Leukemia & Lymphoma Society | Donate Today!

Do you know a hero or are one yourself? Submit your story for a chance to have your story included in the Community of Heroes for a common cause.

This field is required Select your state Alabama Alaska American Samoa Arizona Arkansas California Colorado Connecticut Delaware District of Columbia Florida Georgia Guam Hawaii Idaho Illinois Indiana Iowa Kansas Kentucky Louisiana Maine Maryland Massachusetts Michigan Minnesota Mississippi Missouri Montana Nebraska Nevada New Hampshire New Jersey New Mexico New York North Carolina North Dakota Northern Mariana Islands Ohio Oklahoma Oregon Pennsylvania Puerto Rico Rhode Island South Carolina South Dakota Tennessee Texas Utah Vermont Virginia Virgin Islands Washington West Virginia Wisconsin Wyoming Alberta British Columbia Manitoba New Brunswick Newfoundland Northwest Territories Nova Scotia Nunavut Ontario Prince Edward Island Quebec Saskatchewan Yukon

This field is required

I hereby authorize and permit The Leukemia & Lymphoma Society or its authorized agent, without compensation therefore, permission to photograph, publish, reproduce, record and use, with or without my name or the name of the person for whom I am the parent/guardian. This includes but is not limited to photographs, quotes and/or text, motion pictures, videotapes, website information or audiotapes of and/or by me or the person for whom I am the parent/guardian. I release The Leukemia & Lymphoma Society from any and all legal liability that may arise from the release of information requested. I agree that all text, photographs, motion pictures, negatives, prints and transparencies, videotapes and audio tapes made of and or by me or the person for whom I am the parent/guardian by or for The Leukemia & Lymphoma Society, shall be the exclusive property of The Leukemia & Lymphoma Society which in its sole discretion may use this material as it sees fit.

* This field is required

* Please fix the validation error messages in the Form

Your story was not submitted due to a connection error. Please try again later.

Continue reading here:
Leukemia & Lymphoma Society | Donate Today!

Supercourse: Epidemiology, the Internet, and Global Health

ABCDEFGHIJKLMNOPQRSTUVWXYZ

Academic research council

Achievements public health

Achievements public health

Acne therapeutic strategies

Acute coronary symptoms

Acute coronary syndromes

Adenoviridae and iridoviridae

Adherence hypertension treatment

Administration management medical organizations

Adolescent health risk behavior

Adolescents reproductive health

Adolescents reproductive health

Adverse drug reactions

Advocacy strategy planning

African sleeping sickness

Aids/ hiv current senario

Airborne contaminants

Air pollution armenia

Air pollution armenia

American heart association

Aminoglycosidearginine conjugates

Analytic epidemiology

Anaplasmosis taxonomic

Anemia family practice

Anger regulation interventions

Antimicrobial resistance

Antimicrobrial peptides

Antiretroviral agents

Assessing disease frequency

Assessment bioterrorism threat

Assessment nutritional

Assistive technology devices

Attack preparedness events

Avian influenza: zoonosis

Bacterial membrane vesicles

Bacterial vaginosis pregnancy

Bases of biostatistics

Behaviour medical sciences

Betaserk treatment stroke

Bias confounding chance

Bimaristans (hospitals) islamic

Binomial distribution

Biochemical system medicine

Biological challenges

Biological epidemiologic studies

Biostatistics

Biostatistics public health

Blood donors non-donors

Blood glucose normaization

Bmj triages manuscripts

Body fluid volume regulation

Bolonya declaration education

Bone marrow transplantation

Breast self examination

Bronchial asthma treatmen

Building vulnerability

Burden infectious diseases

Burnout in physicians

Cncer en mxico

Cancer survivorship research

Canine monocytic ehrlichiosis

Capability development

Capture-recapture techniques

Cardiology practice grenada

Cardiometabolic syndrome

Cardiopulmonary resuscitation

Cardio-respiratory illness

Cardiovascular disease

Cardiovascular disease black

Cardiovascular disease prevention

Cardiovascular diseases

Cardiovascular system

Carpal tunnel syndrome

Caseous lymphadenitis

Cause epidemiological approach

Central nervous system

Cervical cancer screening

Changing interpretations

Chemical weapon bioterrorism

Chemiosmotic paradigm

Chickenpox children pregnancy

Child health kazakhstan

Childhood asthma bedding.

Childhood asthma prevalence

Childhood diabetes mellitus

Childhood hearing impairment

Children september 11th attacks

China

Chinese herbal medicines

Chns hypertension control

Cholera global health

Cholesterol education program

Chronic disease management

Chronic fatigue syndrome

Chronic liver disease

Chronic lung diseases

Chronic noncommunicable diseases

Chronic obstructive pulmonary disease

Chronic pulmonary heart

Read more here:
Supercourse: Epidemiology, the Internet, and Global Health

Generation of Induced Pluripotent Stem Cells with …

Induced pluripotent stem cells (iPSCs) share many characteristics with embryonic stem cells, but lack ethical controversy. They provide vast opportunities for disease modeling, pathogenesis understanding, therapeutic drug development, toxicology, organ synthesis, and treatment of degenerative disease. However, this procedure also has many potential challenges, including a slow generation time, low efficiency, partially reprogrammed colonies, as well as somatic coding mutations in the genome. Pioneered by Shinya Yamanaka's team in 2006, iPSCs were first generated by introducing four transcription factors: Oct 4, Sox 2, Klf 4, and c-Myc (OSKM). Of those factors, Klf 4 and c-Myc are oncogenes, which are potentially a tumor risk. Therefore, to avoid problems such as tumorigenesis and low throughput, one of the key strategies has been to use other methods, including members of the same subgroup of transcription factors, activators or inhibitors of signaling pathways, microRNAs, epigenetic modifiers, or even differentiation-associated factors, to functionally replace the reprogramming transcription factors. In this study, we will mainly focus on the advances in the generation of iPSCs with substitutes for OSKM. The identification and combination of novel proteins or chemicals, particularly small molecules, to induce pluripotency will provide useful tools to discover the molecular mechanisms governing reprogramming and ultimately lead to the development of new iPSC-based therapeutics for future clinical applications.

Original post:
Generation of Induced Pluripotent Stem Cells with ...

Somatic cell – Wikipedia

A somatic (Greek: /soma = body) or vegetal cell is any biological cell forming the body of an organism; that is, in a multicellular organism, any cell other than a gamete, germ cell, gametocyte or undifferentiated stem cell.[1]

In contrast, gametes are cells that fuse during sexual reproduction, germ cells are cells that give rise to gametes, and stem cells are cells that can divide through mitosis and differentiate into diverse specialized cell types. For example, in mammals, somatic cells make up all the internal organs, skin, bones, blood and connective tissue, while mammalian germ cells give rise to spermatozoa and ova which fuse during fertilization to produce a cell called a zygote, which divides and differentiates into the cells of an embryo. There are approximately 220 types of somatic cells in the human body.[1]

Theoretically, these cells are not germ cells (the source of gametes), they never transmit to their descendants the mutations they have undergone. However, in sponges, non-differentiated somatic cells form the germ line and, in Cnidaria, differentiated somatic cells are the source of the germline.

The word "somatic" is derived from the Greek word sma, meaning "body".

As multicellularity evolved many times, sterile somatic cells did too. The evolution of an immortal germline producing specialized somatic cells involved the emergence of mortality, and can be viewed in its simplest version in volvocine algae.[2] Those species with a separation between sterile somatic cells and a germ line are called Weismannists. However, Weismannist development is relatively rare (e.g., vertebrates, arthropods, Volvox), as great part of species have the capacity for somatic embryogenesis (e.g., land plants, most algae, many invertebrates).[3][4]

Like all cells, somatic cells contain DNA arranged in chromosomes. If a somatic cell contains chromosomes arranged in pairs, it is called diploid and the organism is called a diploid organism. (The gametes of diploid organisms contain only single unpaired chromosomes and are called haploid.) Each pair of chromosomes comprises one chromosome inherited from the father and one inherited from the mother. For example, in humans, somatic cells contain 46 chromosomes organized into 23 pairs. By contrast, gametes of diploid organisms contain only half as many chromosomes. In humans, this is 23 unpaired chromosomes. When two gametes (i.e. a spermatozoon and an ovum) meet during conception, they fuse together, creating a zygote. Due to the fusion of the two gametes, a human zygote contains 46 chromosomes (i.e. 23 pairs).

However, a large number of species have the chromosomes in their somatic cells arranged in fours ("tetraploid") or even sixes ("hexaploid"). Thus, they can have diploid or even triploid germline cells. An example of this is the modern cultivated species of wheat, Triticum aestivum L., a hexaploid species whose somatic cells contain six copies of every chromatid.

In recent years, the technique of cloning whole organisms has been developed in mammals, allowing almost identical genetic clones of an animal to be produced. One method of doing this is called "somatic cell nuclear transfer" and involves removing the nucleus from a somatic cell, usually a skin cell. This nucleus contains all of the genetic information needed to produce the organism it was removed from. This nucleus is then injected into an ovum of the same species which has had its own genetic material removed. The ovum now no longer needs to be fertilized, because it contains the correct amount of genetic material (a diploid number of chromosomes). In theory, the ovum can be implanted into the uterus of a same-species animal and allowed to develop. The resulting animal will be a nearly genetically identical clone to the animal from which the nucleus was taken. The only difference is caused by any mitochondrial DNA that is retained in the ovum, which is different from the cell that donated the nucleus. In practice, this technique has so far been problematic, although there have been a few high-profile successes, such as Dolly the Sheep and, more recently, Snuppy, the first cloned dog. Somatic cells have also been collected in the practice of cryoconservation of animal genetic resources as a means of conserving animal genetic material, including to clone livestock.

Development of biotechnology has allowed for the genetic manipulation of somatic cells. This biotechnology deals with some ethical controversy in human genetic engineering.

Originally posted here:
Somatic cell - Wikipedia

Embryonic and Somatic Stem Cells, Whats the Difference?

Stem cells are undifferentiated cells found in body having capability to develop into different types of cells in the body. Whenever there is a cell division of stem cells, each stem cell has the ability to remain a stem cell or become another type of cell having more specialized functions. In body, stem cells can differentiate into other kind of body cells by having the following capabilities;

1) Proliferation; The stem cells are capable to renew even after long periods of inactivity and capable of self-renewal during cell division. 2) Unspecialized; They are unspecialized which later give arise to specialized cells. 3) Differentiation; They are able to differentiate them under special conditions and functions.

There are two kinds of stem cell:

1) Embryonic stem cells. Those stem cells are derived from developing embryo. These cells are mostly use for in-vitro fertilization. 2) Somatic stem cells. Commonly known as adult stem cell. They are defined according to their location within the body.

Embryonic stem cells Somatic Pluripotent. Non pluripotent. Can easily grow in culture Not that easy. A large number of cells are required for stem cell replacement therapies.

The somatic stem cells can be further classified into mesenchymal stem cells and hematopoietic stem cells. The somatic stem cells are found in specific region of organ known as stem cell niche. The organs from where stem cell niche are found are in brain, skeletal muscle, gut, liver, pancreas, bone marrow, ovarian epithelium teeth, and testis.

The Somatic stem cells have been demonstrated as; Hematopoietic stem cells; Give rise to all kinds of blood cells. Mesenchymal stem cells; Give rise to all kinds of bone cells. Neural stem cells; Give rise to all kinds of neuronal and non neuronal cells in brain. Epithelial stem cells; Give rise to different kinds of cells of digestive tract. Skin stem cells; give rise to all the epidermis and ketatinocytes of skin.

Read the original post:
Embryonic and Somatic Stem Cells, Whats the Difference?

Stem cell controversy – Wikipedia

The stem cell controversy is the consideration of the ethics of research involving the development, use, and destruction of human embryos. Most commonly, this controversy focuses on embryonic stem cells. Not all stem cell research involves the human embryos. For example, adult stem cells, amniotic stem cells, and induced pluripotent stem cells do not involve creating, using, or destroying human embryos, thus are minimally, if at all, controversial. Many less controversial sources of acquiring stem cells include using cells from the umbilical cord, breast milk, and bone marrow, which are not pluripotent.

For many decades, stem cells have played an important role in medical research, beginning in 1868 when Ernst Haeckel first used the phrase to describe the fertilized egg which eventually gestates into an organism. The term was later used in 1886 by William Sedgwick to describe the parts of a plant that grow and regenerate. Further work by Alexander Maximow and Leroy Stevens introduced the concept that stem cells are pluripotent, i.e. able to become many types of different cell. This significant discovery led to the first human bone marrow transplant by E. Donnal Thomas in 1968, which although successful in saving lives, has generated much controversy since. This has included the many complications inherent in stem cell transplantation (almost 200 allogeneic marrow transplants were performed in humans, with no long-term successes before the first successful treatment was made), through to more modern problems, such as how many cells are sufficient for engraftment of various types of hematopoietic stem cell transplants, whether older patients should undergo transplant therapy, and the role of irradiation-based therapies in preparation for transplantation.

The discovery of adult stem cells led scientists to develop an interest in the role of embroynic stem cells, and in separate studies in 1981 Gail Martin and Martin Evans derived pluripotent stem cells from the embryos of mice for the first time. This paved the way for Mario Capecchi, Martin Evans, and Oliver Smithies to create the first knockout mouse, ushering in a whole new era of research on human disease.

In 1998, James Thomson and Jeffrey Jones derived the first human embryonic stem cells, with even greater potential for drug discovery and therapeutic transplantation. However, the use of the technique on human embryos led to more widespread controversy as criticism of the technique now began from the wider non-scientific public who debated the moral ethics of questions concerning research involving human embryonic cells.

Since pluripotent stem cells have the ability to differentiate into any type of cell, they are used in the development of medical treatments for a wide range of conditions. Treatments that have been proposed include treatment for physical trauma, degenerative conditions, and genetic diseases (in combination with gene therapy). Yet further treatments using stem cells could potentially be developed due to their ability to repair extensive tissue damage.[1]

Great levels of success and potential have been realized from research using adult stem cells. In early 2009, the FDA approved the first human clinical trials using embryonic stem cells. These can become any cell type of the body, excluding placental cells. This ability is called pluripotency. Only cells from an embryo at the morula stage or earlier are truly totipotent, meaning that they are able to form all cell types including placental cells. Adult stem cells are generally limited to differentiating into different cell types of their tissue of origin. However, some evidence suggests that adult stem cell plasticity may exist, increasing the number of cell types a given adult stem cell can become.

Many of the debates surrounding human embryonic stem cells concern issues such as what restrictions should be made on studies using these types of cells. At what point does one consider life to begin? Is it just to destroy an embryo cell if it has the potential to cure countless numbers of patients? Political leaders are debating how to regulate and fund research studies that involve the techniques used to remove the embryo cells. No clear consensus has emerged. Other recent discoveries may extinguish the need for embryonic stem cells.[2]

Much of the criticism has been a result of religious beliefs, and in the most high-profile case, Christian US President George W Bush signed an executive order banning the use of federal funding for any cell lines other than those already in existence, stating at the time, "My position on these issues is shaped by deeply held beliefs," and "I also believe human life is a sacred gift from our creator."[3] This ban was in part revoked by his successor Barack Obama, who stated "As a person of faith, I believe we are called to care for each other and work to ease human suffering. I believe we have been given the capacity and will to pursue this research and the humanity and conscience to do so responsibly." [4]

Some stem cell researchers are working to develop techniques of isolating stem cells that are as potent as embryonic stem cells, but do not require a human embryo.

Foremost among these was the discovery in August 2006 that adult cells can be reprogrammed into a pluripotent state by the introduction of four specific transcription factors, resulting in induced pluripotent stem cells.[5] This major breakthrough won a Nobel Prize for the discoverers, Shinya Yamanaka and John Gurdon.[6]

In an alternative technique, researchers at Harvard University, led by Kevin Eggan and Savitri Marajh, have transferred the nucleus of a somatic cell into an existing embryonic stem cell, thus creating a new stem cell line.[7]

Researchers at Advanced Cell Technology, led by Robert Lanza and Travis Wahl, reported the successful derivation of a stem cell line using a process similar to preimplantation genetic diagnosis, in which a single blastomere is extracted from a blastocyst.[8] At the 2007 meeting of the International Society for Stem Cell Research (ISSCR),[9] Lanza announced that his team had succeeded in producing three new stem cell lines without destroying the parent embryos. "These are the first human embryonic cell lines in existence that didn't result from the destruction of an embryo." Lanza is currently in discussions with the National Institutes of Health to determine whether the new technique sidesteps U.S. restrictions on federal funding for ES cell research.[10]

Anthony Atala of Wake Forest University says that the fluid surrounding the fetus has been found to contain stem cells that, when used correctly, "can be differentiated towards cell types such as fat, bone, muscle, blood vessel, nerve and liver cells". The extraction of this fluid is not thought to harm the fetus in any way. He hopes "that these cells will provide a valuable resource for tissue repair and for engineered organs, as well".[11]

The status of the human embryo and human embryonic stem cell research is a controversial issue, as with the present state of technology, the creation of a human embryonic stem cell line requires the destruction of a human embryo. Most of these embryos are discarded. Stem cell debates have motivated and reinvigorated the pro-life movement, whose members are concerned with the rights and status of the embryo as an early-aged human life. They believe that embryonic stem cell research instrumentalizes and violates the sanctity of life and is tantamount to murder.[12] The fundamental assertion of those who oppose embryonic stem cell research is the belief that human life is inviolable, combined with the belief that human life begins when a sperm cell fertilizes an egg cell to form a single cell. The view of those in favor is that these embryos would otherwise be discarded, and if used as stem cells, they can survive as a part of a living human being.

A portion of stem cell researchers use embryos that were created but not used in in vitro fertility treatments to derive new stem cell lines. Most of these embryos are to be destroyed, or stored for long periods of time, long past their viable storage life. In the United States alone, an estimated at least 400,000 such embryos exist.[13] This has led some opponents of abortion, such as Senator Orrin Hatch, to support human embryonic stem cell research.[14] See also embryo donation.

Medical researchers widely report that stem cell research has the potential to dramatically alter approaches to understanding and treating diseases, and to alleviate suffering. In the future, most medical researchers anticipate being able to use technologies derived from stem cell research to treat a variety of diseases and impairments. Spinal cord injuries and Parkinson's disease are two examples that have been championed by high-profile media personalities (for instance, Christopher Reeve and Michael J. Fox, who have lived with these conditions, respectively). The anticipated medical benefits of stem cell research add urgency to the debates, which has been appealed to by proponents of embryonic stem cell research.

In August 2000, The U.S. National Institutes of Health's Guidelines stated:

...research involving human pluripotent stem cells...promises new treatments and possible cures for many debilitating diseases and injuries, including Parkinson's disease, diabetes, heart disease, multiple sclerosis, burns and spinal cord injuries. The NIH believes the potential medical benefits of human pluripotent stem cell technology are compelling and worthy of pursuit in accordance with appropriate ethical standards.[15]

In 2006, researchers at Advanced Cell Technology of Worcester, Massachusetts, succeeded in obtaining stem cells from mouse embryos without destroying the embryos.[16] If this technique and its reliability are improved, it would alleviate some of the ethical concerns related to embryonic stem cell research.

Another technique announced in 2007 may also defuse the longstanding debate and controversy. Research teams in the United States and Japan have developed a simple and cost-effective method of reprogramming human skin cells to function much like embryonic stem cells by introducing artificial viruses. While extracting and cloning stem cells is complex and extremely expensive, the newly discovered method of reprogramming cells is much cheaper. However, the technique may disrupt the DNA in the new stem cells, resulting in damaged and cancerous tissue. More research will be required before noncancerous stem cells can be created.[17][18][19][20]

Update article to include 2009/2010 current stem cell usages in clinical trials.[21][22] The planned treatment trials will focus on the effects of oral lithium on neurological function in people with chronic spinal cord injury and those who have received umbilical cord blood mononuclear cell transplants to the spinal cord. The interest in these two treatments derives from recent reports indicating that umbilical cord blood stem cells may be beneficial for spinal cord injury and that lithium may promote regeneration and recovery of function after spinal cord injury. Both lithium and umbilical cord blood are widely available therapies that have long been used to treat diseases in humans.

This argument often goes hand-in-hand with the utilitarian argument, and can be presented in several forms:

This is usually presented as a counter-argument to using adult stem cells as an alternative that does not involve embryonic destruction.

This argument is used by opponents of embryonic destruction, as well as researchers specializing in adult stem cell research.

Pro-life supporters often claim that the use of adult stem cells from sources such as umbilical cord blood has consistently produced more promising results than the use of embryonic stem cells.[30] Furthermore, adult stem cell research may be able to make greater advances if less money and resources were channeled into embryonic stem cell research.[31]

In the past, it has been a necessity to research embryonic stem cells and in doing so destroy them for research to progress.[32] As a result of the research done with both embryonic and adult stem cells, new techniques may make the necessity for embryonic cell research obsolete. Because many of the restrictions placed on stem cell research have been based on moral dilemmas surrounding the use of embryonic cells, there will likely be rapid advancement in the field as the techniques that created those issues are becoming less of a necessity.[33] Many funding and research restrictions on embryonic cell research will not impact research on IPSCs (induced pluripotent stem cells) allowing for a promising portion of the field of research to continue relatively unhindered by the ethical issues of embryonic research.[34]

Adult stem cells have provided many different therapies for illnesses such as Parkinson's disease, leukemia, multiple sclerosis, lupus, sickle-cell anemia, and heart damage[35] (to date, embryonic stem cells have also been used in treatment),[36] Moreover, there have been many advances in adult stem cell research, including a recent study where pluripotent adult stem cells were manufactured from differentiated fibroblast by the addition of specific transcription factors.[37] Newly created stem cells were developed into an embryo and were integrated into newborn mouse tissues, analogous to the properties of embryonic stem cells.

Austria, Denmark, France, Germany, and Ireland do not allow the production of embryonic stem cell lines,[38] but the creation of embryonic stem cell lines is permitted in Finland, Greece, the Netherlands, Sweden, and the United Kingdom.[38]

In 1973, Roe v. Wade legalized abortion in the United States. Five years later, the first successful human in vitro fertilization resulted in the birth of Louise Brown in England. These developments prompted the federal government to create regulations barring the use of federal funds for research that experimented on human embryos. In 1995, the NIH Human Embryo Research Panel advised the administration of President Bill Clinton to permit federal funding for research on embryos left over from in vitro fertility treatments and also recommended federal funding of research on embryos specifically created for experimentation. In response to the panel's recommendations, the Clinton administration, citing moral and ethical concerns, declined to fund research on embryos created solely for research purposes,[39] but did agree to fund research on leftover embryos created by in vitro fertility treatments. At this point, the Congress intervened and passed the Dickey Amendment in 1995 (the final bill, which included the Dickey Amendment, was signed into law by Bill Clinton) which prohibited any federal funding for the Department of Health and Human Services be used for research that resulted in the destruction of an embryo regardless of the source of that embryo.

In 1998, privately funded research led to the breakthrough discovery of human embryonic stem cells (hESC). This prompted the Clinton administration to re-examine guidelines for federal funding of embryonic research. In 1999, the president's National Bioethics Advisory Commission recommended that hESC harvested from embryos discarded after in vitro fertility treatments, but not from embryos created expressly for experimentation, be eligible for federal funding. Though embryo destruction had been inevitable in the process of harvesting hESC in the past (this is no longer the case[40][41][42][43]), the Clinton administration had decided that it would be permissible under the Dickey Amendment to fund hESC research as long as such research did not itself directly cause the destruction of an embryo. Therefore, HHS issued its proposed regulation concerning hESC funding in 2001. Enactment of the new guidelines was delayed by the incoming George W. Bush administration which decided to reconsider the issue.

President Bush announced, on August 9, 2001, that federal funds, for the first time, would be made available for hESC research on currently existing embryonic stem cell lines. President Bush authorized research on existing human embryonic stem cell lines, not on human embryos under a specific, unrealistic timeline in which the stem cell lines must have been developed. However, the Bush Administration chose not to permit taxpayer funding for research on hESC cell lines not currently in existence, thus limiting federal funding to research in which "the life-and-death decision has already been made".[44] The Bush Administration's guidelines differ from the Clinton Administration guidelines which did not distinguish between currently existing and not-yet-existing hESC. Both the Bush and Clinton guidelines agree that the federal government should not fund hESC research that directly destroys embryos.

Neither Congress nor any administration has ever prohibited private funding of embryonic research. Public and private funding of research on adult and cord blood stem cells is unrestricted.

In April 2004, 206 members of Congress signed a letter urging President Bush to expand federal funding of embryonic stem cell research beyond what Bush had already supported.

In May 2005, the House of Representatives voted 238194 to loosen the limitations on federally funded embryonic stem-cell researchby allowing government-funded research on surplus frozen embryos from in vitro fertilization clinics to be used for stem cell research with the permission of donorsdespite Bush's promise to veto the bill if passed.[45] On July 29, 2005, Senate Majority Leader William H. Frist (R-TN), announced that he too favored loosening restrictions on federal funding of embryonic stem cell research.[46] On July 18, 2006, the Senate passed three different bills concerning stem cell research. The Senate passed the first bill (the Stem Cell Research Enhancement Act) 6337, which would have made it legal for the federal government to spend federal money on embryonic stem cell research that uses embryos left over from in vitro fertilization procedures.[47] On July 19, 2006 President Bush vetoed this bill. The second bill makes it illegal to create, grow, and abort fetuses for research purposes. The third bill would encourage research that would isolate pluripotent, i.e., embryonic-like, stem cells without the destruction of human embryos.

In 2005 and 2007, Congressman Ron Paul introduced the Cures Can Be Found Act,[48] with 10 cosponsors. With an income tax credit, the bill favors research upon nonembryonic stem cells obtained from placentas, umbilical cord blood, amniotic fluid, humans after birth, or unborn human offspring who died of natural causes; the bill was referred to committee. Paul argued that hESC research is outside of federal jurisdiction either to ban or to subsidize.[49]

Bush vetoed another bill, the Stem Cell Research Enhancement Act of 2007,[50] which would have amended the Public Health Service Act to provide for human embryonic stem cell research. The bill passed the Senate on April 11 by a vote of 63-34, then passed the House on June 7 by a vote of 247176. President Bush vetoed the bill on July 19, 2007.[51]

On March 9, 2009, President Obama removed the restriction on federal funding for newer stem cell lines. [52] Two days after Obama removed the restriction, the president then signed the Omnibus Appropriations Act of 2009, which still contained the long-standing Dickey-Wicker provision which bans federal funding of "research in which a human embryo or embryos are destroyed, discarded, or knowingly subjected to risk of injury or death;"[53] the Congressional provision effectively prevents federal funding being used to create new stem cell lines by many of the known methods. So, while scientists might not be free to create new lines with federal funding, President Obama's policy allows the potential of applying for such funding into research involving the hundreds of existing stem cell lines as well as any further lines created using private funds or state-level funding. The ability to apply for federal funding for stem cell lines created in the private sector is a significant expansion of options over the limits imposed by President Bush, who restricted funding to the 21 viable stem cell lines that were created before he announced his decision in 2001.[54] The ethical concerns raised during Clinton's time in office continue to restrict hESC research and dozens of stem cell lines have been excluded from funding, now by judgment of an administrative office rather than presidential or legislative discretion.[55]

In 2005, the NIH funded $607 million worth of stem cell research, of which $39 million was specifically used for hESC.[56]Sigrid Fry-Revere has argued that private organizations, not the federal government, should provide funding for stem-cell research, so that shifts in public opinion and government policy would not bring valuable scientific research to a grinding halt.[57]

In 2005, the State of California took out $3 billion in bond loans to fund embryonic stem cell research in that state.[58]

China has one of the most permissive human embryonic stem cell policies in the world. In the absence of a public controversy, human embryo stem cell research is supported by policies that allow the use of human embryos and therapeutic cloning.[59]

According to Rabbi Levi Yitzchak Halperin of the Institute for Science and Jewish Law in Jerusalem, embryonic stem cell research is permitted so long as it has not been implanted in the womb. Not only is it permitted, but research is encouraged, rather than wasting it.

However in order to remove all doubt [as to the permissibility of destroying it], it is preferable not to destroy the pre-embryo unless it will otherwise not be implanted in the woman who gave the eggs (either because there are many fertilized eggs, or because one of the parties refuses to go on with the procedurethe husband or wifeor for any other reason). Certainly it should not be implanted into another woman.... The best and worthiest solution is to use it for life-saving purposes, such as for the treatment of people that suffered trauma to their nervous system, etc.

Similarly, the sole Jewish majority state, Israel, permits research on embryonic stem cells.

The Catholic Church opposes human embryonic stem cell research calling it "an absolutely unacceptable act." The Church supports research that involves stem cells from adult tissues and the umbilical cord, as it "involves no harm to human beings at any state of development."[60]

The Southern Baptist Convention opposes human embryonic stem cell research on the grounds that "Bible teaches that human beings are made in the image and likeness of God (Gen. 1:27; 9:6) and protectable human life begins at fertilization."[61] However, it supports adult stem cell research as it does "not require the destruction of embryos."[61]

The United Methodist Church opposes human embryonic stem cell research, saying, "a human embryo, even at its earliest stages, commands our reverence."[62] However, it supports adult stem cell research, stating that there are "few moral questions" raised by this issue.[62]

The Assemblies of God opposes human embryonic stem cell research, saying, it "perpetuates the evil of abortion and should be prohibited."[63]

The religion of Islam favors the stance that scientific research and development in terms of stem cell research is allowed as long as it benefits society while using the least amount of harm to the subjects. "Stem cell research is one of the most controversial topics of our time period and has raised many religious and ethical questions regarding the research being done. With there being no true guidelines set forth in the Qur'an against the study of biomedical testing, Muslims have adopted any new studies as long as the studies do not contradict another teaching in the Qur'an. One of the teachings of the Qur'an states that Whosoever saves the life of one, it shall be if he saves the life of humankind (5:32), it is this teaching that makes stem cell research acceptable in the Muslim faith because of its promise of potential medical breakthrough."[64]

The First Presidency of The Church of Jesus Christ of Latter-day Saints "has not taken a position regarding the use of embryonic stem cells for research purposes. The absence of a position should not be interpreted as support for or opposition to any other statement made by Church members, whether they are for or against embryonic stem cell research.[65]

View post:
Stem cell controversy - Wikipedia

Platelet-rich plasma – Wikipedia

Platelet-rich plasma (abbreviation: PRP) is blood plasma that has been enriched with platelets. As a concentrated source of autologous platelets, PRP contains several different growth factors and other cytokines that can stimulate healing of soft tissue. Platelet-rich plasma therapy is an old therapy and used extensively in specialities of dermatology, orthopedics and dentistry. Platelet rich plasma therapy utilizes growth factors present in alpha granules of platelets in an autologous manner. Main indications in dermatology for PRP are androgenetic alopecia, wound healing, face rejuvenation etc. For preparation of PRP, various protocols are used and no standard protocol exists but main principles essentially involve concentrating platlets in a concentration of 35 times the physiological value and then injecting this concentrated plasma in the tissue where healing or effect is desired.[1] As of 2016, no large-scale randomized controlled trials have confirmed the efficacy of PRP as a treatment for musculoskeletal or nerve injuries, the accelerated healing of bone grafts, or the reduction of androgenic hair loss.

PRP was first developed in the 1970s and first used in Italy in 1987 in an open heart surgery procedure.[citation needed] PRP therapy began gaining popularity[where?] in the mid 1990s.[citation needed] It has since been applied to many different medical fields such as cosmetic surgery, dentistry, sports medicine and pain management.[citation needed]

The number of peer reviewed publications studying the PRP's efficacy has increased dramatically since 2007.[2]

The efficacy of certain growth factors in healing various injuries and the concentrations of these growth factors found within PRP are the theoretical basis for the use of PRP in tissue repair.[3] The platelets collected in PRP are activated by the addition of thrombin and calcium chloride, which induces the release of the mentioned factors from alpha granules. The growth factors and other cytokines present in PRP include:[3][4]

As of 2009[update] there have been two PRP preparation methods approved by the U.S. Food and Drug Administration.[5] Both processes involve the collection of the patient's whole blood (that is anticoagulated with citrate dextrose) before undergoing two stages of centrifugation (TruPRP) (Harvest) (Pure PRP) designed to separate the PRP aliquot from platelet-poor plasma and red blood cells.[5] In humans, the typical baseline blood platelet count is approximately 200,000 per L; therapeutic PRP concentrates the platelets by roughly five-fold.[6] There is broad variability in the production of PRP by various concentrating equipment and techniques.[7][8][9]

In humans, PRP has been investigated and used as a clinical tool for several types of medical treatments, including nerve injury,[4] chronic tendinitis,[10][11][12]plantar fasciitis,[13]osteoarthritis,[14]cardiac muscle injury,[15] and androgenic alopecia,[16][17] for bone repair and regeneration,[18] in plastic surgery,[19]colorectal surgery[20] and oral surgery[21]

PRP has received attention in the popular media as a result of its use in treating sports injuries in professional athletes.[22][23][24][25]

The cost of a PRP treatment in the U.S. has been quoted as $1000 out-of-pocket expenses, as it is usually not covered by health insurance.[25]

PRP has been used experimentally in the treatment of empty nose syndrome[26]

As of 2016[update] results of basic science and preclinical trials have not yet been confirmed in large-scale randomized controlled trials. A 2009 systematic review of the scientific literature found there were few randomized controlled trials that adequately evaluated the safety and efficacy of PRP treatments and concluded that PRP was "a promising, but not proven, treatment option for joint, tendon, ligament, and muscle injuries".[27]

A 2010 Cochrane analysis on PRP use in sinus lifts during dental implant placement found no evidence that PRP offered any benefit.[21]

As of 2011, PRP use for nerve injury and sports medicine has produced "promising" but "inconsistent" results in early trials.[4]

A 2013 review stated more evidence was needed to determine PRP's effectiveness for hair regrowth.[28]

A 2014 Cochrane analysis for PRT use to treat musculoskeletal injuries found very weak (very low quality) evidence for a decrease in pain in the short term, up to three months and no difference in function in the short, medium or long term. There was weak evidence that suggested that harm occurred at comparable, low rates in treated and untreated people.[29]

A 2016 systematic review and meta-analysis of randomized controlled clinical trials for PRP use to augment bone graft found only one study reporting a significant difference in bone augmentation, while four studies found no significant difference.[30]

Since 2004, proponents of PRP therapy have argued that negative clinical results are associated with poor-quality PRP produced by inadequate single spin devices. The fact that most gathering devices capture a percentage of a given thrombocyte count could bias results, because of inter-individual variability in the platelet concentration of human plasma and more would not necessarily be better.[6] The variability in platelet concentrating techniques may alter platelet degranulation characteristics that could affect clinical outcomes.[4]

Platelet-rich plasma is used in horses for treatment of equine lameness due to tendon and ligament injury, wounds, fractures, bone cysts, and osteoarthritis.[citation needed]

Some concern exists as to whether PRP treatments violate anti-doping rules.[3] As of 2010 it was not clear if local injections of PRP could have a systemic impact on circulating cytokine levels, affecting doping tests and whether PRP treatments have systemic anabolic effects or affect performance.[3] In January 2011, the World Anti-Doping Agency removed intramuscular injections of PRP from its prohibitions after determining that there is a "lack of any current evidence concerning the use of these methods for purposes of performance enhancement".[31]

According to the Baltimore Sun, Zach Britton had PRP injections in his left shoulder in March 2012, Orioles first baseman Chris Davis underwent two PRP injections to speed the healing and recovery of an oblique injury in April 2014, and Dylan Bundy had the procedure in April before undergoing Tommy John surgery in June 2014.[32]

Continued here:
Platelet-rich plasma - Wikipedia

Induced pluripotent stem cells and Parkinson’s disease …

Review Authors

Correspondence: T. Wang, Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Road, Wuhan 430022, Hubei, China; E-mail: wangtaowh@hust.edu.cn

Many neurodegenerative disorders, such as Parkinson's disease (PD), are characterized by progressive neuronal loss in different regions of the central nervous system, contributing to brain dysfunction in the relevant patients. Stem cell therapy holds great promise for PD patients, including with foetal ventral mesencephalic cells, human embryonic stem cells (hESCs) and human induced pluripotent stem cells (hiPSCs). Moreover, stem cells can be used to model neurodegenerative diseases in order to screen potential medication and explore their mechanisms of disease. However, related ethical issues, immunological rejection and lack of canonical grafting protocols limit common clinical use of stem cells. iPSCs, derived from reprogrammed somatic cells, provide new hope for cell replacement therapy. In this review, recent development in stem cell treatment for PD, using hiPSCs, as well as the potential value of hiPSCs in modelling for PD, have been summarized for application of iPSCs technology to clinical translation for PD treatment.

adenoviral iPSCs

cluster of differentiation

cynomolgus macaque

dopamine

dopamine transporter

deep brain stimulation

flow cytometric analysis

glucocerebrosidase

human embryonic stem cells

human induced pluripotent stem cells

Lewy bodies

leucine-rich repeat kinase 2

mouse embryonic fibroblasts

major histocompatibility complex

mitochondrial DNA

neural stem cells

Parkinson's disease

subgranular zone

-synuclein

substantia nigra pars compacta

subventricular zone

valproic acid

zinc finger nuclease

zonisamide

Parkinson's disease (PD) is the second most common neurodegenerative disorder, and concerns progressive loss of dopaminergic (DA) neurons in the substantia nigra pars compacta (SNpc) of the midbrain [1]. The crucial pathological feature of PD is presence of Lewy bodies (LBs), which are abnormal aggregates of -synuclein (SNCA) protein. Reported standardized incidence rates of PD are 818 per 100 000 person-years worldwide [2]. In China, prevalence of PD for those aged 65 years is 1.7% [3]. PD patients suffer from motor symptoms such as rest tremor, bradykinesia, rigidity and abnormal gait. Non-motor symptoms, such as olfactory dysfunction, psychiatric changes and sleep disorders, further impair PD patients' quality of life. Up to now, multiple factors have been found to involve pathogenesis of PD, including genetic susceptibility, environmental toxins, interruption of autophagy, neuroinflammation and most importantly advancing age. Although the precise mechanisms underlying the pathogenesis of PD are not well understood, interactions within these pathogenic factors give rise to loss of DA neurons within the SNpc. Unfortunately, current pharmacological and surgical treatments provide only insufficient symptomatic relief, but cannot reverse nor slow down the underlying loss of midbrain DA neurons. Stem cell transplantation, however, holds great promise in the treatment of PD.

Neural stem cells (NSCs) provide a potential endogenous source for neuron replacement therapy in neurodegenerative disorders such as PD. One of the most important essential features of NSCs is their proliferation potential. It has already been indicated that NSCs can differentiate directly into DA neurons and Suksuphew and Noisa have shown that they have high possibility for producing two undifferentiated daughter cells at early stages of development (symmetric division), and later cell division for production of differentiated neurons plus glial cells (asymmetric division) [4]. A further feature of NSCs' multipotency is their potential to differentiate into astrocytes and oligodendrocytes, as well as into neurons [5, 6]. NSCs in the subventricular zone (SVZ) can differentiate into olfactory neurons, while those of the subgranular zone (SGZ) can differentiate into granular neurons of the dentate gyrus [7]. Furthermore, when implanted into developing eyes, hippocampal NSCs have exhibited several morphological and immunological properties of retinal cells, including photoreceptors [7]. Differentiation of adult NSCs can be influenced by their local environment as well as by intrinsic programmes [8].

Human ESCs were the first human stem cells to be identified and cultured, by Thomson et al. in 1998 [9], and were proven at that time to be self-renewing and pluripotent. These properties indicated hESCs as having great promise for cell transplantation therapy. However, ethical concerns arose immediately as generation of hESCs requires destruction of the fertilized human embryo. A further significant problem with transplanting stem cells is associated with immunological rejection after transplantation of specific cells derived from allogeneic hESCs. In 2006, Yamanaka et al. reported generation of ES-like pluripotent stem cells from somatic fibroblasts, the so-called iPSCs [10]. Since then, many methods have been explored to generate hiPSCs from a wide variety of easily accessible source tissues, including skin, adipose and blood cells [11-14]. Unlike hESCs, there are no ethical issues preventing use of iPSCs. Refinement of reprogramming methods now allows for iPSC generation without genomic integration of reprogramming factors, using expression plasmids, non-integrating viruses, recombinant proteins, small molecules and synthetically modified mRNAs or miRNAs [15]. Here, we review the existing iPSC-based models and treatments, with particular emphasis on PD, and explore the challenges associated with cell therapies using iPSCs-derived DA neurons, which have thus far hindered expansion of this research.

Cell replacement therapy with foetal ventral midbrain (VM) DA neurons has been shown, in some ways, to be beneficial to PD patients. Dopaminergic neurons lost in PD are primarily of the VM, and VM DA neurons arise from floor plate cells during embryonic development. The earliest study of DA neuron differentiation from mouse ESCs was performed by Lee et al. in 2000 [16]. This group generated CNS progenitor populations from ESCs, expanded the cells and promoted their differentiation into dopaminergic and serotonergic neurons in the presence of mitogens and specific signalling molecules. Their differentiation involved a number of steps: generation of embryoid bodies (EBs) without retinoic acid (RA) treatment in a serum-containing medium, use of a defined medium to select for CNS stem cells, proliferation of CNS stem cells in the presence of mitogen, basic fibroblast growth factor (bFGF), and differentiation of the stem cells by removal of the mitogen in serum-free medium [16]. Finally, the differentiation medium consisted of N2 medium supplemented with laminin, cAMP and ascorbic acid (AA). Sonic hedgehog (SHH), FGF8 and AA also enhanced differentiation to DA fate, and increased yield of ES-derived TH+ neurons. The cells were incubated under differentiation conditions for 615 days at the last stage, in order to increase numbers of TH+ neurons and DA level [16]. While this protocol succeeded in generating DA neurons at relatively high efficiency, extensive studies in Parkinsonian animals are needed to further assess complete function and safety of ESC-derived DA neurons in vivo [16]. Efficiency and purification of generated cell populations also needs to be improved by genetic methods.

After some months, Kawasaki and colleagues introduced an efficient method for generating neurons from ESCs by using PA6-derived stromal cell-derived inducing activity (SDIA) in a serum-free condition requiring neither EBs nor RA treatment [17]. High proportions of TH+ neurons producing DA were obtained from SDIA-treated ESCs. When transplanted, SDIA-induced dopaminergic neurons integrated into the mouse striatum and remained positive for TH expression [17]. In accordance with Lee's group, Kawasaki et al. also avoided using RA in their experiment, as RA seemed to perturb neural patterning and neuronal identities in EBs, as a strong teratogen. Efficiency of DA neuron induction in the SDIA method is as high as maximum efficiency (~30%) obtained by Lee's method with SHH, FGF8 and ascorbate treatment [17]. Neural induction by SDIA provided a new powerful tool for both basic neuroscience research and therapeutic applications.

Low efficiency of generation of DA neurons from primary cultures of foetal neonatal cells, or adult stem cells, limits their therapeutic potential as donor cells [18]. In effort to improve efficiency of DA neuron generation, survival and maturation in vitro, Kim et al. used a cytomegalovirus plasmid (pCMV) driving expression of rat Nurr1 complementary DNA modified to establish stable Nurr1 ESC lines [18]. Nurr1 ESCs raised the proportion of TH+ neurons up to 78%, combined with their previous five-stage differentiation method [16, 18]. They also demonstrated that these DA neurons from ESCs could functionally integrate into host tissue as well as lead to recovery in a rodent model of PD [18].

These results have subsequently been replicated using hESCs with some modifications, but efficiency was not satisfactory [19-21]. Perrier et al. have reported that co-culture of hESCs on MS5 stroma can yield highly efficient differentiation into midbrain DA neurons [22]. Neural differentiation of hESCs was induced by means of co-culture on MS5, MS5-Wnt or S2 stroma at comparable efficiencies. Growth factors were added in various combinations and at various time points. Rosette structures were harvested mechanically from feeder layers on day 28 of differentiation and gently replanted on 15 g/ml polyornithine/1 g/ml laminin-coated culture dishes in N2 medium supplemented with SHH, FGF8, AA and BDNF. These cells were resuspended in N2 medium, and replated again on to polyornithine/laminin-coated culture dishes in the presence of SHH, FGF8, AA and BDNF. After additional 79 days culture, cells were found to have differentiated in the absence of SHH and FGF8 but in the presence of BDNF, glial cell line-derived neurotrophic factor, transforming growth factor type 3, dibutyryl cAMP and AA [22]. The workers observed that exposure to SHH and FGF8 from day 12 to day 20 differentiation, followed by differentiation in the presence of AA and BDNF, resulted in 3-fold increase in TH+ cells. Up to 79% of all the neurons express TH, the rate-limiting enzyme in the synthesis of DA. In addition to TH expression, cells in these cultures expressed key markers associated with normal midbrain DA neurons. However, the high-yield midbrain DA neuron derivation protocol reported here need to be transplantated into pre-clinical animal models of PD [22]. Beyond this, cell survival and long-term maintenance of phenotype are essential parameters for testing in vivo.

In 2008, a group of Korean scientists reported a method for differentiating hESCs into functional TH+ neurons, with up to near 86% total hESC-derived neurons, the highest purity ever reported [23]. The unique feature of their protocol was generation of pure spherical neural masses (SNMs). These SNMs could be expanded for long periods without losing their differentiation capability and could be coaxed into DA neurons efficiently within a relatively short time (approximately 2 weeks) when needed. SNM culture and DA neuron derivation from the SNMs did not need feeder cells, which reduced risks of contamination by unwanted cells and pathogens. More importantly, their hESC-derived DA neurons induced clear behavioural recovery after transplantation in a Parkinsonian rat model, indicating their functionality in vivo [23].

It has been reported that bone marrow mesenchymal stem cells (BMSCs) can differentiate into not only osteogenic, adipogenic, chondrogenic cells, but also into other lineages including myogenic, hepatic and neurogenic cells [24]. Furthermore, they are inducible to differentiate into cells with the DA neuronal phenotype suggested by expression of TH, DAT markers, as well as synthesis and secretion of DA after appropriate stimuli [25]. Previous studies have shown that human BMSC engraftment can alleviate motor dysfunction in Parkinsonian animal models, but with limited efficacy and but few engrafted cells surviving. Our team transplanted equal amounts of hBMSCs into hemi-lesioned Parkinsonian rats with supplementation of bFGF, to assess whether a combination of bFGF and hBMSC therapy would enhance treatment effectiveness in PD rat models [26]. As a result, bFGF promoted hBMSCs to transdifferentiate towards neural-like lineages in vitro [26]. In addition, hBMSC transplantation alleviated motor functional asymmetry, and prevented DA neurons from loss in the PD model, while bFGF administration enhanced neurodifferentiation capacity and therapeutic effect [26].

Similar strategies have been applied for differentiating hiPSCs into DA neurons. Cooper et al. postulated that a major limitation for experimental studies of current ESC/iPSC differentiation protocols, was lack of VM DA neurons of stable phenotype, as defined by expression marker code FOXA2/TH/-tubulin [27]. They demonstrated a combination of three modifications that were required to produce VM DA neurons. First, early and specific exposure to low-dose RA improved regional identity of neural progenitor cells derived from pluripotent stem cells. Secondly, a high activity form of human SHH established a sizeable FOXA2+ neural progenitor cell population in vitro. Thirdly, early exposure to FGF8a, rather than FGF8b, and WNT1 were required for robust differentiation of the FOXA2+ floor plate-like human neural progenitor cells into FOXA2+ DA neurons [27]. FOXA2+ DA neurons were also generated when this protocol was adapted to feeder-free conditions. In summary, their new human ESC and iPSC differentiation protocol can generate human VM DA neurons as required for relevant new bioassays, drug discovery and cell-based therapies for PD [27].

The majority of PD cases are sporadic with unknown cause. Age, oxidative stress, toxin and environmental factors are risk factors [2], and remaining 10% is familial PD, where several causative genes have been identified [28]. Before stem cell modelling appeared, the most used cell or animal models for PD were generated with toxins such as rotenone, 6-OHDA, MPTP or genetic models. The relationship between MPTP and PD was found in a cluster of young drug addicts, by Davis et al. in 1979 [29]. MPTP easily crosses the bloodbrain barrier (BBB) where it is oxidized in glial cells into MPP+. MPP+ competes with DA for the DA transporter and after entering neurons, it exerts its toxic effect by inactivating complex I of the ETC [30]. MPTP is commonly used to model for PD in primates and rodents in that the drug kills dopaminergic neurons, allowing researchers to study neuronal circuitry with reduced dopaminergic involvement [31]. Many workers have demonstrated that MPTP administration is able to reproduce most, but not all, the clinical and pathological hallmarks of PD in monkeys [32-34] and, at least degeneration of dopaminergic neurons, in mice [35]. Similar to MPTP, the pesticide rotenone disrupts complex I function of mitochondria [36]. Our team has demonstrated that rotenone models for PD appear to mimic most clinical features of idiopathic PD and recapitulate the slow and progressive loss of DA neurons and LB formation in the nigral-striatal system [36]. Both MPTP and rotenone have been important for establishment of PD animal models. However, while they promote dopaminergic neuron death with associated motor impairment, their side effects and lack of specificity are major drawbacks [31]. 6-OHDA, a selective catecholaminergic neurotoxin, is used to generate lesions in the nigrostriatal DA neurons in rats [37]. Unlike MPTP, 6-OHDA cannot cross the BBB. So, 6-OHDA must be injected into the SNc, medial forebrain bundle or striatum, to induce Parkinsonism, rather than systemic administration [38]. Intrastriatal injection of 6-OHDA causes progressive retrograde neuronal degeneration in the SNc and VTA [39, 40]. Genetic models provide us with better understanding of underlying genetic forms of PD, even though their pathological and behavioural phenotypes are often quite different from the human condition [41]. Many genetic variant models, including SNCA, LRRK2, PINK, Parkin, DJ-1 and Glucocerebrosidase (GBA), have been generated to explore inherent mechanisms in PD [42-45]. The well-established genetic models are able to interpret pathogenesis of only 510% familial PD, however, without replicating the entire genetic background of the patients in vitro [46]. Moreover, differences between species in displaying neurodegenerative phenotypes make it difficult to extrapolate results obtained from animal models to humans [47]. The discovery of iPSCs has for the first time enabled us to reproduce DA neurons from individuals who suffered from familial or sporadic PD [47, 48]. Moreover, these iPSC models allow us to explore pathogenic factors and discover interactions between genetic and exogenous factors involved in the pathogenesis of PD. As individuals' responses to drug compounds varies, patient-specific iPSCs may be used to distinguish between those individuals likely to respond to new therapeutics and those who are not, and more accurately predict toxicity and efficacy in screening drugs, from mechanisms, in comparison to animal models.

Differentiation of DA neurons from iPSCs has been demonstrated to be relatively robust and reproducible, allowing for generation of disease models from patients carrying a variety of mutations in key genes implicated in familial PD, including PARK2, PINK1, LRRK2, SNCA and GBA [49, 50]. Among genetic risk factors, Parkin (PARK2) is the most frequently mutated gene that has causally been linked to autosomal recessive early-onset familial PD [51]. In patients with PD onset before the age 45, PARK2 mutations are seen in up to 50% of familial cases and about 15% of sporadic cases [52]. Parkin knockout mouse models display some abnormalities, but do not fully recapitulate the pathophysiology of human PARK2. Jiang et al. generated iPSCs from normal subjects and PD patients with Parkin mutations and demonstrated that loss of Parkin in human midbrain DA neurons greatly increased expression of monoamine oxidases and oxidative stress, significantly reduced DA uptake and increased spontaneous DA release [53]. These results suggest that Parkin controls dopamine utilization in human midbrain DA neurons by enhancing the precision of DA neurotransmission and suppressing DA oxidation [53]. Imaizumi et al. used PARK2 patients' specific iPSCs-derived neurons to recapitulate pathogenic changes in the brain of PARK2 patients [54]. The data indicated that PARK2 iPSC-derived neurons exhibited increased oxidative stress, impaired mitochondrial homoeostasis and SNCA accumulation [54]. Recently, Ren et al. showed that the complexity of neuronal processes and microtubule stability were significantly reduced in iPSC-derived neurons from PD patients with Parkin mutations [55], suggesting that Parkin maintains morphological complexity of human neurons by stabilizing microtubules [55]. Shaltouki et al. observed reduced DA differentiation, accompanied by reduced mitochondrial volume ratio and abnormal mitochondrial ultrastructure, consistent with the current model of PARK2 mutations [48].

PINK1 functions upstream of Parkin, and is involved in recruiting Parkin to damaged mitochondria, for example, following mitochondrial depolarization [56]. Mutations in either PARK2 or PINK1 result in impaired mitophagy. Cooper et al. found that PINK1 mutant iPSC-derived DA neuronal cells were more sensitive to cell death and production of ROS elicited by mitochondrial and oxidative stressors, and further showed increased basal oxygen consumption and proton leakage suggestive of intrinsically damaged mitochondria [57]. Moreover, cell vulnerability associated with mitochondrial function in iPSC-derived neural cells could be rescued with coenzyme Q10, rapamycin or LRRK2 kinase inhibitor GW5074. These data demonstrate that iPSC-derived neural cells are sensitive models for measuring vulnerability and doseresponses of candidate neuroprotective molecules and might help to identify disease causes and better individualize treatment efficacy [57].

Mutations in leucine-rich repeat kinase 2 (LRRK2) are associated with sporadic and familial forms of PD. Nguyen et al. have reported that DA neurons derived from G2019S mutation-iPSCs have high levels of expressions of key oxidative stress-response genes and SNCA protein [58]. The mutant neurons were more sensitive to caspase-3 activation and cell death caused by exposure to stress agents, such as hydrogen peroxide, MG-132 and 6-hydroxydopamine [58]. Cooper et al. indicated that LRRK2 G2019S and R1441C mutations reduced availability of substrates for oxidative phosphorylation, and were associated with disrupted mitochondrial movement in PD patient-specific iPSCs [57]. Laurie et al. further demonstrated the mechanisms by which LRRK2 mutations lead to loss of mitochondrial function. Their data revealed that mitochondrial DNA (mtDNA) damage was induced in neural cells by PD-associated mutations in LRRK2, and this phenotype could be functionally reversed or prevented by zinc finger nuclease (ZFN)-mediated genome editing in iPSCs [59]. These results indicate that mtDNA damage is likely to be a critical early event in neuronal dysfunction that leads ultimately to LRRK2-related PD [59].

The first genetic cause identified for familial PD was SNCA, as PD can be caused by mutations in SNCA or by overexpression of normal SNCA via gene duplication or triplication, consistent with a gain-of-function mechanism. iPSC-derived midbrain DA cultures from SNCA triplication patients exhibit several disease-related phenotypes in culture, including accumulation of SNCA, inherent overexpression of markers of oxidative stress and sensitivity to peroxide-induced oxidative stress [60]. iPSCs, reprogrammed from patients with the most common A53T-SNCA mutation, had high nitric oxide and 3-NT levels compared to controls [61].

GBA mutations, which cause the lysosomal storage disorder Gaucher disease, have recently been linked to a 5-fold greater risk of developing Parkinsonism than non-carrier individuals [62], and are the strongest genetic risk factor for PD known to date. GBA1 mutated iPSC-derived neurons have low glucocerebrosidase activity and protein levels, and high SNCA levels as well as autophagic and lysosomal defects [63]. Mutant neurons display dysregulation of calcium homoeostasis and increased vulnerability to stress responses involving elevation of cytosolic calcium [63]. These findings using iPSC technology, have provided evidence for a link between GBA1 mutations and complex changes in autophagic/lysosomal system and intracellular calcium homoeostasis, underlying vulnerability to neurodegeneration. As monozygotic twins share identical genetic makeup, twin studies have been valuable for dissecting complex gene-environmental interactions in PD. Woodard et al., using iPSC technology [64], investigated a unique set of monozygotic twins and found that SNCA clearance was impaired in midbrain DA neurons carrying GBA N370S regardless of disease status. Moreover, DA levels of twins discordant for PD were different, suggesting that non-genetic factors further perturbed DA homoeostasis in addition to GBA mutations. These results verified the interactions between genetic and environmental factors in the progress of PD, and offer a theoretical basis for personalized medicine in PD (Table 1).

R42P

EX3DEL R275W

More sensitive to cell death and production of ROS elicited by mitochondrial and oxidative stressors, and increased basal oxygen consumption and proton leakage

Phenotype rescue using coenzyme Q10, rapamycin or the LRRK2 kinase inhibitor GW5074

Increased expression of key oxidative stress-response genes and -synuclein

More sensitive to caspase-3 activation and cell death caused by exposure to stress agents

L444P,

N370S

Reduced glucocerebrosidase activity and protein levels, increased -synuclein levels as well as autophagic and lysosomal defects

Dysregulation of calcium homoeostasis and increased vulnerability to stress responses involving elevation of cytosolic calcium

Although the cause of sporadic PD is not fully understood, various factors including environmental toxins, genetic susceptibility and age, have been implicated. Isogenic hiPSC PD models show that toxin-induced nitrosative/oxidative stress results in S-nitrosylation of transcription factor MEF2C and this redox reaction inhibits MEF2C-PGC1 transcriptional network, contributing to mitochondrial dysfunction and apoptotic cell death [65], suggesting that the MEF2C-PGC1 pathway may be a new drug target for PD. The advance of iPSC technology now enables widespread development of PD models for dissecting molecular mechanisms that contribute to its disease pathogenesis.

As previously mentioned, there is currently no cure for PD except for some extent of relieving the symptoms. Current treatments include the use of oral medication of l-DOPA dopamine receptor agonists, MAO-B, apomorphine in more serious cases, continuous intestinal infusion of l-DOPA, and deep brain stimulation (DBS) in subthalamic nucleus and globus pallidus by using surgically implanted electrodes [66]. l-DOPA is the gold standard for treatment of PD. Up to now, no medical nor surgical therapy has been shown to provide superior anti-parkinsonian benefits than can be achieved with l-DOPA [67]. Unfortunately, its therapeutic effect is reduced after around 35 years use [68]. The problems and limitations associated with long-term use of l-DOPA, including on-off fluctuations and emergence of dyskinesia, facilitating exploration of better ways to restore dopamine neurotransmission. Dopamine receptor agonists are used as the first choice to delay initiation of l-DOPA treatment, with longer plasma elimination half-lives than l-DOPA. Their mechanism of action are by stimulation of presynaptic and postsynaptic DA receptors so that their use has therefore been considered to be opportunity to improve continuous drug delivery [67]. Selegiline was the first selective, irreversible inhibitor of monoamine oxidase type B (MAO-B) used in treatment of PD, which can stabilize DA levels in the synaptic cleft [67]. Because of its capacity for interfering with oxidative stress and for blocking MPTP toxicity, selegiline has been tested in the first major trial as a putative disease-modifying agent [67]. Rasagiline is another MAO-B inhibitor, with different metabolites than selegiline, successfully developed for PD therapy. Good tolerance to rasagiline and its ease of use make it an appealing option at the start of therapy [67]. DBS as surgical treatment has some serious limitations. It is costly and can produce cognitive disorders, which may be permanent [68]. All of these treatments have considerable side effects such as ultimate loss of drug effect (wearing off) during disease progression, occurrence of dyskinesia (notably with l-DOPA) use, and appearance of non-motor symptoms that are largely refractory to dopaminergic medication [69]. The concept of using cell transplantation to substitute for loss of DA neurons in the brains of PD patients has evolved. In addition to conventional clinical treatments, such as pharmaceutical drugs and DBS, cell replacement therapy has offered a novel basis for development of effective therapeutic strategies for PD. In 1987, Brundin et al. first transplanted human VM tissue into the striatum of PD patients in Sweden, and the era of cell therapy for PD patients started [70]. Various source tissues have been assessed for therapeutic replacement of DA neurons, such as hESC, hiPSC or DA grafts directly converted from somatic cells. Our team also has transplanted DiI-labelled human umbilical cord mesenchymal stem cells (HUMSCs) to rotenone-induced hemiparkinsonian rats [71]. We showed that intra-CPu transplantation of DiI-labelled HUMSCs 4 weeks after rotenone administration ameliorated APO-induced rotations gradually over a period of 12 months, indicating long-term therapeutic effect of this approach [71]. By monitoring red fluorescence of DiI, we found that HUMSCs migrated in the lesioned cerebral hemisphere, from CPu to SNc, or even to the opposite hemisphere through the corpus callosum. HUMSCs survived for up to 12 months after transplantation, and differentiated into Nestin-, NSE-, GFAP- and TH-positive cells in the CPu and TH+ cells in the SNc. No tumour-like structures was observed in implanted CPu [71]. As reported, vascular endothelial growth factor (VEGF) is a neurotrophic factor which has been proven to promote growth and survival of DA neurons in VM explants and animal models for PD [72-74]. Our previous work has also indicated that relatively low-level expression of VEGF in the striatum protects DA neurons of Parkinsonian rats [75]. Next, we developed a more effective neurorestorative and neuroregenerative therapy combining VEGF and HUMSC [76]. As a result, intrastriatal infusion VEGF-expressing HUMSCs to rotenone-induced Parkinsonian rats provided a significant behavioural improvement, more significant than HUMSC transplantation alone, and resulted in revival of TH immunoreactivity in the lesioned striatum and SNc [76]. Importantly, VEGF expression enhanced neuroprotective effects by promoting DA neuron-orientated differentiation of the HUMSCs. Thus, our findings have presented the suitability of HUMSC as a vector for gene therapy and suggested that stem cell engineering with VEGF may improve transplantation strategies for PD treatment [76].

iPSCs, induced pluripotent cells, have the potential capacity for self-renewal and are able to differentiate into any somatic cells, including DA neurons [77]. Alternatively, iPSCs have properties similar to ESCs but can be generated from adult human cells such as skin, adipose tissue and fibroblasts [10]. Thus, they are ethically more acceptable than some other stem cells sources. In theory, iPSCs from patients are without risk of immunological rejection for autografting [78].

Before successfully generating hiPSCs, many efforts had been made in animal experiments. In 2006, Yamanaka et al. generated iPSCs from mouse embryonic fibroblasts (MEF) and adult mouse tail-tip fibroblasts, by retrovirus-mediated transfection of four transcription factors, Oct3/4, Sox2, c-Myc and Klf4 [10]. One year later, iPSCs were derived by the same group [79] by viral reprogramming of human skin fibroblasts with the same four factors. These studies opened a new avenue for generating patient- and disease-specific pluripotent stem cells. Wernig et al. used 6-OHDA-lesioned rats to examine whether DA neurons derived from directly reprogrammed fibroblasts had therapeutic potential for PD animals [78]. As a result, in the striatum of rats grafted with differentiated iPSCs, a large number of TH+ cells with complex morphologies have been observed, these grafted stem cells were also positive for En1, VMAT2 and DAT. Four of the five transplanted animals which contained large numbers of TH+ neurons showed marked recovery of rotation behaviour 4 weeks after transplantation [78].

Kikuchi et al. first grafted hiPSC-derived DA neurons into the brains of an MPTP-lesioned Parkinsonian monkey, which survived as DA neurons as long as 6 months [80]. In order to reduce immune rejection, Deleidi et al. generated iPSCs from cynomolgus macaque (CM) skin fibroblasts carrying specific major histocompatibility complex (MHC) haplotypes, observing that neither tumour formation nor inflammatory reactions occurred in the transplanted animals, up to 6 months after transplantation [81]. Concerning the aspect of directed differentiation of iPSC into DA neurons, Snchez-Danes et al. indicated lentiviral vectors driving controlled expression of LMX1A was an efficient way to generate enriched populations of human VM DA neurons [82]. However, Mak et al. revealed that the protocol using dorsomorphin and SB431542 to replace SHH with purmorphamine or smoothened agonist could greatly improve conversion of hiPSCs to the neuronal lineage [83]. These histocompatible iPSCs may allow pre-clinical validation of safety and efficacy of iPSCs for PD.

Two commonly used anti-convulsants drugs, valproic acid (VPA) and zonisamide (ZNS), have been tested to promote differentiation of iPSC-derived DA neurons [84]. As iPSC-derived donor cells inevitably contain tumourigenic or inappropriate cells, finding better protocols to purify and sort iPSCs is urgent. Doi et al. have shown that hiPSC-derived DA progenitor cells can be efficiently isolated by cell sorting using a floor plate marker, CORIN [85]. When transplanted into 6-OHDA-lesioned rats, CORIN+ cells survived and differentiated into midbrain DA neurons in vivo, resulting in significant improvement in motor behaviour, without tumour formation [85]. Recently, Hallett et al. analysed CM iPSC-derived midbrain DA neurons for up to 2 years following autologous transplantation in a PD model. They observed that unilateral engraftment of CM-iPSCs provided gradual onset of functional motor improvement, and increased motor activity, without any need for immunosuppression. Postmortem analyses demonstrated robust survival of midbrain-like DA neurons and extensive outgrowth into the transplanted putamen [86]. These experiments offered strong immunological, functional and biological rationales for using midbrain DA neurons derived from iPSCs for future cell replacement in PD (Table 2).

Although pre-clinical studies concerning iPSCs-derived cell therapies have shown great achievement, yet, some limitations hinder clinical usage of iPSCs for PD treatment. Tumourigenicity of iPSCs is an an important putative problem. Murine iPSCs and ESCs both form teratomas when transplanted into syngeneic mice. Also, hiPSCs and hESCs generate teratomas when injected into immunodeficient mice [87]. A standardized sensitive teratoma assay to detect low numbers of tumour-forming cells within a therapeutic cell preparation would be highly valuable. Gropp et al. presented detailed characterization of an efficient, quantitative, sensitive and easy-to-perform teratoma assay [87]. These tumours may be benign (also, they may be malignant), although even so can become fatal when very large. In some studies, when teratomas have been removed from mice, the animals survived [88]. Importantly, aggressiveness of teratocarcinomas from iPSCs is greater than that of ESCs [88]. Differences in oncogenicity between ESCs and iPSCs might be due to their different approach of being derived [88]. As hiPSCs have been first derived by transduction of human dermal fibroblasts with integrating viruses carrying four transcription factors Oct4, Sox2, c-Myc and Klf4 [79], c-Myc is a well-established oncogene while the other three transcription factors are known to be highly expressed in various types of cancer [89-91]. Yamanaka et al. subsequently reported a further Myc family member, L-Myc, as well as C-Myc mutants (W136E and dN2), all of which indicated little transformational activity, promoting hiPSC generation more efficiently and specifically compared to WT C-Myc [32]. A further cause of tumourigenicity may be attributed to random integration of foreign DNA into the host genome disrupting important genes or activating oncogenes, potentially leading to uncontrollable growth of cells [92].

Stadtfeld et al. generated mouse iPSCs from fibroblasts and liver cells by using non-integrating adenoviruses transiently expressing Oct4, Sox2, Klf4 and c-Myc [93]. These adenoviral iPSCs (adeno-iPSCs) showed DNA demethylation characteristic of reprogrammed cells, expressed endogenous pluripotency genes and formed teratomas [93]. Their work indicated that insertional mutagenesis was not required for in vitro reprogramming [93]. Thus, more than 2 years after establishment of iPSC technology by Yamanaka's group, these newly generated adeno-iPSCs have been the first reported reprogrammed pluripotent stem cells with evidence of complete lack of viral transgene integration [94]. Yamanaka et al. described an alternative method to generate iPSCs from MEFs by continual transfection of plasmid vectors free from plasmid integration [95]; this protocol took around 2 months to complete, from MEF isolation to iPSC establishment. The virus-free technique reduced safety concerns for iPSC generation and application, and have provided a source of cells for investigation of mechanisms underlying reprogramming and pluripotency [95]. Introducing mRNA directly into host cells without altering their genomic makeup or using episomal DNA-based vectors which seldom integrate into the host genome, holds the potential to solve this problem, by providing sufficient reprogramming factor expression for successful transformation of somatic cells [96-98].

Anokye-Danso et al. showed that expression of the miR302/367 cluster rapidly and efficiently reprogramed mouse and human somatic cells to an iPSC state without requirement for exogenous transcription factors [99]. This miRNA-based reprogramming approach was two orders of magnitude more efficient than standard Oct4/Sox2/Klf4/Myc-mediated methods, and the miR302/367 iPSCs displayed characteristics similar to the Oct4/Sox2/Klf4/Myc-iPSCs [99].

A further problem which hinders iPSCs treatment is that the therapeutic effect can be influenced by inherent pathogenic features of PD. Interactions between genetic and exogenous factors result in its pathogenesis. A general concern about use of autologous iPSC transplantation is whether underlying PD-associated genetic mutations presented in transplanted neurons increases vulnerability of iPSC-derived midbrain DA neurons to disease pathology. Environmental factors and age contribute largely to the pathogenesis of PD. Thus, iPSC-derived neurons represent a reasonable strategy for more advantages. It has been shown that LBs, the pathological features of PD, can be found in grafts of foetal VM tissue [100, 101]. The first reason is that LB pathology is a reaction to inflammation from host brain tissues, possibly mediated by cell stress induced by reactive microglia [102]. Second, the spread of SNCA into the graft from the host may contribute to these pathological changes [103, 104]. However, Barker et al. believe that these pathological changes are not likely to limit widespread adoption of cell treatments, as numbers of cells with LB-like pathology in the grafts were small compared to numbers of healthy cells [105-107]. Patients can still be functionally stable, more than a decade after such a graft, at a time when accumulation of SNCA had been observed [108, 109].

Apart from the potential risk of tumourigenicity or inherent pathogenic features deriving from donor cells, pluripotent stem cell-derived cell populations for therapies also confer a risk for the contamination of transplantation cell populations with residual pluripotent cells [110]. In order to resolve this issue, several sorting methods have been developed for enrichment of differentiated neural cell populations and elimination of pluripotent stem cells, using flow cytometric analysis (FACS) or MACS [111]. Different combinations of CD markers have been explored to purify heterogeneous pluripotent stem cell-derived neural cell populations. Pruszak et al. identified a cluster of differentiation (CD) surface antigen code for the neural lineage, based on combinatorial FACS of three distinct populations derived from hESCs: combinatorial CD15/CD24/CD29 marker profiles [112]. They found that CD15(+)/CD29(HI)/CD24(LO) surface antigen expression defined NSCs, and this could eliminate tumour formation in vivo, resulting in pure neuronal grafts [112]. Yuan et al. performed an unbiased FACS- and image-based immunophenotyping analysis using 190 antibodies to cell surface markers on pluripotent stem cells [113]. From this analysis they isolated a population of NSC that were CD184(+)/CD271(-)/CD44(-)/CD24(+) from neural induction cultures of hESCs and hiPSCs [113]. To improve the sorting method, Sundberg et al. sorted primate iPSC-derived neural cell population with NCAM+/CD29low selection [111]. They demonstrated that teh NCAM+/CD29low selection method enriched the FOXA2/TH and EN1/TH+ DA neurons in vitro compared to unsorted cell populations from >10% prior to sorting to > 35% after sorting. Importantly, sorting with NCAM+/CD29low selection prior to transplantation eliminated non-neural tumourigenic cells from the grafts and significantly increased the number of TH+ cells in the cell grafts compared to unsorted cell populations [111].

In this review, we have summarized a number of scientific and ethical issues in modelling and treatment with iPSCs for PD. iPSCs can be employed as relevant Parkinsonian cell models, for drug screening, studying disease progression and most importantly for treatment of PD by transplantation techniques. Compared with other stem cells, iPSCs stand out for their powerful pluripotency, few ethical issues and less immune rejection, although there are still several issues that need to be solved prior to translation of iPSCs into the clinical setting. First, the exact mechanisms of how transplanted cells restore host brain function and how to connect them with circumjacent brain tissues have not been yet elucidated. Second, tumourigenicity of iPSCs may surpass their therapeutic effects. Ultimately, iPSC, derived from autologous PD patients, may contain pathogenic gene mutations that affect prognosis of transplantation therapy. With improvements in differentiation methodologies and better understanding of pathogenesis of PD through patient-specific iPSCs, iPSC therapy can be a potential alternative for PD treatment combined with traditional drug development platforms and gene therapy.

This work was supported by grants 31171211 and 81471305 from the National Natural Science Foundation of China (to TW), grant 81200983 from the National Natural Science Foundation of China (to NX), grant 81301082 from the National Natural Science Foundation of China (to JSH), grant 2012B09 from China Medical Foundation (to NX) and grant 0203201343 from Hubei Molecular Imaging Key Laboratory (to NX). The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.

There are no actual or potential conflicts of interest.

2016 John Wiley & Sons Ltd

Request Permissions

Please enable Javascript to view the related content of this article.

Advertisement

Follow this link:
Induced pluripotent stem cells and Parkinson's disease ...

Induced Pluripotent Stem Cells: A New Frontier for Stem …

Induced pluripotent stem cells (iPSCs) are the newest member of a growing list of stem cell populations that hold great potential for use in cell-based treatment approaches in the dental field. This review summarizes the dental tissues that have successfully been utilized to generate iPSC lines, as well as the potential uses of iPSCs for tissue regeneration in different dental applications. While iPSCs display great promise in a number of dental applications, there are safety concerns with these cells that need to be addressed before they can be used in clinical settings. This review outlines some of the apprehensions to the use of iPSCs clinically, and it details approaches that are being employed to ensure the safety and efficacy of these cells. One of the major approaches being investigated is the differentiation of iPSCs prior to use in patients. iPSCs have successfully been differentiated into a wide range of cells and tissue types. This review focuses on 2 differentiation approaches-the differentiation of iPSCs into mesenchymal stem cells and the differentiation of iPSCs into osteoprogenitor cells. Both these resulting populations of cells are particularly relevant to the dental field.

International & American Associations for Dental Research 2015.

Read the rest here:
Induced Pluripotent Stem Cells: A New Frontier for Stem ...

What are embryonic stem cells or ES cells?

Embryonic stem cells are derived from early stage embryos (called blastocysts) that are five to seven days old. In Australia these blastocysts are donated for research with consent from patients who have completed treatment for infertility, and have surplus embryos. The embryos donated and used in research would otherwise be discarded. At this stage of development the blastocyst is a hollow ball of about 150 cells and no bigger than a pinhead. The blastocyst is a unique stage of development, as there are two different types of cells, an outer layer called the trophectoderm, and a small group of approximately 30 cells called the inner cell mass. The inner cell mass is what ultimately becomes the embryo, and the trophectoderm becomes the placenta.

Embryonic stem cells are isolated from the blastocyst when the inner cell mass is removed and cultured in the laboratory. During this process the blastocyst is destroyed.Once the cells have been isolated they can be grown continuously in a laboratory culture dish that contains a nutrient-rich culture medium. As the stem cells divide and spread over the surface of the dish some are removed to populate fresh subcultures to form a stem cell line. Because these cells have the ability to keep dividing (self-renewing), large numbers of embryonic stem cells can be grown in the laboratory and also frozen for future use. As a result, established embryonic stem cell lines can be maintained in laboratories for research, shared between researchers and maybe ultimately used in cell-based therapies.

It has been legal to use human embryos for stem cell research in Australia since 2002 but only where scientists meet strict regulatory requirements such as obtaining a licence for the creation of the human embryonic stem cells.

To learn more about embryonic stem cells watchCreation of human embryonic stem cell linesin our video library.

To learn more about the Australian laws that regulate this area of science watchThe Stem Cell Debateor visitIs stem cell research legal in Australia?

Continued here:
What are embryonic stem cells or ES cells?