated with working with ES cells. These iPS cells also have potential in creating and examining new disease models and developing more efficient drug treatments.[6] Another feature of these cells is that they provide researchers with a human cell sample, as opposed to simply using an animal with similar DNA, for drug testing.

One major problem with iPS cells is the way in which the cells are reprogrammed. Using gene-carrying viruses has the potential to cause iPS cells to develop into cancerous cells.[1] Also, an implant made using undifferentiated iPS cells, could cause a teratoma to form. Any implant that is generated from using these iPS cells would only be viable for transplant into the original subject that the cells were taken from. In order for these iPS cells to become viable in therapeutic use, there are still many steps that must be taken.[5][7]

Read more from the original source:
IPS Cell Therapy - Genetherapy

To Read More: IPS Cell Therapy – Genetherapy
categoriaIPS Cell Therapy commentoComments Off on IPS Cell Therapy – Genetherapy | dataNovember 8th, 2013
Read All

Induced pluripotent stem cell therapy – Wikipedia, the free …

By Sykes24Tracey

In 2006, Shinya Yamanaka of Kyoto University in Japan was the first to disprove the previous notion that reversible cell differentiation of mammals was impossible. He reprogrammed a fully differentiated mouse cell into a pluripotent stem cell by introducing four genes, Oct-4, SOX2, KLF4, and Myc, into the mouse fibroblast through gene-carrying viruses. With this method, he and his coworkers created induced pluripotent stem cells (iPS cells), the key component in this experiment.[1] Scientists have been able to conduct experiments that show the ability of iPS cells to treat and even cure diseases. In this experiment, tests were run on mice with inherited sickle cell anemia.Skin cells were turned into cells containing genes that transformed the cells into iPS cells. These replaced the diseased sickled cells, curing the test mice. The reprogramming of the pluripotent stem cells in mice was successfully duplicated with human pluripotent stem cells within about a year of the experiment on the mice.

Sickle cell anemia is a disease in which the body produces abnormally shaped red blood cells. Red blood cells are flexible and round, moving easily through the blood vessels. Infected cells are shaped like a crescent or sickle (the namesake of the disease). As a result of this disorder the hemoglobin protein in red blood cells is faulty. Normal hemoglobin bonds to oxygen, then releases it into cells that need it. The blood cell retains its original form and is cycled back to the lungs and re-oxygenated.

Sickle cell hemoglobin, however, after giving up oxygen, cling together and make the red blood cell stiff. The sickle shape also makes it difficult for the red blood cell to navigate arteries and causes blockages.[2] This can cause intense pain and organ damage. The sickled red blood cells are fragile and prone to rupture. When the number of red blood cells decreases from rupture (hemolysis), anemia is the result. Sickle cells also die in 1020 days as opposed to the traditional 120-day lifespan of a normal red blood cell.

Sickle cell anemia is inherited as an autosomal (meaning that the gene is not linked to a sex chromosome) recessive condition.[2] This means that the gene can be passed on from a carrier to his or her children. In order for sickle cell anemia to affect a person, the gene must be inherited from both the mother and the father, so that the child has two recessive sickle cell genes (a homozygous inheritance). People who inherit one sickle cell gene from one parent and one normal gene from the other parent, i.e. heterozygous patients, have a condition called sickle cell trait. Their bodies make both sickle hemoglobin and normal hemoglobin. They may pass the trait on to their children.

The effects of sickle cell anemia vary from person to person. People who have the disease suffer from varying degrees of chronic pain and fatigue. With proper care and treatment, the quality of health of most patients will improve. Doctors have learned a great deal about sickle cell anemia since its discovery in 1979. They know its causes, its effects on the body, and possible treatments for complications. Sickle cell anemia has no widely available cure. A bone marrow transplant is the only treatment method currently recognized to be able to cure the disease, though it does not work for every patient. Finding a donor is difficult and the procedure could potentially do more harm than good. Treatments for sickle cell anemia are generally aimed at avoiding crises, relieving symptoms, and preventing complications. Such treatments may include medications, blood transfusions, and supplemental oxygen.

During the first step of the experiment, skin cells (also known as fibroblasts) were collected from infected test mice and put in a culture. The fibroblasts were reprogrammed by infecting them with retroviruses that contained genes common to embryonic stem cells. These genes were the same four used by Yamanaka (Oct-4, SOX2, KLF4, and Myc) in his earlier study. The investigators were trying to produce cells with the potential to differentiate into any type of cell needed (i.e. pluripotent stem cells). As the experiment continued, the fibroblasts multiplied into identical copies of iPS cells. The cells were then treated to form the mutation needed to reverse the anemia in the mice. This was accomplished by restructuring the DNA containing the defective globin gene into DNA with the normal gene through the process of homologous recombination. The iPS cells then differentiated into blood stem cells, or hematopoietic stem cells. The hematopoietic cells were injected back into the infected mice, where they proliferate and differentiate into normal blood cells, curing the mice of the disease.[3][4][verification needed]

To determine whether the mice were cured from the disease, the scientists checked for the usual symptoms of sickle cell disease. They examined the blood for mean corpuscular volume (MCV) and red cell distribution width (RDW) and urine concentration defects. They also checked for sickled red blood cells. They examined the DNA through gel electrophoresis, checking for bands that display an allele that causes sickling. Compared to the untreated mice with the disease, which they used as a control, the treated animals had marked increases in RBC counts, healthy hemoglobin, and packed cell volume levels.[5]

Researchers examined the urine concentration defect, which results from RBC sickling in renal tubules and consequent reduction in renal medullary blood flow, and the general deteriorated systemic condition reflected by lower body weight and increased breathing.[5] They were able to see that these parts of the body of the mice had healed or improved. This indicated that all hematological and systemic parameters of sickle cell anemia improved substantially and were comparable to those in control mice.[5] They cannot say if this will work in humans because a safe way to inject the genes for the induced pluripotent cells is still needed.[citation needed]

The reprogramming of the induced pluripotent stem cells in mice was successfully duplicated in humans within a year of the successful experiment on the mice. This reprogramming was done in several labs and it was shown that the iPS cells in humans were almost identical to original embryonic stem cells (ES cells) that are responsible for the creation of all structures in a fetus.[1] An important feature of iPS cells is that they can be generated with cells taken from an adult, which would circumvent many of the ethical problems associated with working with ES cells. These iPS cells also have potential in creating and examining new disease models and developing more efficient drug treatments.[6] Another feature of these cells is that they provide researchers with a human cell sample, as opposed to simply using an animal with similar DNA, for drug testing.

One major problem with iPS cells is the way in which the cells are reprogrammed. Using gene-carrying viruses has the potential to cause iPS cells to develop into cancerous cells.[1] Also, an implant made using undifferentiated iPS cells, could cause a teratoma to form. Any implant that is generated from using these iPS cells would only be viable for transplant into the original subject that the cells were taken from. In order for these iPS cells to become viable in therapeutic use, there are still many steps that must be taken.[5][7]

Read more from the original source:
Induced pluripotent stem cell therapy - Wikipedia, the free ...

To Read More: Induced pluripotent stem cell therapy – Wikipedia, the free …
categoriaIPS Cell Therapy commentoComments Off on Induced pluripotent stem cell therapy – Wikipedia, the free … | dataNovember 8th, 2013
Read All

iPSCTherapy.com: Induced Pluripotent Stem Cell therapy Information …

By Dr. Matthew Watson

There have been hundreds of science fiction stories and books written about growing organs in scientific laboratories as replacements for those that no longer function properly, or about injecting scientifically transmuted cells into ailing patients that can repair the broken cells within their bodies, bringing them back to robust health. In todays language what they were talking about was Induced Pluripotent Stem Cell (iPSC) Therapy.

Here, in the early 21st century, the gap between science fiction and science truth is closing at a record rate due to the rapid progress made in iPSC Therapy research, especially over the last three years.

After the virtual stop order placed on embryonic cell stem research in 2001, the race to find an alternative type of stem cell began in earnest, and in 2006 Shinya Yamanaka of Kyoto University in Japan announced his teams successful reprogramming of mouse cells into iPSCs. This was the breakthrough that made it possible for stem cell research to continue without the use of controversial embryonic stem cells.

The next major announcement came in 2007, again from Yamanaka in Japan, followed by one only a few weeks later by James A. Thompson from the University of Wisconsin, detailing the making of iPSC from adult human cells. Again, neither used embryos in their experiments.

From that time on the goal has been developing stem cell science that will eventually be safe iPS Cell Therapy modalities to be used in Regenerative or Reparative Medicine. What kinds of illnesses or diseases will iPSC Therapies be used to treat in the future? Only a partial list would include:

The world of iPSC Therapy research is wide open today and its on the move! This website is dedicated to bringing you first, the story of stem cell research, both embryonic and iPStem Cell, and the controversy surrounding them, as well as the most up to date information in the easiest to understand language about major milestone accomplishments in the field.

If you were to go back 100 years you would be amazed by how primitive medicine was. Even 60 years ago there were no organ transplants, no cystoscopic surgeries, and there was a massive polio outbreak in the United States that closed public swimming pools and beaches and other public gathering places across the country for the summer. Who can tell where medicine will be in 10 or 15 years? There is no predicting, but with the rapid advancement of the last few years and the bright promise shown so far, iPSC Therapy is sure to play a major role.

Continued here:
iPSCTherapy.com: Induced Pluripotent Stem Cell therapy Information ...

To Read More: iPSCTherapy.com: Induced Pluripotent Stem Cell therapy Information …
categoriaIPS Cell Therapy commentoComments Off on iPSCTherapy.com: Induced Pluripotent Stem Cell therapy Information … | dataNovember 8th, 2013
Read All

Human muscle stem cell therapy gets help from zebrafish

By daniellenierenberg

PUBLIC RELEASE DATE:

7-Nov-2013

Contact: B. D. Colen bd_colen@harvard.edu 617-495-7821 Harvard University

Harvard Stem Cell Scientists have discovered that the same chemicals that stimulate muscle development in zebrafish can also be used to differentiate human stem cells into muscle cells in the laboratory, an historically challenging task that, now overcome, makes muscle cell therapy a more realistic clinical possibility.

The work, published this week in the journal Cell, began with a discovery by Boston Children's Hospital researchers, led by Leonard Zon, MD, and graduate student Cong (Tony) Xu, who tested 2,400 different chemicals in cultures of zebrafish embryo cells to determine if any could increase the numbers of muscle cells formed. Using fluorescent reporter fish in which muscle cells were visible during their creation, the researchers found six chemicals that were very effective at promoting muscle formation.

Zon shared his results with Harvard Department of Stem Cell and Regenerative Biology professor Amy Wagers, PhD, and Mohammadsharif Tabebordbar, a graduate student in her laboratory, who tested the six chemicals in mice. One of the six, called forskolin, was found to increase the numbers of muscle stem cells from mice that could be obtained when these cells were grown in laboratory dishes. Moreover, the cultured cells successfully integrated into muscle when transplanted back into mice.

Inspired by the successful application of these chemicals in mice, Salvatore Iovino, PhD, a joint postdoctoral fellow in the Wagers lab and the lab of C. Ronald Kahn, MD, at the Joslin Diabetes Center, investigated whether the chemicals would also affect human cells and found that a combination of three chemicals, including forskolin, could induce differentiation of human induced pluripotent stem (iPS) cells, made by reprogramming skin cells. Exposure of iPS cells to these chemicals converted them into skeletal muscle, an outcome the Wagers and Kahn labs had been striving to achieve for years using conventional methods. When transplanted into a mouse, the human iPS-derived muscle cells also contributed to muscle repair, offering early promise that this protocol could provide a route to muscle stem cell therapy in humans.

The interdisciplinary, cross-laboratory collaboration between Zon, Wagers, and Kahn highlights the advantage of open exchange between researchers. "If we had done this screen directly on human iPS cells, it would have taken at least 10 times as long and cost 100 times as much," said Wagers. "The zebrafish gave us a big advantage here because it has a fast generation time, rapid development, and can be easily and relatively cheaply screened in a culture dish."

"This research demonstrates that over 300 million years of evolution, the pathways used in the fish are conserved through vertebrates all the way up to the human," said Wagers' fellow HSCRB professor Leonard Zon, chair of the Harvard Stem Cell Institute Executive Committee and director of the stem cell program at Boston Children's Hospital. "We can now make enough human muscle progenitors in a dish to allow us to model diseases of the muscle lineage, like Duchenne muscular dystrophy, conduct drug screens to find chemicals that correct those disease, and in the long term, efficiently transplant muscle stem cells into a patient."

In a similar biomedical application, Kahn, who is chief academic officer at the Joslin, plans to apply the new ability to quickly produce muscle stem cells for diabetes research. His lab will generate iPS-derived muscle cells from people who are at risk for diabetes and people who have diabetes to identify alterations that lead to insulin resistance in the muscle.

Originally posted here:
Human muscle stem cell therapy gets help from zebrafish

To Read More: Human muscle stem cell therapy gets help from zebrafish
categoriaIPS Cell Therapy commentoComments Off on Human muscle stem cell therapy gets help from zebrafish | dataNovember 8th, 2013
Read All

What are induced pluripotent stem cells or iPS cells? – Stem Cells …

By Dr. Matthew Watson

In November 2007 scientists announced they had developed a new way to cause mature human cells to resemble pluripotent stem cells - similar in many ways to human embryonic stem cells. By simply altering the expression of just four genes using genetic modification, the mature cells were 'induced' to become more primitive, stem cells and were referred to as 'induced' pluripotent stem (iPS) cells.

Initially iPS cells were generated using viruses to change gene expression, however since the initial discovery, technologies for reprogramming cells are moving very quickly and researchers are now investigating the use of new methods that do not use viruses which can cause permanent and potentially harmful changes in the cells. If they are able to be made safely, and on a large scale, iPS cells could possibly be used to provide a source of cells to replace cells damaged following illness or disease. It may even be possible to make stem cells for therapy from a patient's own cells and thereby avoid the use of anti-rejection medications.

However, right now scientists are using this method to create disease specific cells for research by taking a cells - maybe from a skin biopsy - from a patient with a genetic disorder, such as Huntingtons disease, and then using the iPS cells to study the disease in the laboratory. Scientist hope that such an approach will help them understand the development and progression of certain diseases, and assist in the development and testing of new drugs to treat disease.

While the discovery of iPS cells was a very important development, more research needs to be done to discover if they will offer the same research value as embryonic stem cells and if they will be as useful for therapy.

To learn more about iPS cells watch What are induced pluripotent stem cells? in our video library.

Read more here:
What are induced pluripotent stem cells or iPS cells? - Stem Cells ...

To Read More: What are induced pluripotent stem cells or iPS cells? – Stem Cells …
categoriaIPS Cell Therapy commentoComments Off on What are induced pluripotent stem cells or iPS cells? – Stem Cells … | dataNovember 3rd, 2013
Read All

Stem Cell Definitions | California’s Stem Cell Agency

By JoanneRUSSELL25

En Espaol

The term stem cell by itself can be misleading. There are many different types of stem cells, each with very different potential to treat disease. The so-called adult stem cells come from any organ, from the fetus through the adult. These are also called tissue stem cells. The so-called pluripotent cells, which have the ability to form all cells in the body, can be either embryonic or induced pluripotent stem (iPS) cells.

All stem cells, whether they are tissue stem cells or pluripotent cells, have the ability to divide and create an identical copy of themselves. This process is called self-renewal. The cells can also divide to form cells that go on to develop into mature tissue types such as liver, lungs, brain, or skin.

Embryonic stem cells exist only at the earliest stages of embryonic development and go on to form all the cells of the adult body. In humans, these cells no longer exist after about five days of development.

When removed and grown in a lab dish these stem cells can continue dividing indefinitely, retaining the ability to form the more than 200 adult cell types. Because the cells have the potential to form so many different adult tissues they are also called pluripotent ("pluri" = many, "potent" = potentials) stem cells.

James Thomson, a professor of Anatomy at the University of Wisconsin, isolated the first human embryonic stem cells in 1998. He now shares a joint appointment at the University of California, Santa Barbara.

Irv Weissman talks about the difference between adult and embryonic stem cells (3:29)

Pluripotent means many (pluri) potentials (potent). In other words, these cells have the potential of taking on many fates in the body, including all of the more than 200 different cell types. Embryonic stem cells are pluripotent, as are iPS cells that are reprogrammed from adult tissues. When scientists talk about pluripotent stem cells they mostly mean either embryonic or iPS cells.

What people commonly call adult stem cells are more accurately called tissue-specific stem cells. These are specialized cells found in tissues of adults, children and fetuses. They are thought to exist in most of the bodys tissues such as the blood, brain, liver, intestine or skin. These cells are committed to becoming a cell from their tissue of origin, but they still have the broad ability to become any one of these cells. Stem cells of the bone marrow, for example, can give rise to any of the red or white cells of the blood system. Stem cells in the brain can form all the neurons and support cells of the brain, but cant form non-brain tissues. Unlike embryonic stem cells, researchers have not been able to grow adult stem cells indefinitely in the lab.

In recent years, scientists have found stem cells in the placenta and in the umbilical cord of newborn infants. Although these cells come from a newborn they are like adult stem cells in that they are already committed to becoming a particular type of cell and cant go on to form all tissues of the body. The cord blood cells that some people bank after the birth of a child are a form of adult blood-forming stem cells.

See the original post here:
Stem Cell Definitions | California's Stem Cell Agency

To Read More: Stem Cell Definitions | California’s Stem Cell Agency
categoriaIPS Cell Therapy commentoComments Off on Stem Cell Definitions | California’s Stem Cell Agency | dataNovember 3rd, 2013
Read All

Induced pluripotent stem cell – Wikipedia, the free encyclopedia

By Dr. Matthew Watson

Induced pluripotent stem cells,[1] commonly abbreviated as iPS cells or iPSCs are a type of pluripotent stem cell artificially derived from a non-pluripotent cell typically an adult somatic cell by inducing a "forced" expression of specific genes.

Induced pluripotent stem cells are similar to natural pluripotent stem cells, such as embryonic stem (ES) cells, in many aspects, such as the expression of certain stem cell genes and proteins, chromatin methylation patterns, doubling time, embryoid body formation, teratoma formation, viable chimera formation, and potency and differentiability, but the full extent of their relation to natural pluripotent stem cells is still being assessed.[2] Induced pluripotent cells have been made from adult stomach, liver, skin cells, blood cells, prostate cells and urinary tract cells.[3]

iPSCs were first produced in 2006 from mouse cells and in 2007 from human cells in a series of experiments by Shinya Yamanaka's team at Kyoto University, Japan, and by James Thomson's team at the University of Wisconsin-Madison. For her iPSC research, Dr. Nancy Bachman, of Oneonta, NY, was awarded the Wolf Prize in Medicine in 2012 (along with John B. Gurdon).[4][5][6] For his iPSC discovery (and for deriving the first human embryonic stem cell), James Thomson received the 2011 Albany Medical Center Prize for Biomedical Research and the 2011 King Faisal International Prize, which he shared with Yamanaka. In October 2012, Yamanaka and fellow stem cell researcher John Gurdon were awarded the Nobel Prize in Physiology or Medicine "for the discovery that mature cells can be reprogrammed to become pluripotent."[7]

iPSCs are an important advance in stem cell research, as they may allow researchers to obtain pluripotent stem cells, which are important in research and potentially have therapeutic uses, without the controversial use of embryos. Because iPSCs are developed from a patient's own somatic cells, it was believed that treatment of iPSCs would avoid any immunogenic responses; however, Zhao et al. have challenged this assumption.[8]

Depending on the methods used, reprogramming of adult cells to obtain iPSCs may pose significant risks that could limit their use in humans. For example, if viruses are used to genomically alter the cells, the expression of cancer-causing genes "oncogenes" may potentially be triggered. In February 2008, scientists announced the discovery of a technique that could remove oncogenes after the induction of pluripotency, thereby increasing the potential use of iPS cells in human diseases.[9] In April 2009, it was demonstrated that generation of iPS cells is possible without any genetic alteration of the adult cell: a repeated treatment of the cells with certain proteins channeled into the cells via poly-arginine anchors was sufficient to induce pluripotency.[10] The acronym given for those iPSCs is piPSCs (protein-induced pluripotent stem cells).

Dedifferentiation to totipotency or pluripotency: an overview of methods. Various methods exist to revert adult somatic cells to pluripotency or totipotency. In the case of totipotency, reprogramming is mediated through a mature metaphase II oocyte as in somatic cell nuclear transfer (Wilmut et al., 1997). Recent work has demonstrated the feasibility of enucleated zygotes or early blastomeres chemically arrested during mitosis, such that nuclear envelope break down occurs, to support reprogramming to totipotency in a process called chromosome transfer (Egli and Eggan, 2010). Direct reprogramming methods support reversion to pluripotency; though, vehicles and biotypes vary considerably in efficiencies (Takahashi and Yamanaka, 2006). Viral-mediated transduction robustly supports dedifferentiation to pluripotency through retroviral or DNA-viral routes but carries the onus of insertional inactivation. Additionally, epigenetic reprogramming by enforced expression of OSKM through DNA routes exists such as plasmid DNA, minicircles, transposons, episomes and DNA mulicistronic construct targeting by homologous recombination has also been demonstrated; however, these methods suffer from the burden to potentially alter the recipient genome by gene insertion (Ho et al., 2010). While protein-mediated transduction supports reprogramming adult cells to pluripotency, the method is cumbersome and requires recombinant protein expression and purification expertise, and reprograms albeit at very low frequencies (Kim et al., 2009). A major obstacle of using RNA for reprogramming is its lability and that single-stranded RNA biotypes trigger innate antiviral defense pathways such as interferon and NF-B-dependent pathways. In vitro transcribed RNA, containing stabilizing modifications such as 5-methylguanosine capping or substituted ribonucleobases, e.g. pseudouracil, is 35-fold more efficient than viral transduction and has the additional benefit of not altering the somatic genome (Warren et al., 2010). An overarching goal of reprogramming methods is to replace genes with small molecules to assist in reprogramming. No cocktail has been identified to completely reprogram adult cells to totipotency or pluripotency, but many examples exist that improve the overall efficiency of the process and can supplant one or more genes by direct reprogramming routes (Feng et al., 2009; Zhu et al., 2010).

iPS cells are typically derived by transfection of certain stem cell-associated genes into non-pluripotent cells, such as adult fibroblasts, although this technique is becoming less popular since it is known to be prone to inducing cancer formation. Transfection is typically achieved through viral vectors, such as retroviruses. Transfected genes include the master transcriptional regulators Oct-3/4 (Pou5f1) and Sox2, although it is suggested that other genes enhance the efficiency of induction. After 34 weeks, small numbers of transfected cells begin to become morphologically and biochemically similar to pluripotent stem cells, and are typically isolated through morphological selection, doubling time, or through a reporter gene and antibiotic selection.

Induced pluripotent stem cells were first generated by Shinya Yamanaka's team at Kyoto University, Japan in 2006. Yamanaka used genes that had been identified as particularly important in embryonic stem cells (ESCs), and used retroviruses to transduce mouse fibroblasts with a selection of those genes. Eventually, four key pluripotency genes essential for the production of pluripotent stem cells were isolated; Oct-3/4, SOX2, c-Myc, and Klf4. Cells were isolated by antibiotic selection of Fbx15+ cells. However, this iPS cell line showed DNA methylation errors compared to original patterns in ESC lines and failed to produce viable chimeras if injected into developing embryos.

In June 2007, the same group published a breakthrough study along with two other independent research groups from Harvard, MIT, and the University of California, Los Angeles, showing successful reprogramming of mouse fibroblasts into iPS cells and even producing viable chimera. These cell lines were also derived from mouse fibroblasts by retroviral mediated reactivation of the same four endogenous pluripotent factors, but the researchers now selected a different marker for detection. Instead of Fbx15, they used Nanog which is an important gene in ESCs. DNA methylation patterns and production of viable chimeras (and thereby contributing to subsequent germ-line production) indicated that Nanog is a major determinant of cellular pluripotency.[11][12][13][14][15]

Unfortunately, two of the four genes used (namely, c-Myc and KLF4) are oncogenic, and 20% of the chimeric mice developed cancer. In a later study, Yamanaka reported that one can create iPSCs even without c-Myc. The process takes longer and is not as efficient, but the resulting chimeras didn't develop cancer.[16]

Go here to see the original:
Induced pluripotent stem cell - Wikipedia, the free encyclopedia

To Read More: Induced pluripotent stem cell – Wikipedia, the free encyclopedia
categoriaIPS Cell Therapy commentoComments Off on Induced pluripotent stem cell – Wikipedia, the free encyclopedia | dataNovember 3rd, 2013
Read All

Read in

By NEVAGiles23

TOKYO: Shinya Yamanaka, fresh from the Nobel Prize for medicine, states that science and ethics must go hand in hand. Interviewed by the Mainichi Shimbun after the award, he said: "I would like to invite ethical experts as teachers at my laboratory and work to guide iPS [induced pluripotent stem] cell research from that direction as well. The work of a scientific researcher is just one part of the equation. "

Yamanaka, 50, found that adult cells can be transformed into cells in their infancy, stem cells (iPS), which are, so to speak, the raw material for the reconstruction of tissue irreparably damaged by disease. For regenerative medicine the implications of Yamanaka's discovery are obvious. Adult skin cells can for example be reprogrammed and transformed into any other cell that is desired: from the skin to the brain, from the skin to the heart, from the skin to elements that produce insulin.

"Their discovery - says the statement of the jury that awarded him the Nobel Prize on October 8 - has revolutionized our understanding of how cells and organisms develop. Through the programming of human cells, scientists have created new opportunities for the study of diseases and development of methods for the diagnosis and therapy ".

These "opportunities" are not only "scientific", but also "ethical". Much of the scientific research and global investment is in fact launched to design and produce stem cells from embryos, arriving at the point of manipulating and destroying them, facing scientists with enormous ethical problems.

" Ethics are really difficult - Yamanaka explainsto Mainichi - In the United States I began work on mouse experiments, and when I returned to Japan I learned that human embryonic stem cells had been created. I was happy that they would contribute to medical science, but I faced an ethical issue. I started iPS cell research as a way to do good things as a researcher, and I wanted to do what I could to expand the merits of embryonic stem cells. If we make sperm or eggs from iPS cells, however, it leads to the creation of new life, so the work I did on iPS cells led to an ethical problem. If we don't prepare debates for ethical problems in advance, technology will proceed ahead faster than we think.. "

The "ethical question" Yamanaka pushed to find a way to "not keep destroying embryos for our research."

Speaking with his co-workers at the University of Kyoto, immediately after receiving the award, Yamanaka showed dedication and modesty.

"Now - he said - I strongly feel a sense of gratitude and responsibility" gratitude for family and friends who have supported him in a demanding journey of discovery that lasted decades; responsibility for a discovery that gives hope to millions of patients. Now iPS cells can grow into any tissue of the human body allowing regeneration of parts so far irretrievably lost due to illness.

His modesty also led him to warn against excessive hopes. To a journalist who asked him for a message to patients and young researchers awaiting the results of his research heresponded: "The iPS cells are also known as versatile cells, and the technology may be giving the false impression to patients that they could be cured any day now. It will still take five or 10 years of research before the technology is feasible. There are over 200 researchers at my laboratory, and I want patients to not give up hope"

"Dozens of times - he continued - I tried to get some results and I have often failed in the experiments .... Many times I was tempted to give up or cry. Without the support of my family, I could not have continued this search. From now on I will be facing the moment of truth. I would like to return to my laboratory as quickly as possible. "

Link:
Read in

To Read More: Read in
categoriaIPS Cell Therapy commentoComments Off on Read in | dataOctober 12th, 2012
Read All

10/11/2012 10:05 JAPAN Nobel Prize for Yamanaka, scientific research and ethics must go hand in hand

By Dr. Matthew Watson

10/11/2012 10:05 JAPAN Nobel Prize for Yamanaka, scientific research and ethics must go hand in hand by Pino Cazzaniga Research on iPS (induced pluripotent stem cells) can produce stem cells from adult cells, for use in regenerative medicine. Shinya Yamanakas discovery reveals that research on embryonic stem cells is unnecessary, saving the lives of many embryos. The Japanese researcher has searched for new ways driven by ethical question.

Tokyo (AsiaNews) - Shinya Yamanaka, fresh from the Nobel Prize for medicine, states that science and ethics must go hand in hand. Interviewed by the Mainichi Shimbun after the award, he said: "I would like to invite ethical experts as teachers at my laboratory and work to guide iPS [induced pluripotent stem] cell research from that direction as well. The work of a scientific researcher is just one part of the equation. "

Yamanaka, 50, found that adult cells can be transformed into cells in their infancy, stem cells (iPS), which are, so to speak, the raw material for the reconstruction of tissue irreparably damaged by disease. For regenerative medicine the implications of Yamanaka's discovery are obvious. Adult skin cells can for example be reprogrammed and transformed into any other cell that is desired: from the skin to the brain, from the skin to the heart, from the skin to elements that produce insulin.

"Their discovery - says the statement of the jury that awarded him the Nobel Prize on October 8 - has revolutionized our understanding of how cells and organisms develop. Through the programming of human cells, scientists have created new opportunities for the study of diseases and development of methods for the diagnosis and therapy ".

These "opportunities" are not only "scientific", but also "ethical". Much of the scientific research and global investment is in fact launched to design and produce stem cells from embryos, arriving at the point of manipulating and destroying them, facing scientists with enormous ethical problems.

" Ethics are really difficult - Yamanaka explainsto Mainichi - In the United States I began work on mouse experiments, and when I returned to Japan I learned that human embryonic stem cells had been created. I was happy that they would contribute to medical science, but I faced an ethical issue. I started iPS cell research as a way to do good things as a researcher, and I wanted to do what I could to expand the merits of embryonic stem cells. If we make sperm or eggs from iPS cells, however, it leads to the creation of new life, so the work I did on iPS cells led to an ethical problem. If we don't prepare debates for ethical problems in advance, technology will proceed ahead faster than we think.. "

The "ethical question" Yamanaka pushed to find a way to "not keep destroying embryos for our research."

Speaking with his co-workers at the University of Kyoto, immediately after receiving the award, Yamanaka showed dedication and modesty.

"Now - he said - I strongly feel a sense of gratitude and responsibility" gratitude for family and friends who have supported him in a demanding journey of discovery that lasted decades; responsibility for a discovery that gives hope to millions of patients. Now iPS cells can grow into any tissue of the human body allowing regeneration of parts so far irretrievably lost due to illness.

Go here to see the original:
10/11/2012 10:05 JAPAN Nobel Prize for Yamanaka, scientific research and ethics must go hand in hand

To Read More: 10/11/2012 10:05 JAPAN Nobel Prize for Yamanaka, scientific research and ethics must go hand in hand
categoriaIPS Cell Therapy commentoComments Off on 10/11/2012 10:05 JAPAN Nobel Prize for Yamanaka, scientific research and ethics must go hand in hand | dataOctober 11th, 2012
Read All

FRC Supports Alliance Defending Freedom, Jubilee Campaign Cert Petition to Supreme Court on Stem Cell Funding

By JoanneRUSSELL25

WASHINGTON, Oct. 10, 2012 /PRNewswire-USNewswire/ --Alliance Defending Freedom and the Jubilee Campaign together with Tom Hungar of Gibson, Dunn & Crutcher today filed a petition for certiorari with the U.S. Supreme Court in the case Sherley v. Sebelius, which seeks to end federal funding of human embryonic stem cell research.

Of the petition David Prentice, Ph.D., senior fellow for life sciences at the Family Research Council's Center for Human Life and Bioethics, made the following comments:

"Even as the Nobel Prize committee honors Japanese scientist Shinya Yamanaka for introducing ethical induced pluripotent stem (iPS) cells to the field of medicine, the Obama administration is fighting to continue wasting taxpayer money on unethical embryonic stem cell research, which relies on the destruction of young human life. A plain reading of federal law would specifically prohibit funding of embryonic stem cell research. After years of wasting taxpayer dollars as well as lives on ethically-tainted experiments, it's time for the federal government to start putting that money into lifesaving and ethical adult stem cell research, the gold standard for patient treatments. Such research is saving thousands of lives now lives like that of Chloe Levine who beat cerebral palsy with the help of adult stem cells. Each precious life at every stage and every age deserves our respect, and we should devote our resources and time to the ethical stem cell research that has the best chance of preserving life adult stem cells.

"We are pleased to see this suit move forward, and hope that the Supreme Court will agree to its review and uphold the clear intent of federal law to protect human life from experimentation."

To watch a video about Chloe Levine and adult stem cell therapy, click here : http://www.youtube.com/watch?feature=player_embedded&v=ojjT4yRd5Es

To learn more about adult stem cells, click here : http://www.stemcellresearchfacts.org/

SOURCE Family Research Council

Follow this link:
FRC Supports Alliance Defending Freedom, Jubilee Campaign Cert Petition to Supreme Court on Stem Cell Funding

To Read More: FRC Supports Alliance Defending Freedom, Jubilee Campaign Cert Petition to Supreme Court on Stem Cell Funding
categoriaIPS Cell Therapy commentoComments Off on FRC Supports Alliance Defending Freedom, Jubilee Campaign Cert Petition to Supreme Court on Stem Cell Funding | dataOctober 11th, 2012
Read All

NeuroGeneration Recruits Top Scientist To Direct New Division of Biotherapeutics and Drug Discovery In La Jolla, CA

By Sykes24Tracey

LOS ANGELES and LA JOLLA, Calif., Oct. 5, 2012 /PRNewswire/ -- A senior research neuroscientist from The Salk Institute,Mohamedi Kagalwala PhD, has been recruited to head NeuroGeneration's new laboratories in La Jolla, California. Dr. Kagalwala, an expert on neural stem cells, will become director of the new Division of Biotherapeutics and Drug Discovery.

"I am extremely pleased to lead NeuroGeneration's new Division and expand its technology of adult neural stem cells for Parkinson's disease. It will allow us to develop personalized iPS cell therapies for degenerative brain disorders," said Dr. Kagalwala. "In addition, by investigating intrinsic neurogenesis and brain repair mechanisms, our team will be able to modify discrete molecular mechanisms during aging and neurodegenerative changes. We will then be in a better position to influence the environment, either with drugs or cellular therapies, to prevent the progression of disease and facilitate brain repair."

This new Division will complement the neural stem cell therapy studies for Parkinson's disease and other Atypical Parkinsonism led by Dr. Michel Levesque, NeuroGeneration's scientific founder.

Within the new facility providing core state of the art technologies, NeuroGeneration will expand its bioinformatic platforms to include personalized neurogenomic, analysis for drug target discovery for aging, Parkinson's disease, Stroke, Amyotrophic Lateral Sclerosis, Alzheimer's disease, Multiple Sclerosis, Epilepsy, Depression and Schizophrenia.

ABOUT NEUROGENERATION:

NeuroGeneration is a life science company designing new cellular therapies and biological modulators for the prevention and treatment of neurodegenerative disorders. The company has completed a Phase I clinical trial for Parkinson's disease using adult-derived autologous neural stem cells. It intends to complete a Phase II study for the treatment of Parkinson's disease as soon as it receives final approval from the FDA. NeuroGeneration's Division of Biotherapeutics and Drug Discovery offers molecular products using its drug discovery platforms to target neuroprotective and endogenous repair mechanisms.

FOR MORE INFORMATION CONTACT:

NeuroGeneration Laboratories Division of Biotherapeutics and Drug Discovery 3210 Merryfield Row San Diego, CA92121

Patricia Eastman NeuroGeneration,Inc 8670 Wilshire Blvd, Suite 201 Los Angeles, CA 90211 USA Tel.:1-310-659-3880 Email: info@neurogeneration.com http://www.neurogeneration.com

Read the rest here:
NeuroGeneration Recruits Top Scientist To Direct New Division of Biotherapeutics and Drug Discovery In La Jolla, CA

To Read More: NeuroGeneration Recruits Top Scientist To Direct New Division of Biotherapeutics and Drug Discovery In La Jolla, CA
categoriaIPS Cell Therapy commentoComments Off on NeuroGeneration Recruits Top Scientist To Direct New Division of Biotherapeutics and Drug Discovery In La Jolla, CA | dataOctober 5th, 2012
Read All

International Stem Cell Corp Granted Key Patent for Liver Disease Program

By NEVAGiles23

CARLSBAD, CA--(Marketwire - Sep 25, 2012) - International Stem Cell Corporation ( OTCQB : ISCO ) (www.internationalstemcell.com) ("ISCO" or "the Company") a California-based biotechnology company, today announced that the United States Patent and Trademark Office (USPTO) has granted the Company a patent for a method of creating pure populations of definitive endoderm, precursor cells to liver and pancreas cells, from human pluripotent stem cells.This patent is a key element of ISCO's metabolic liver disease program and allows the Company to produce the necessary quantities of precursor cells in a more efficient and cost effective manner.

The patent, 8,268,621, adds to the Company's growing portfolio of proprietary technologies relating to the development of potential treatments for incurable diseases using human parthenogenetic Stem Cells (hpSC).Human parthenogenetic stem cells are unique pluripotent stem cells that offer the possibility to reduce the cost of health care while avoiding the ethical issues that surround the use of fertilized human embryos.Aside from the Company's current liver disease program, this new patented method can be used as a route to create pancreatic and endocrine cells that could be used in future studies of diabetes and other metabolic disorders.

ISCO currently has the largest collection of hpSC including cell lines which immune match the donor, as is the case with induced pluripotent stem cells (iPS), and cell lines which immune-match millions of individuals and potentially reduce tissue rejection issues.The Company is focusing its therapeutic development efforts on three clinical applications where cell and tissue therapy is already proven but where there currently is an insufficient supply of safe and efficacious cells: Parkinson's disease, inherited/metabolic liver diseases and corneal blindness.

About International Stem Cell Corporation

International Stem Cell Corporation is focused on the therapeutic applications of human parthenogenetic stem cells (hpSCs) and the development and commercialization of cell-based research and cosmetic products.ISCO's core technology, parthenogenesis, results in the creation of pluripotent human stem cells from unfertilized oocytes (eggs) hence avoiding ethical issues associated with the use or destruction of viable human embryos.ISCO scientists have created the first parthenogenetic, homozygous stem cell line that can be a source of therapeutic cells for hundreds of millions of individuals of differing genders, ages and racial background with minimal immune rejection after transplantation. hpSCs offer the potential to create the first true stem cell bank, UniStemCell. ISCO also produces and markets specialized cells and growth media for therapeutic research worldwide through its subsidiary Lifeline Cell Technology (www.lifelinecelltech.com), and stem cell-based skin care products through its subsidiary Lifeline Skin Care (www.lifelineskincare.com). More information is available at http://www.internationalstemcell.com.

To receive ongoing corporate communications via email, visit: http://www.b2i.us/irpass.asp?BzID=1468&to=ea&s=0

To like our Facebook page or follow us on Twitter for company updates and industry related news, visit: http://www.facebook.com/InternationalStemCellCorporation and http://www.twitter.com/intlstemcell

Safe harbor statement

Statements pertaining to anticipated developments, the potential use of technologies to develop therapeutic products and other opportunities for the company and its subsidiaries, along with other statements about the future expectations, beliefs, goals, plans, or prospects expressed by management constitute forward-looking statements. Any statements that are not historical fact (including, but not limited to statements that contain words such as "will," "believes," "plans," "anticipates," "expects" or "estimates") should also be considered to be forward-looking statements. Forward-looking statements involve risks and uncertainties, including, without limitation, risks inherent in the development and/or commercialization of potential products and the management of collaborations, regulatory approvals, need and ability to obtain future capital, application of capital resources among competing uses, and maintenance of intellectual property rights. Actual results may differ materially from the results anticipated in these forward-looking statements and as such should be evaluated together with the many uncertainties that affect the company's business, particularly those mentioned in the cautionary statements found in the company's Securities and Exchange Commission filings. The company disclaims any intent or obligation to update forward-looking statements.

Continued here:
International Stem Cell Corp Granted Key Patent for Liver Disease Program

To Read More: International Stem Cell Corp Granted Key Patent for Liver Disease Program
categoriaIPS Cell Therapy commentoComments Off on International Stem Cell Corp Granted Key Patent for Liver Disease Program | dataSeptember 30th, 2012
Read All

Page 20«..10..19202122..»


Copyright :: 2024