Page 32«..1020..31323334..4050..»

Archive for the ‘Skin Stem Cells’ Category

Stem Cell Therapy for Anti-Aging and Sexual Performance …

Stem Cell Therapy has been around for quite some time, but due to high cost it was primarily used for recovery in athletes and the financial elite. However, with the progression of science and knowledge, stem cell therapy has become much more widely used and financially attainable.

Tampa Rejuvenation is the first in Tampa Bay to utilize the benefits of stem cell therapy for the purpose of anti-aging and sexual performance. We realize as our patients age, their bodies no longer have the regenerative properties to attain the desired results from using their growth factors alone as with our PRP, or Plasma Rich Platelet, therapy. Although many patients will still yield improvement with the PRP alone, the magnitude of cytokines and growth factors in your blood as you age will deplete with age. By implementing stem cell therapy, the number of growth factors are exponential allowing our bodies to regenerate on a magnitude that is otherwise unattainable with some results lasting for 3-5 years.

Stem Cell Therapy can be used to restore vitality to the skin, encourage the growth of hair, and even restore sexual performance and pleasure.

The rest is here:
Stem Cell Therapy for Anti-Aging and Sexual Performance ...

Stem cell numbers in a damaged knee – Dr. Marc Darrow is a …

Are there enough stem cells in your knees to heal the damage of osteoarthritis? If yes, why arent those stem cells fixing your knees now? Is it a lack of numbers?

Marc Darrow MD, JD. Thank you for reading my article. You can ask me your questions about bone marrow derived stem cells using the contact form below.

In 2011, doctors at the University of Aberdeen published research in the journal Arthritis and rheumatism that provided the first evidence that resident stem cells in the knee joint synovium underwent proliferation (multiplied) and chondrogenic differentiation (made themselves into cartilage cells) following injury.(1)This paper, presenting the idea that stem cells in an injured knee increased in numbers in preparation of healing has been cited by more than 40 medical studies.

If the stem cells in your knee synovial lining are abundant and have the ability to rebuild cartilage after injury, why isnt your knee fixing itself?

One of those 40 studies was performed by researchers at theUniversity of Calgary in 2012. Among their questions, if the stem cells in the knee synovial lining are abundant and have the ability to rebuild cartilage after injury, why isnt the knee fixing itself? Here is what they published:

Since osteoarthritis leads to a progressive loss of cartilage and synovial progenitors (rebuilding) cells have the potential to contribute to articular cartilage repair, the inability of osteoarthritis synovial fluid Mesenchymal progenitor cells (stem cell growth factors) to spontaneously differentiate into chondrocytes suggests that cell-to-cell aggregation and/or communication may be impaired in osteoarthritis and somehow dampen the normal mechanism of chondrocyte replenishment from the synovium or synovial fluid. Should the cells of the synovium or synovial fluid be a reservoir of stem cells for normal articular cartilage maintenance and repair, these endogenous sources of chondro-biased cells would be a fundamental and new strategy for treating osteoarthritis and cartilage injury if this loss of aggregation & differentiation phenotype can be overcome.(2)

This research was supported in a study from December 2017 In Nature reviews. The paper suggested that recognizing that joint-resident stem cells are comparatively abundant in the joint and occupy multiple niches (from the center of the joint to the out edges) will enable the optimization of single-stage therapeutic interventions for osteoarthritis.(3) The idea is to get these native stem cells to repair.

Now we know that there are many stem cells in the knee, when there is an injury there are more stem cells. If we can figure out how to get these stem cells turned on to the healing mode, the knee could heal itself of early stage osteoarthritis. So the problem is not the number of stem cells, BUT, communication.

This failure to communicate was also seen in other research. In 2016, another heavily cited paper, this time fromTehran University for Medical Sciences, noted that despite their larger numbers,the native stem cells act chaotically and are unable to regroup themselves into a healing mechanism and repair the bone, cartilage and other tissue. Introducing bone marrow stem cells into this environmentgets the native stem cells in line and redirects them to perform healing functions. The joint environmentis changed from chaotic to healing because of communication.(4) It should be pointed out that 62 medical studies cited the research in this papers findings).

A recentpaper from a research team inAustralia confirms how this change of joint environment works. It starts with cell signalling a new communication network is built.

University of Iowa research published in theJournal of orthopaedic research

Serious meniscus injuries seldom heal and increase the risk for knee osteoarthritis; thus, there is a need to develop new reparative therapies. In that regard, stimulating tissue regeneration by autologous (from you, not donated) stem/progenitor cells has emerged as a promising new strategy.

(The research team) showed previously that migratory chondrogenic progenitor cells (mobile cartilage growth factors) were recruited to injured cartilage, where they showed a capability in situ (on the spot) tissue repair. Here, we tested the hypothesis that the meniscus contains a similar population of regenerative cells.

Explant studies revealed that migrating cells were mainly confined to the red zone (where the blood is and its growth factors) in normal menisci: However, these cells were capable of repopulating defects made in the white zone (the desert area where no blood flows. Migrating cell numbers increased dramatically in damaged meniscus. Relative to non-migrating meniscus cells, migrating cells were more clonogenic, overexpressed progenitor cell markers, and included a larger side population. (They were ready to heal) Gene expression profiling showed that the migrating population was more similar tochondrogenic progenitor cells (mobile cartilage growth factors) than other meniscus cells. Finally, migrating cells equaledchondrogenic progenitor cells in chondrogenic potential, indicating a capacity for repair of the cartilaginous white zone of the meniscus. These findings demonstrate that, much as in articular cartilage, injuries to the meniscus mobilize an intrinsic progenitor cell population with strong reparative potential.(6)

The intrinsic progenitor cell population with strong repair potential are in your knee waiting to be mobilized.

So what are we to make of this research?There are a lot of stem cells in a knee waiting to repair. The problem is they are confused and not getting the correct instructions. Stem cell therapy can fix the communication problem and begin the repair process anew.

A leading provider of bone marrow derived stem cell therapy, Platelet Rich Plasma and Prolotherapy11645 WILSHIRE BOULEVARD SUITE 120, LOS ANGELES, CA 90025

PHONE: (800) 300-9300

1 Kurth TB, Dellaccio F, Crouch V, Augello A, Sharpe PT, De Bari C. Functional mesenchymal stem cell niches in adult mouse knee joint synovium in vivo. Arthritis Rheum. 2011 May;63(5):1289-300. doi: 10.1002/art.30234.

2 Krawetz RJ, Wu YE, Martin L, Rattner JB, Matyas JR, Hart DA. Synovial Fluid Progenitors Expressing CD90+ from Normal but Not Osteoarthritic Joints Undergo Chondrogenic Differentiation without Micro-Mass Culture. Kerkis I, ed.PLoS ONE. 2012;7(8):e43616. doi:10.1371/journal.pone.0043616.

3 McGonagle D, Baboolal TG, Jones E. Native joint-resident mesenchymal stem cells for cartilage repair in osteoarthritis. Nature Reviews Rheumatology. 2017 Dec;13(12):719.

4Davatchi F, et al. Mesenchymal stem cell therapy for knee osteoarthritis: 5 years follow-up of three patients. Int J Rheum Dis. 2016 Mar;19(3):219-25.

5. Freitag J, Bates D, Boyd R, Shah K, Barnard A, Huguenin L, Tenen A.Mesenchymal stem cell therapy in the treatment of osteoarthritis: reparative pathways, safety and efficacy a review.BMC Musculoskelet Disord. 2016 May 26;17(1):230. doi: 10.1186/s12891-016-1085-9. Review.

6 Seol D, Zhou C, et al. Characteristics of meniscus progenitor cells migrated from injured meniscus. J Orthop Res. 2016 Nov 3. doi: 10.1002/jor.23472.

Read the original post:
Stem cell numbers in a damaged knee - Dr. Marc Darrow is a ...

Where Do Stem Cells Come From? | Basics Of Stem Cell …

Where do stem cells come from? Learn the basics of master cells to better understand their therapeutic potential.

In this article:

Where do stem cells come from? You have probably heard of thewonders of stem cell therapy. Not only do stem cells make research for future scientific breakthroughs possible, but they also provide the basis for many medical treatments today. So, where exactly are they from, and how are they different from regular cells? The answer depends on the types of stem cells in question.

There are two main types of stem cells adult and embryonic:

Beyond the two broader categories, there are sub-categories. Each has its own characteristics. For researchers, the different types of stem cells serve specific purposes.

Many tissues throughout the adult human body contain stem cells. Scientists previously believed adult stem cells to be inferior to human embryonic stem cells for therapeutic purposes. Theydid not believe adult stem cells to be as versatile as embryonic stem cells (ESCs), because they are not capable of becoming all 200 cell types within the human body.

While this theoryhas notbeen entirely disproved, encouraging evidence suggests that adult stem cells can develop into a variety of new types of cells. They can also affect repair through other mechanisms.

In August 2017, the number of stem cell publications registered in PubMed, a government database, surpassed 300,000. Stem cells are also being explored in over 4,600 cell therapy clinical trials worldwide. Some of the earliest forms of adult stem cell use include bone marrow and umbilical cord blood transplantation.

It should be noted that while the term adult stem cell is used for this type of cell, it is not descriptive of age, because adult stem cells can come from children. The term simply helps to differentiate stem cells derived from living humans as opposed to embryonic stem cells.

Embryonic stem cells are controversial because they are made from embryos that are created but not used by fertility clinics.

Because adult stem cells are somewhat limited in the cell types they can become, scientists developed a way to genetically reprogram cells into what is called an inducedpluripotent stem cell or iPS cell. In creating inducedpluripotent stem cells, researchers hope to blend the usefulness of adult stem cells with the promise of embryonic stem cells.

Both embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs) are known as pluripotent stem cells.

Pluripotent stem cells are a type of cell that has the capacity to divide indefinitely and create any cell found within the three germ layers of an organism: ectoderm (cells forming the skin and nervous system), endoderm (cells forming pancreas, liver, endocrine gland, and gastrointestinal and respiratory tracts), and mesoderm (cells forming connective tissues, and other related tissues, muscles, bones, most of the circulatory system, and cartilage).

Embryonic stem cells can grow into a much wider range of cell types, but they also carry the risk of immune system rejection in patients. In contrast, adult stem cells are more plentiful, easier to harvest, and less controversial.

Embryonic stem cells come from embryos harvested shortly after fertilization (within 4-5 days). These cells are made when the blastocysts inner cell mass is transferred into a culture medium, allowing them to develop.

At 5-6 days post-fertilization, the cells within the embryo start to specialize. At this time, they no longer are able to become all of the cell types within the human body. They are no longer pluripotent.

Because they are pluripotent, embryonic stem cells can be used to generate healthy cells for disease patients. For example, they can be grown into heart cells known as cardiomyocytes. These cells may have the potential to be injected into an ailing patients heart.

Harvesting stem cells from embryos is controversial, so there are guidelines created by the National Institutes of Health (NIH) that allow the public to understand what practices are not allowed.

Scientists can harvest perinatal stem cells from a variety of tissues, but the most common sources include:

The umbilical cord attaches a mother to her fetus. It is removed after birth and is a valuable source of stem cells. The blood it contains is rich in hematopoietic stem cells (HSC). It also contains smaller quantities of another cell type known as mesenchymal stem cells (MSCs).

The placenta is a large organ that acts as a connector between the mother and the fetus. Both placental blood and tissue are also rich in stem cells.

Finally, there is amniotic fluid surrounding a baby while it is in utero. It can be harvested if a pregnant woman needs a specialized kind of test known as amniocentesis. Both amniotic fluid and tissue contain stem cells, too.

Adult stem cells are usually harvested in one of three ways:

The blood draw, known as peripheral blood stem cell donation, extracts the stem cells directly from a donors bloodstream. The bone marrow stem cells come from deep within a bone often a flat bone such as the hip. Tissue fat is extracted from a fatty area, such as the waist.

Embryonic donations are harvested from fertilized human eggs that are less than five days old. The embryos are not grown within a mothers or surrogates womb, but instead, are multiplied in a laboratory. The embryos selected for harvesting stem cell are created within invitro fertilization clinics but are not selected for implantation.

Amniotic stem cells can be harvested at the same time that doctors use a needle to withdraw amniotic fluid during a pregnant womans amniocentesis. The same fluid, after being tested to ensure the babys health, can also be used to extract stem cells.

As mentioned, there is another source for stem cells the umbilical cord. Blood cells from the umbilical cord can be harvested after a babys birth. Cells can also be extracted from the postpartumhuman placenta, which is typically discarded as medical waste following childbirth.

The umbilical cord and the placenta are non-invasive sources of perinatal stem cells.

People who donate stem cells through the peripheral blood stem cell donor procedure report it to be a relativelypainless procedure. Similar to giving blood, the procedure takes about four hours. At a clinic or hospital, an able medical practitioner draws the blood from the donors vein in one of his arms using a needle injection. The technician sends the drawn blood into a machine, which extracts the stem cells. The blood is then returned to the donors body via a needle injected into the other arm. Some patients experience cramping or dizziness, but overall, its considered a painless procedure.

If a blood stem cell donor has a problem with his or her veins, a catheter may be injected in the neck or chest. The donor receives local anesthesia when a catheter-involved donation occurs.

During a bone marrow stem cell donor procedure, the donor is put under heavy sedation in an operating room. The hip is often the site chosen to harvest the bone marrow. More of the desired red marrow is found in flat bones, such as those in the pelvic region. The procedure takes up to two hours, with several extractions made while the patient is sedated. Although the procedure is painless due to sedation, recovery can take a couple of weeks.

Bone marrow stem cell donation takes a toll on the donorbecause it involves the extraction of up to 10 percent of the donors marrow. During the recovery period, the donors body gradually replenishes the marrow. Until that happens, the donor may feel fatigued and sore.

Some clinics offer regenerative and cosmetic therapies using the patients own stem cells derived from the fat tissue located on the sides of the waistline. Considered a simple procedure, clinics do this for therapeutic reasons or as a donation for research.

Stem cells differ from the trillions of other cells in your body. In fact, stem cells make up only a small fraction of the total cells in your body. Some people have a higher percentage of stem cells than others. But, stem cells are special because they are the mothers from which specialized cells grew and developed within us. When these cells divide, they become daughters. Some daughter cells simply self-replicate, while others form new kinds of cells altogether. This is the main way stem cells differ from other body cells they are the only ones capable of generating new cells.

The ways in which stem cells can directly treat patients grow each year. Regenerative medicine now relies heavily on stem cell applications. This type of treatment replaces diseased cells with new, healthy ones generated through donor stem cells. The donor can be another person or the patient themselves.

Sometimes, stem cells also exert therapeutic effects by traveling through the bloodstream to sites that need repair or by impacting their micro-environment through signaling mechanisms.

Some types of adult stem cells, like mesenchymal stem cells (MSCs), are well-known for exerting anti-inflammatory and anti-scarring effects. MSCs can also positively impact the immune system.

Conditions and diseases which stem cell regeneration therapy may help include Alzheimers disease, Parkinsons disease, and multiple sclerosis (MS). Heart disease, certain types of cancer, and stroke victims may also benefit in the future. Stem cell transplant promises advances in treatment for diabetes, spinal cord injury, severe burns, and osteoarthritis.

Researchers also utilize stem cells to test new drugs. In this case, an unhealthy tissue replicates into a larger sample. This method enables researchers to test various therapies on a diseased sample, rather than on an ailing patient.

Stem cell research also allows scientists to study how both healthy and diseased tissue grows and mutates under various conditions. They do this by harvesting stem cells from the heart, bones, and other body areas and studying them under intensive laboratory conditions. In this way, they get a better understanding of the human body, whether healthy or sick.

With the following stem cell transplant benefits, its not surprising people would like to try the therapy as another treatment option.

Physicians harvest stem cell from either the patient or a donor. For an autologous transplant, there is no risk of transferring any disease from another person. For an allogeneic transplant, the donor is meticulously screened before the therapy to make sure they are compatible with the patient and have healthy sources of stem cells.

One common and serious problem of transplants is the risk of rejecting the transplanted organs, tissues, stem cells, and others. With autologous stem cell therapy, the risk is avoided primarily because it comes from the same person.

Because stem cell transplants are typically done through infusion or injection, the complex and complicated surgical procedure is avoided. Theres no risk of accidental cuts and scarring post-surgery.

Recovery time from surgeries and other types of treatments is usually time-consuming. With stem cell therapy, it could only take about 3 months or less to get the patient back to their normal state.

As the number of stem cell treatments dramatically grew over the years, its survival rate also increased. A study published in the Journal of Clinical Oncology showed there was a significant increase in survival rate over 12 years among participants of the study. The study analyzed results from over 38,000 stem cell transplants on patients with blood cancers and other health conditions.

One hundred days following transplant, the researchers observed an improvement in the survival rate of patients with myeloid leukemia. The significant improvements we saw across all patient and disease populations should offer patients hope and, among physicians, reinforce the role of blood stem cell transplants as a curative option for life-threatening blood cancers and other diseases.

With the information above, people now have a better understanding of the answer to the question Where do stem cells come from? Stem cells are a broad topic to comprehend, and its better to go back to its basics to learn its mechanisms. This way, a person can have a piece of detailed knowledge about these master cells from a scientific perspective.

If you found this blog valuable, subscribe to BioInformants stem cell industry updates.

As the first and only market research firm to specialize in the stem cell industry, BioInformant research is cited by The Wall Street Journal, Xconomy, AABB, and Vogue Magazine. Bringing you breaking news on an ongoing basis, we encourage you to join more than half a million loyal readers, including physicians, scientists, executives, and investors.

What do you understand aboutthe basics of stem cells? Share your thoughts in the comments section below.

Up Next:Top 5 TED Talks About Stem Cells

Where Do Stem Cells Come From? | Basics Of Stem Cell Therapy

Read more:
Where Do Stem Cells Come From? | Basics Of Stem Cell ...

Stem Cells – The Hastings Center

By Insoo Hyun

Stem cells are undifferentiated cells that have the capacity to renew themselves and to specialize into various cell types, such as blood, muscle, and nerve cells. Embryonic stem cells, derived from five-day-old embryos, eventually give rise to all the different cells and organ systems of the embryo. Embryonic stem cells are pluripotent, because they are capable of differentiating along each of the three germ layers of cells in the embryo, as well as producing the germ line (sperm and eggs). The three germ layers are the ectoderm (skin, nerves, brain), the mesoderm (bone, muscle), and the endoderm (lungs, digestive system).

During later stages of human development, minute quantities of more mature stem cells can be found in most tissue and organ systems, such as bone marrow, the skin, and the gut. These are somatic stem cells, responsible for renewing and repairing the bodys specialized cells. Although the lay public often refers to them as adult stem cells, researchers prefer to call them multipotent because they are less versatile than pluripotent stem cells, and because they are present from the fetal stage of development and beyond. Multipotent stem cells can only differentiate into cells related to the tissue or organ systems from which they originated for instance, multipotent blood stem cells in bonemarrow can develop into different types of blood cells, but not into nerve cells or heart cells.

While multipotent stem cell research has been around for nearly 50 years and has led to clinical therapies for leukemia and other blood disorders, the field of human embryonic stem cell research is still relatively new, and basic discoveries have yet to be directly transitioned into clinical treatments. Human embryonic stem cells were first isolated and maintained in culture in 1998 by James Thomson and colleagues at the University of Wisconsin. Since then, more than a thousand different isolateslines of self-renewing embryonic stem cellshave been created and shared by researchers worldwide.

The main ethical and policy issues with stem cells concern the derivation and use of embryonic stem cells for research. A vocal minority of Americans objects to the destruction of embryos that occurs when stem cells are derived. Embryonic stem cell research is especially controversial for those who believe that five-day-old preimplantation human embryos should not be destroyed no matter how valuable the research may be for society.

To bypass this ethical controversy, the Presidents Council on Bioethics recommended in 2005 that alternative sources of pluripotent stem cells be pursued. Some alternatives have been developed, most notably, the induced pluripotent stem (iPS) cells human skin cells and other body cells reprogrammed to behave like embryonic cells. But embryonic stem cell research will remain needed because there are some questions only they have the potential to answer.

Embryonic stem cells are necessary for several aims of scientific and biomedical research. They include addressing fundamental questions in developmental biology, such as how primitive cells differentiate into more specialized cells and how different organ systems first come into being. By increasing our knowledge of human development, embryonic stem cells may also help us better understand the causes of fetal deformations.

Other important applications lie in the areas of disease research and targeted drug development. By deriving and studying embryonic or other pluripotent stem cells that are genetically-matched to diseases such as Parkinsons disease and juvenile diabetes, researchers are able to map out the developmental course of complex medical conditions to understand how, when, and why diseased specialized cells fail to function properly in patients. Such disease-in-a-dish model systems provide researchers with a powerful new way to study genetic diseases. Furthermore, researchers can aggressively test the safety and efficacy of new, targeted drug interventions on tissue cultures of living human cells derived from disease-specific embryonic stem cells. This method of testing can reduce the risks associated with human subjects research.

One possible way of deriving disease-specific stem cells is through a technique called somatic cell nuclear transfer (SCNT), otherwise known as research cloning. By replacing the DNA of an unfertilized egg with the DNA of a cell from a patients body, researchers are able to produce embryonic stem cells that are genetically-matched to the patient and his or her particular disease. SCNT, however, is technically challenging and requires the collection of high-quality human eggs from female research volunteers, who must be asked to undergo physically burdensome procedures to extract eggs.

A much more widespread and simpler technique for creating disease-specific stem cells was pioneered in 2006 by Shinya Yamanaka and colleagues in Kyoto, Japan. They took mouse skin cells and used retroviruses to insert four genes into them to to create iPS cells. In 2007, teams led by Yamanaka, James Thomson, and George Daley each used similar techniques to create human iPS cells. The iPS cell approach is promising because disease-specific stem cells could be created using skin or blood samples from patients and because, unlike SCNT, it does not require the procurement of human eggs for research.

However, despite these advances, scientists do not believe iPS cells can replace human embryonic stem cells in research. For one, embryonic stem cells must be used as controls to assess the behavior and full scientific potential of iPS cells. Furthermore, iPS cells may not be able to answer some important questions about early human development. And safety is a major issue for iPS cell research aimed at clinical applications, since the cell reprogramming process can cause harmful mutations in the stem cells, increasing the risk of cancer. In light of these and other concerns, iPS cells may perhaps prove to be most useful in their potential to expand our overall understanding of stem cell biology, the net effect of which will provide the best hope of discovering new therapies for patients.

Many who oppose embryonic stem cell research believe for religious or other personal reasons that all preimplantation embryos have a moral standing equal to living persons. On the other hand, those who support embryonic stem cell research point out that not all religious traditions grant full moral standing to early-stage human embryos.

According to Jewish, Islamic, Hindu, and Buddhist traditions, as well as many Western Christian views, moral standing arrives much later during the gestation process, with some views maintaining that the fetus must first reach a stage of viability where it would be capable of living outside the womb. Living in a pluralistic society such as ours, supporters argue, means having to tolerate differences in religious and personal convictions over such theoretical matters as when, during development, moral standing first appears.

Other critics of embryonic stem cell research believe that all preimplantation embryos have the potential to become full-fledged human beings and that they should never have this potential destroyed. In response, stem cell supporters argue that it is simply false that all early-stage embryos have the potential for complete human life many fertility clinic embryos are of poor quality and therefore not capable of producing a pregnancy (although they may yield stem cells). Similarly, as many as 75% to 80% of all embryos created through intercourse fail to implant. Furthermore, no embryos have the potential for full human life until they are implanted in a womans uterus, and until this essential step is taken an embryos potential exists only in the most abstract and hypothetical sense.

Despite the controversies, embryonic stem cell research continues to proceed rapidly around the world, with strong public funding in many countries. In the U.S., federal money for embryonic stem cell research is available only for stem cell lines that are on the National Institutes of Health stem cell registry. However, no federal funds may be used to derive human embryonic stem cell lines; NIH funds may only be used to study embryonic stem cells that were derived using other funding sources.

Despite the lack of full federal commitment to funding embryonic stem cell research in the U.S., there are wide-ranging national regulatory standards. The National Academy of Sciences established guidelines in 2005 for the conduct of human embryonic stem cell research. (See Resources.) According to these guidelines, all privately and publicly funded scientists working with embryonic stem cells should have their research proposals approved by local embryonic stem cell research oversight (ESCRO) committees. ESCRO committees are to include basic scientists, physicians, ethicists, legal experts, and community members to look at stem-cell-specific issues relating to the proposed research. These committees are also to work with local ethics review boards to ensure that the donors of embryos and other human materials are treated fairly and have given their voluntary informed consent to stem cell research teams. Although these guidelines are voluntarily, universities and other research centers have widely accepted them.

At the global level, in 2016 the International Society for Stem Cell Research (ISSCR) released a comprehensive set of professional guidelines for human stem cell research, spanning both bench and clinical stem cell research. (See Resources.) Unlike the NAS guidelines, the ISSCR guidelines go beyond American standards, adding, for example, the recommendation that stem cell lines be banked and freely distributed to researchers around the world to facilitate the fields progress on just and reasonable terms.The potential for over-commercialization and restrictive patenting practices is a major problem facing the stem cell field today, which may delay or reduce the broad public benefit of stem cell research. The promise of broad public benefit is one of thejustifying conditions for conducting stem cell research; without the real and substantial possibility for public benefit, stem cell research loses one of its most important moral foundations.

However, providing useful stem-cell-based therapies in the future is not a simple proposition, either. Developing a roadmap to bring stem cell research into the clinic will involve many complex steps, which the new ISSCR guidelines help address. They include:

These and other difficult issues must be sorted out if stem cell research in all its forms is to fulfill its promise.

STEM CELL GLOSSARY

Newer ethical issues in stem cell research go far beyond the embryo debate, since they encompass all stem cell types, not just human embryonic stem cells, and because they involve human subjects who, despite what one may think about the moral status of preimplantation embryos, are unequivocally moral persons. No other emerging issue better encapsulates the above concern than the growing phenomenon of stem cell tourism. At present, stem cell-based therapies are the clinical standard of care for only afew conditions, such as hematopoietic stem cell transplants for leukemia and epithelial stem cell-based treatments for burns and corneal disorders. Unfortunately, some unscrupulous clinicians around the world are exploiting patients hopes by purporting to provide for large sums of money effective stem cell therapies for many other conditions. These so-called stem cell clinics advance claims about their proffered stem cell therapies without credible scientific rationale, transparency, oversight, or patient protections.

The administration of unproven stem cell interventions outside of carefully regulated research protocols endangers patients and jeopardizes the legitimate progress of translational stem cell scientific research. Patients who travel for unproven stem cell therapies put themselves at risk of physical and financial harm.

The ISSCR guidelines are a good point for thinking about this important problem. The guidelines allow for exceptional circumstances in which clinicians might attempt medically innovative care in a very small number of seriously ill patients, subject to stringent oversight criteria. These criteria include: independent peer review of the proposed innovative procedure and its scientific rationale; institutional accountability; rigorous informed consent and close patient monitoring; transparency; timely adverse event reporting; and a commitment by clinician-scientists to move to a formal clinical trial in a timely manner after experience with at most a few patients. By juxtaposing some current stem cell clinics against the standards outlined in the ISSCR guidelines, one may easily identify some clinics shortcomings and call into question the legitimacy of their purported claims of providing innovative care to patients.

Moving beyond past debates about embryo status to issues concerning the uses of all varieties of stem cells, one can begin to focus the bioethical discourse on areas that have a much broader consensus base of shared values, such as patient and research subject protections and justice. Justice may also call on regulatory and oversight bodies to include a greater involvement of community and patient advocates in the oversight of research. Dealing with the bioethics of stem cell research demands that we wrestle with these and other tough questions.

Insoo Hyun, PhD, is an associate professor of bioethics at Case Western Reserve University.

See the original post here:
Stem Cells - The Hastings Center

StemFactor – Skin Growth Factor Serum

"I started using Osmosis about two weeks ago and only off and on. I had several skin tags on my face, which a doctor want to charge 100 dollars per tag to remove. The StemFactor dried them up and I have been removing them when washing. Unbelievable. I thought I had to live with and the best part, no hyperpigmentation. Thanks!" ~ B Andrews

Esthetician Marianne Kehoe, has worked with thousands of clients and numerous products for more than 20 years. "Between Catalyst and StemFactor, the results Im seeing in my practice are phenomenal, repeatedly, according to Kehoe, including with post-operative scars and people who have been exposed to the California sun for years."

"Over the past month I have noticed a significant improvement in my skin through using Osmosis skincare. I began with a starter pack then introduced Stem Factor - this has resulted in a huge difference to both the acne prone areas as well as contributing to the overall glow of my skin. I have had problems with my skin for a number of years and in particular Osmosis Stem Factor is the only product I've found that continues to improve skin appearance." ~ Emma

Read the original here:
StemFactor - Skin Growth Factor Serum

Stem Cell Basics A Closer Look at Stem Cells

About stem cells

Stem cells are the foundation of development in plants, animals and humans. In humans, there are many different types of stem cells that come from different places in the body or are formed at different times in our lives. These include embryonic stem cells that exist only at the earliest stages of development and various types oftissue-specific(oradult)stem cells that appear during fetal development and remain in our bodies throughout life.Stem cells are defined by two characteristics:

Beyond these two things, though, stem cells differ a great deal in their behaviors and capabilities.

Embryonic stem cells arepluripotent, meaning they can generate all of the bodys cell types but cannot generate support structures like the placenta and umbilical cord.

Other cells aremultipotent,meaning they can generate a few different cell types, generally in a specific tissue or organ.

As the body develops and ages, the number and type of stem cells changes. Totipotent cells are no longer present after dividing into the cells that generate the placenta and umbilical cord. Pluripotent cells give rise to the specialized cells that make up the bodys organs and tissues. The stem cells that stay in your body throughout your life are tissue-specific, and there is evidence that these cells change as you age, too your skin stem cells at age 20 wont be exactly the same as your skin stem cells at age 80.

Learn more about different types of stem cellshere.

View original post here:
Stem Cell Basics A Closer Look at Stem Cells

TREATMENTS – IMAGE NOW. Age later.

Our signature chemical peels help to reverse the visible effects of damage in two ways. First, they power away dull, dead cells to illuminate the skin and reduce the appearance of fine lines, wrinkles, age spots, clogged pores and blemishes. Then, they support collagen for firmer-looking skin over time. Ask your esthetician which IMAGE chemical peel will best target your skins individual needs.

I PEEL | WRINKLE LIFT

Ultra-resurfacing blend of glycolic acid combined with retinol to visibly reduce the appearance of fine lines and wrinkles.

Skin type indications: Aging, wrinkles, rough complexion, uneven skin tone, smokers skin, tired/dull skin, oily/acne

I PEEL | WRINKLE LIFT FORTE

This advanced treatment is formulated with additional glycolic acid and an innovative blend of firming and anti-aging properties, to visibly reduce the appearance of fine lines and wrinkles.

Skin type indications: Advanced aging, wrinkles, rough complexion, uneven skin tone, smokers skin, tired/dull skin, oily/acne

I PEEL | PERFECTION LIFT

This distinct blend of active exfoliants works synergistically to visibly reduce the appearance of fine lines, correct uneven skin tone, smooth rough texture and reduce acne blemishes.

Skin type indications: Aging, pigmentation, acne

I PEEL | PERFECTION LIFT FORTE

This concentrated blend of lactic acid, salicylic acid and resorcinol works synergistically to quickly and effectively reduce the appearance of advanced aging, pigmentation and acne. This extra strength treatment reveals a younger you in a single treatment.

Skin type indications: Advanced aging, pigmentation, acne

I PEEL | ACNE LIFT

Blend of AHAs and BHAs with protective agents to effectively treat all grades of acne.

Acne, oily, acne-prone, aging

I PEEL | BETA LIFT

This powerful non-blended salicylic acid treatment quickly and effectively targets and improves moderate/severe acne. Skin type indications: Acne, oily, aging

I PEEL | LIGHTENING LIFT

Lactic acid blended with kojic acid and a cocktail of brightening agents to reduce all forms of pigmentation.

Skin type indications: Pigmentation, aging, dry/dehydrated, uneven skin tone, age spots, redness-prone

I PEEL | LIGHTENING LIFT FORTE

This results-driven treatment combines the most innovative and effective botanical brighteners with echinacea, plant-derived stem cells and anti-aging peptides for youthful, illuminated skin.

Skin type indications: Advanced pigmentation, aging, dry/dehydrated, uneven skin tone, age spots, redness-prone

See the article here:
TREATMENTS - IMAGE NOW. Age later.

Types of Stem Cells A Closer Look at Stem Cells

Tissue-specific stem cells

Tissue-specific stem cells (also referred to assomaticoradultstem cells) are more specialized than embryonic stem cells. Typically, these stem cells can generate different cell types for the specific tissue or organ in which they live.

For example, blood-forming (orhematopoietic) stem cells in the bone marrow can give rise to red blood cells, white blood cells and platelets. However, blood-forming stem cells dont generate liver or lung or brain cells, and stem cells in other tissues and organs dont generate red or white blood cells or platelets.

Some tissues and organs within your body contain small caches of tissue-specific stem cells whose job it is to replace cells from that tissue that are lost in normal day-to-day living or in injury, such as those in your skin, blood, and the lining of your gut.

Tissue-specific stem cells can be difficult to find in the human body, and they dont seem to self-renew in culture as easily as embryonic stem cells do. However, study of these cells has increased our general knowledge about normal development, what changes in aging, and what happens with injury and disease.

Read this article:
Types of Stem Cells A Closer Look at Stem Cells

AnteAge Stem Cell Skin Care Reviewed

Serum: KEY ACTIVE INGREDIENTS:Stem CytokinesCarnosineNiacinamide (vit B3)Palmitoyl OligopeptidePalmitoyl Tetrapeptide-7Yerba MatGreenTea ExtractCatechins & Flavonoids

INGREDIENTS:Mesenchymal Stem Cell Cytokines,Water (Aqua), Palmitoyl Oligopeptide, Niacinamide (Vitamin B3), Palmitoyl Tetrapeptide-7, PPG-3 Benzyl Myristate, Dimethyl Isosorbide, Carnosine, Hydrolyzed Myrtus Communis (True Myrtle) Leaf Extract, Polyacrylate-13, Camellia Sinensis (Green Tea) Leaf Extract, Maltodextrin, Ilex Paraguariensis (Paraguay) Leaf (Yerba Mate) Extract, Cetearyl Ethylhexanoate, Polyisobutene, Phenoxyethanol (Preservative), Caprylyl Glycol (NaturallyDerived Preservative), Polysorbate-20 (Plant Derived), Chlorphenesin, TetrasodiumEDTA, Citric Acid (Naturally Derived) Accelerator: KEY ACTIVE INGREDIENTS:

INGREDIENTS: Mesenchymal Stem Cell Cytokines, Water (Aqua), Glycerin (Plant Derived), C12-15 Alkyl Benzoate, PPG-3 Benzyl Myristate, Carthamus Tinctorius (Safflower) Seed Oil, Alcohol, Cetearyl Alcohol (Plant Derived), Tocopheryl Acetate (Vitamin E Acetate), Polysorbate-20 (Plant Derived), Cetearyl Glucoside,Tetrahexyldecyl Ascorbate (Vitamin C Ester), Simmondsia Chinensis (Jojoba) Seed Oil, Limnanthes Alba (Meadowfoam) Seed Oil, Dimethyl Isosorbide, Butylene Glycol, Polysorbate-60 (Plant Derived), Glyceryl Stearate (Plant Derived),Lecithin, Hydroxyethyl Acrylate/Sodium Acryloyl Dimethyl Taurate Copolymer, SoybeanGlycerides, Arachidyl Alcohol, Soy Isoflavones, Phenoxyethanol (Preservative), Helianthus Annuus (Hybrid Sunflower) Oil, Butyrospermum Parkii (Shea Butter) Fruit, Bisabolol,Arbutin, Caprylyl Glycol (Naturally Derived Preservative), Behenyl Alcohol, Lonicera Japonica (Honeysuckle) Extract (Natural Preservative), Foeniculum Vulgare (Fennel) Fruit Extract, Camellia Oleifera (ORGANIC) Black Tea, Algae (Seaweed) Extract,Xanthan Gum (Natural Thickener), Saccharum Officinarum (Sugar Cane), Chlorphenesin, Squalane (Plant Derived), Retinol (Vitamin A), Ubiquinone (Coenzyme Q10), Panthenol (Pro-Vitamin B5), Allantoin (Comfrey Root Derived), Citrus MedicaLimonum (Lemon) Fruit Extract, Citrus Aurantium Dulcis (Sweet Neroli Orange) Fruit, Tetrasodium EDTA, Pyrus Malus (Apple) Fruit Juice, Sodium Hyaluronate, Camellia Sinensis (Green Tea) Leaf Extract, Arachidyl Glucoside, Vitis Vinifera (Grape) SeedExtract, Salix Alba (Willow) Bark Extract, Vaccinium Myrtillus (Bilberry) Extract, Phyllanthus Emblica (Amla) Extract, Thioctic Acid (a-Lipoic Acid), Sodium Hydroxide (pH Modifier)

Read more here:
AnteAge Stem Cell Skin Care Reviewed

Stem Cell Transplants in Cancer Treatment – National …

Stem cell transplants are procedures that restore blood-forming stem cells in people who have had theirs destroyed by the very high doses of chemotherapy or radiation therapy that are used to treat certain cancers.

Blood-forming stem cells are important because they grow into different types of blood cells. The main types of blood cells are:

You need all three types of blood cells to be healthy.

In a stem cell transplant, you receive healthy blood-forming stem cells through a needle in your vein. Once they enter your bloodstream, the stem cells travel to the bone marrow, where they take the place of the cells that were destroyed by treatment. The blood-forming stem cells that are used in transplants can come from the bone marrow, bloodstream, or umbilical cord. Transplants can be:

To reduce possible side effects and improve the chances that an allogeneic transplant will work, the donors blood-forming stem cells must match yours in certain ways. To learn more about how blood-forming stem cells are matched, see Blood-Forming Stem Cell Transplants.

Stem cell transplants do not usually work against cancer directly. Instead, they help you recover your ability to produce stem cells after treatment with very high doses of radiation therapy, chemotherapy, or both.

However, in multiple myeloma and some types of leukemia, the stem cell transplant may work against cancer directly. This happens because of an effect called graft-versus-tumor that can occur after allogeneic transplants. Graft-versus-tumor occurs when white blood cells from your donor (the graft) attack any cancer cells that remain in your body (the tumor) after high-dose treatments. This effect improves the success of the treatments.

Stem cell transplants are most often used to help people with leukemia and lymphoma. They may also be used for neuroblastoma and multiple myeloma.

Stem cell transplants for other types of cancer are being studied in clinical trials, which are research studies involving people. To find a study that may be an option for you, see Find a Clinical Trial.

The high doses of cancer treatment that you have before a stem cell transplant can cause problems such as bleeding and an increased risk of infection. Talk with your doctor or nurse about other side effects that you might have and how serious they might be. For more information about side effects and how to manage them, see the section on side effects.

If you have an allogeneic transplant, you might develop a serious problem called graft-versus-host disease. Graft-versus-host disease can occur when white blood cells from your donor (the graft) recognize cells in your body (the host) as foreign and attack them. This problem can cause damage to your skin, liver, intestines, and many other organs. It can occur a few weeks after the transplant or much later. Graft-versus-host disease can be treated with steroids or other drugs that suppress your immune system.

The closer your donors blood-forming stem cells match yours, the less likely you are to have graft-versus-host disease. Your doctor may also try to prevent it by giving you drugs to suppress your immune system.

Stem cells transplants are complicated procedures that are very expensive. Most insurance plans cover some of the costs of transplants for certain types of cancer. Talk with your health plan about which services it will pay for. Talking with the business office where you go for treatment may help you understand all the costs involved.

To learn about groups that may be able to provide financial help, go to the National Cancer Institute database, Organizations that Offer Support Services and search "financial assistance." Or call toll-free 1-800-4-CANCER (1-800-422-6237) for information about groups that may be able to help.

When you need an allogeneic stem cell transplant, you will need to go to a hospital that has a specialized transplant center. The National Marrow Donor Program maintains a list of transplant centers in the United States that can help you find a transplant center.

Unless you live near a transplant center, you may need to travel from home for your treatment. You might need to stay in the hospital during your transplant, you may be able to have it as an outpatient, or you may need to be in the hospital only part of the time. When you are not in the hospital, you will need to stay in a hotel or apartment nearby. Many transplant centers can assist with finding nearby housing.

A stem cell transplant can take a few months to complete. The process begins with treatment of high doses of chemotherapy, radiation therapy, or a combination of the two. This treatment goes on for a week or two. Once you have finished, you will have a few days to rest.

Next, you will receive the blood-forming stem cells. The stem cells will be given to you through an IV catheter. This process is like receiving a blood transfusion. It takes 1 to 5 hours to receive all the stem cells.

After receiving the stem cells, you begin the recovery phase. During this time, you wait for the blood cells you received to start making new blood cells.

Even after your blood counts return to normal, it takes much longer for your immune system to fully recoverseveral months for autologous transplants and 1 to 2 years for allogeneic or syngeneic transplants.

Stem cell transplants affect people in different ways. How you feel depends on:

Since people respond to stem cell transplants in different ways, your doctor or nurses cannot know for sure how the procedure will make you feel.

Doctors will follow the progress of the new blood cells by checking your blood counts often. As the newly transplanted stem cells produce blood cells, your blood counts will go up.

The high-dose treatments that you have before a stem cell transplant can cause side effects that make it hard to eat, such as mouth sores and nausea. Tell your doctor or nurse if you have trouble eating while you are receiving treatment. You might also find it helpful to speak with a dietitian. For more information about coping with eating problems see the booklet Eating Hints or the section on side effects.

Whether or not you can work during a stem cell transplant may depend on the type of job you have. The process of a stem cell transplant, with the high-dose treatments, the transplant, and recovery, can take weeks or months. You will be in and out of the hospital during this time. Even when you are not in the hospital, sometimes you will need to stay near it, rather than staying in your own home. So, if your job allows, you may want to arrange to work remotely part-time.

Many employers are required by law to change your work schedule to meet your needs during cancer treatment. Talk with your employer about ways to adjust your work during treatment. You can learn more about these laws by talking with a social worker.

Follow this link:
Stem Cell Transplants in Cancer Treatment - National ...

Apple Stem Cells – The Anti-Aging skin care ingredient …

What are Stem Cells?

Stem cells are super unique in that they have the ability to go through numerous cycles and cell divisions while maintaining the undifferentiated state. Primarily, stem cells are capable of self-renewal and can transform themselves into other cell types of the same tissue. Their crucial role is to replenish dying cells and regenerate damaged tissue. Stem cells have a limited life expectation due to environmental and intrinsic stress factors. Because their life is endangered by internal and external stresses, stem cells have to be protected and supported to delay preliminary aging. In aged bodies, the number and activity of stem cells in reduced.

Until several years ago, the tart, unappealing breed of the Swiss-grown Uttwiler Sptlauber apples, did not seem to offer anything of value. That was until Swiss scientists discovered the unusual longevity of the stem cells that kept these apples alive months after other apples shriveled and fell off their trees. In the rural region of Switzerland, home of these magical apples, it was discovered that when the unpicked apples or tree bark was punctured, Swiss Apple trees have the ability to heal themselves and last longer than other varieties. What was the secret to these apples prolonged lives?

Proven to Diminish the Signs of Aging

These scientists got to work to find out. What they revealed was that apple stem cells work just like human stem cells, they work to maintain and repair skin tissue. The main difference is that unlike apple stem cells, skin stem cells do not have a long lifespan, and once they begin depleting, the signs of aging start kicking in (in the forms of loose skin, wrinkles, the works). Time to harness these apple stem cells into anti aging skin care! Not so fast. As mentioned, Uttwiler Sptlauber apples are now very rare to the point that the extract can no longer be made in a traditional fashion. The great news is that scientists developed a plant cell culture technology, which involves breeding the apple stem cells in the laboratory.

Human stem cells on the skins epidermis are crucial to replenish the skin cells that are lost due to continual shedding. When epidermal stem cells are depleted, the number of lost or dying skin cells outpaces the production of new cells, threatening the skins health and appearance.

Like humans, plants also have stem cells. Enter the stem cells of the Uttwiler Sptlauber apple tree, whose fruit demonstrates an exceptionally long shelf-life. How can these promising stem cells help our skin?

Studies show that apple stem cells boosts production of human stem cells, protect the cell from stress, and decreases wrinkles. How does it work? The internal fluid of these plant cells contains components that help to protect and maintain human stem cells. Apple stem cells contain metabolites to ensure longevity as the tree is known for the fact that its fruit keep well over long periods of time.

When tested in vitro, the apple stem cell extract was applied to human stem cells from umbilical cords and was found to increase the number of the stem cells in culture. Furthermore, the addition of the ingredient to umbilical cord stem cells appeared to protect the cells from environmental stress such as UV light.

Apple stem cells do not have to be fed through the umbilical cord to benefit our skin! The extract derived from the plant cell culture technology is being harnessed as an active ingredient in anti aging skincare products. When delivered into the skin nanotechnology, the apple stem cells provide more dramatic results in decreasing lines, wrinkles, and environmental damage.

Currently referred to as The Fountain of Youth, intense research has proved that with just a concentration level of 0.1 % of the PhytoCellTec (apple stem cell extract) could proliferate a wealth of human stem cells by an astounding 80%! These wonder cells work super efficiently and are completely safe. Of the numerous benefits of apple stems cells, the most predominant include:

See the original post here:
Apple Stem Cells - The Anti-Aging skin care ingredient ...

Stem Cell Use in Skin Care Products? – Science of Skincare

The science behind skin care has been progressing at a faster and faster rate of speed. Twenty years ago, had you mentioned stem cell use in association with mainstream skin care, people would have stared at you as though you had three heads and steered their children in a path far around you.

Reality today paints a much cooler picture. One where stem cells are used to treat a variety of blood and bone marrow diseases, blood cancers, and immune disorders. And we are finding stem cells, both human and plant, on the ingredients lists of some very powerful and effective skin care products. Stem cell use in skin care products is coming of age.

Stem cells are a type of cell that are found in all living things and have the glorious ability to differentiate themselves into many different types of cells. They are capable of becoming any other type of cell in that type of organism and reproducing in a controlled manner. As a result, they are the building blocks of your tissues and have the unique ability to replace damaged and diseased cells. They can proliferate for long periods, dividing themselves over and over again into millions of new cells. That means they can play a pivotal role in how skin repairs itself.

Stem cells are extremely beneficial in the natural process of healing and regeneration, says Jessica Weiser, M.D., a board-certified dermatologist in New York City.

Many beauty products contain stem cells from fruits like Swiss apples, edelweiss, roses, date palms, grape, raspberry, lilac, and gotu kola that have the ability to stay fresh for long periods of times.

Human stem cells come from one of two sources: embryonic stem cells and adult (somatic) stem cells. For the case of skin care, stem cells of the adult origin are used. They remain in the body quietly in a non-dividing state for years until activated by disease or injury.

Because they play an essential role in tissue removal, stem cells residing just below the surface of the skin can help with restorative functions, such as cellular regeneration, and could play a vital role in helping to enhance our ability to repair aging skin.

You start off with an abundance of stem cells in your skin, but you lose them as you age. By the time you hit 50, youve lost about 98% of them.

The working theory is that by applying products containing stem cell extracts, you could encourage the growth of your own skins stem cells and possibly wake them up to trigger their anti-aging effects. Some research suggests that they can promote the production of collagen, which is the bodys firming protein.

Live cells need very specific conditions to remain alive and viable. Its difficult enough to maintain those conditions in a laboratory setting. Skin care products and their environments dont offer those types of conditions. When stem cells are included in skin care products, makers arent looking to provide you with live, functional cells. Extracts from the stem cells, not the actual cells themselves, are usually added to skin care products. Its not possible to maintain live stem cells in cosmetic emulsions, says Zoe Diana Draelos, a consulting professor of dermatology at the Duke University School of Medicine in Durham, North Carolina.

Most stem cell products you see on the shelf dont actually contain stem cells, but rather the proteins and amino acids that those cells secrete. Typically, if you see a product labeled as a stem cell product, youll see the stem cells key substances in the ingredients list. These include ferulic acid, ellagic acid, and quercetin. This is what your body is able to recognize and put to use to help rejuvenate and repair cells. Human stem cell byproducts (from skin or adipose tissue) seem to be the best solution for use in skin care products because of their ability to produce the same types of cellular components that your body uses naturally to maintain a youthful appearance.

Cultivating stem cells is a tedious process involving a very controlled environment without any contaminants in order to yield the most potent, stable, and pure extract. Because of this technology, the cost of stem cell products are usually greater than products without.

MDSUN is a perfect collaboration between medicine and beauty with the ability to deliver the highest quality skin care products, giving you long-lasting radiance and youth. Each formulation is effective, while free of harsh ingredients, perfumes, or chemical scent additives.

They offer multiple options incorporating powerful stem cell technology with proven effective results. The Wrinkle Smoothener reduces wrinkle depth and improves skins texture while quenching skin-damaging free radicals. It can stimulate skin repair and diminish the appearance of aging skin.

The Collagen Lift is a very potent treatment that can deliver obvious results, minimizing the appearance of wrinkles and lines, improving skin texture and tone. This luxurious gel-cream soothes redness and irritations and rejuvenates skin cells for a strong and long-lasting radiant renewal.

The Med-Eye Complex Cream visibly promotes firmness, increases blood circulation and deeply hydrates the eye area to reduce the signs of aging, lending a youthful appearance and glow.

Here is the original post:
Stem Cell Use in Skin Care Products? - Science of Skincare

Stem Cells Used in Anti-Aging Skin Care Radiant RG-Cell

Stem cells are biological cells that are able to stay dormant until triggered to reproduce into new tissue. Found in human embryos and in adult tissue, they can form into any cell type, and help repair organs and skin in the case of injury or other cause of damage.

So is it any surprise that their potential is also being trumpeted in the world of skin care? Cosmetic science has often taken inspiration from hard-core medical breakthroughs, and stem cells appear to possess the ideal skill set to throw the switch on a veritable fountain of youth.

While skin stem cells have found use in treating diseases, stem cells technology in skin care products have been largely based on hype rather than science, but in some cases like RG-CELL, it truly works magic.

The concept of topically applying stem cells, through cream, serum, mask, or facial procedure, with a promise to replenish dying cells and regenerate dying tissues has shown no real scientific evidence that it works.

If youre unfamiliar with the practice, you may question the validity of using live stem cells in anti-aging products when its already an enormous and time consuming challenge to use them in actual organ regenerating procedures.

Firstly, stem cells are highly unstable. They have little to no shelf life. Secondly, they will not enter the deep layers of the skin without an effective skin delivery system. And thirdly, stem cells need specific nutrition via a blood supply in the tissue to survive and function if they were layered onto intact skin the stem cells would just die.

It should be made abundantly clear that, no stem cell skin care products contain actual stem cells. Stem cell based products contain growth factors, along with enzymes and other nutrients, which help the cells grow. Other products dont contain any stem cell-related material at all.

[frame src=https://rg-cell.com/wp-content/uploads/2013/05/stem-cell-skin-care.jpg width=250 height=188 alt=Stem Cell Skin Care align=right]There are 2 ways in which stem cell technology is being used. Firstly, companies are creating products with specialized peptides and enzymes or plant growth factors which, when applied topically on the surface, help protect the human skin from damage and deterioration. Products claiming to contain plant stem cells dont contain human cytokines (or cell messengers), and in fact are really just ground up plant bits. In short, plant stem cell technology cannot effectively impact human stem cells. It can be useful as excellent antioxidants, but marketing has made the benefits bigger than reality.

Secondly, and bearing more scientific evidence, is an alternative application of skin care anti-aging products. These products utilize human stem cell technology, and your skin is the most active participant, NOT plant or apple stem cells. Using ingredients that promote the repair and rejuvenation of your skin by stimulating the activity of your own stem cells in the skin has proven to be safer, more ethical and far more scientifically proven than applying stem cells in a jar. This technology implies a superior product designed specifically to regenerate and rejuvenate your own skin cells.

These products contain epidermal growth factors (EGF) obtained by genetic engineering technology (microbial recombinant) totally identical to natural EGF, known as a BEAUTY FACTOR, boosts and regulates stem cell proliferation. When applied to the skin, stimulate collagen production, improve elasticity, firm sagging skin, improve tone and so much more.

[frame src=https://rg-cell.com/wp-content/uploads/2012/11/nano-encapsulation.jpg width=250 height=190 alt=Skin Delivery System align=right]EGF is a large molecule so it cannot penetrate the skin. In fact, it is too big to fit in between the spaces in cells of our skin. There is also speculation around the length of time, that it can remain stable in a formulation. Clinical studies and research are practically non-existent. Therefore, buyer beware: If you opt for using a product that contains EGF consider whether or not the mechanism of action employed to deliver the ingredient to the dermal layers, will actually work.

Only special technology, can deliver EGF into the skin deeper layers. One of the biggest advances is the use of a patented nano-particulate lipid bi-layer delivery system that allows the products to be delivered deep into the skin where the stem cells live.

RG-Cell uses a unique patented nano-encapsulation technology as its delivery system. This improves the permeation and penetration efficiency of the active ingredients. Owing to this fact, RG-CELL can make valid claims about the efficiency in it is delivery of EGF where it is needed the most. This technology also stabilizes the EGF thereby prolonging its shelf life in the actual product.

Thus we can see that there are already many choices in skin care products with specialized peptides and enzymes or EGFs which, when applied topically stimulate the skins own stem cells. But, only one uses the most advanced technology to deliver nutrients into the skin. Expect many more good choices to be developed in the years to come!

Go here to read the rest:
Stem Cells Used in Anti-Aging Skin Care Radiant RG-Cell

Stem Cell Skin Care – anti-aging cream and hydration Serum

SC21 BioTech: Stem Cell Skin Care Set

SC21 nowoffers a rejuvenating stem cell skin careset that is available to help restore aging skin. At SC21, we have been able to combine human mesenchymal stem cell growth factors, polypeptide complexes, and cytokines, with our day time anti-aging cream & evening hydration serum.

Our SC21 biotechnology scientists have developed a process to isolate potent rejuvenating factors from human stem cells. By resupplying the skin with these powerful missing factors, SC21 Day & Night Stem Cell Skin Care promotes cell renewal, boosts the production of collagen and elastin, restores aging cells, and, ultimately, provides you with more youthful looking skin.

It is important to note that as we age, the stem cell population that is vital in providing healing signals to the skin dramatically diminishes. As a result of this, the rejuvenating components the skin needs to maintain its appearance lessen. By replenishing lost peptides, cytokines & growth factors with the use of a topical product on the skin, we, through the day &night skin care set, are able to effectively re-engage the skins healing process.

The SC21 day & night stem cell skin care rejuvenation set also has a complete solution for restoring aging skin. We have, through the day anti-aging cream & night hydration serum been able to use: human mesenchymal stem cell growth factors, to regenerate human tissues; polypeptide complexes, (which penetrate the epidermis, outer layer of our skin) to send signals to the skin cells and cytokines proteins to send signals between the skin cells.

Focus Ingredient of Growth Factor Skin Care:

Mesenchymal Stem Cell (MSC) Peptide Complex = 15% (cytokines, growth factors, peptide complex)

Other Key Ingredients:

Focus Ingredient of Growth Factor Skin Care:

Mesenchymal Stem Cell (MSC) Peptide Complex = 20%(cytokines, growth factors, peptide complex)

Other Key Ingredients:

Apply 2-3 pumps to clean & dry skin.

Peptides are easier explained as signaling molecules produced by cells to instruct other cells.

As cellular messengers, cytokines influence and control our biological processes from start to finish. There are hundreds of unique cytokines in the human body. Cells talk with cytokines to repair injury, repel microbes, fight infections, and develop immunity.

Growth factors, are, on the other hand, diffusible signaling proteins that stimulate the growth of specific tissues and play a crucial role in promoting cell differentiation and division.

Many modern medical advances, including stem cell breakthroughs, are made possible due to our growing understanding of cytokines & growth factors. We use modern culture techniques (the same type used to produce human insulin and other naturally occurring substances) to grow human stem cells in the laboratory to harvest their regenerative cytokines and growth factors.

Mesenchymal stem cells (MSCs), which are traditionally found in the bone marrow, are used to improve function upon integration because they are self-renewing cells that have the capacity to differentiate, and are capable of replacing and repairing damaged tissues.

MSCs can consequently during culture, produce the following:

Our skin cells are biologically designed to continuously renew themselves, but, starting from our mid 20s, the skin cell renewal process slows down and our skin becomes thinner. This thinning causes us to be more prone to skin damage from external elements.

However, there are other factors that can contribute to our aging process, and in other cases even cause premature aging. Some of these factors include:

More:
Stem Cell Skin Care - anti-aging cream and hydration Serum

Which spare body parts will stem cells deliver first? | Cosmos

On 6 November 1998, the world woke to news of an astonishing discovery. James Thomson and his colleagues at the University of Wisconsin-Madison had generated stem cells from human embryos. Unlike other types of stem cells, these were pluripotent meaning they had the potential to generate any type of body tissue if given the right signals.

For many this news, and the accompanying claims that embryonic stem (ES) cells could revolutionise medicine, appeared to come out of the blue. However, for those of us already working in the stem cell space it was the vital next step in exploring the potential of stem cell science.

Back in 1998, I was a keen PhD student, part of the stem cell research effort at Monash University. I was trying to create pluripotent stem cells from the skin cells of a mouse. The idea was to first clone a mouse embryo from its skin cell and harvest the ES cells. In the lab next door, Ben Reubinoff had been working with Alan Trounson and Martin Pera for several years to see if they could make embryonic stem cells from donated human embryos effectively in parallel to their colleagues in Wisconsin.

There was a lot of excitement about how we might one day be able to use these cells to make replacement body tissues effectively on demand and alleviate suffering for many patients. Although we all recognised this was going to take an enormous amount of effort and time to deliver.

Outside the lab if I mentioned that I worked in stem cell research, I was met with overwhelming curiosity. But people also wondered why we couldnt just use adult stem cells which are found in some of our organs. Many people I spoke to already knew somebody who had been helped by a stem cell transplant using bone marrow or cord blood. Why did we need to use human embryos and ES cells at all?

The reason was, and still is, that adult stem cells are not able to generate any type of tissue because they are not pluripotent. Bone marrow stem cells, for instance, can regenerate an immune system but they cannot regenerate the pancreas or brain tissue. The only source of pluripotent cells was surplus human embryos originally created in an IVF clinic and then donated to research.

In 2007, Japanese scientists made a landmark discovery that side-stepped the need to use embryos. They were able to manipulate ordinary human skin cells to make them pluripotent (a much more elegant and effective approach than my attempts with mice skin cells during my PhD). Dubbed induced pluripotent stem cells or iPSC, these cells share the same desirable features as ES cells. They can be grown in the lab and coaxed to form specific types of body cells.

But both sources of pluripotent stem cells also carry the risk that they could form a tumour if we dont fully direct their developmental fate. Any clinical application must meticulously weed out the stem cells as part of the laboratory recipe used to make the replacement cells. For me, the crucial challenge is how to harness the potential of stem cells to develop safe and effective treatments.

These days, as the head of the outreach and policy program for Stem Cells Australia, a nationwide consortium of Australian stem cell scientists, I spend a lot of my time talking to the public. To some extent Ive become a race caller frequently asked to predict what new treatments are likely to come galloping down the track. Sometimes Im asked to offer an opinion on stem cell treatments that are not on the track at all. Promoted as a sure thing and available now for a price, these interventions lack credible evidence that they work or are even safe. Providers are effectively peddling hope and should be viewed with caution.

Fortunately, we do have providers committed to responsibly advancing the field with lots of bona fide contenders in clinical trials. So with my binoculars firmly in place, here is my reading of whats coming down the track.

Jeffrey Phillips

Leading the charge towards the clinic is a possible treatment for the most common cause of age-related vision loss: macular degeneration. In Australia about one in seven people over the age of 50 have some evidence of this disease. In this condition, damage to the cells at the back of the eye the macula affects central vision and the ability to read, drive and recognise faces. The actual seeing cells in the macula are intact but sight is lost because a tiny underlying patch of darkly pigmented cells are damaged. Known as retinal pigmented epithelial cells or RPE cells, they act like a pit stop team, feeding and clearing away waste for the highly active cells of the retina.

Because the number of RPE cells needed is very small and pluripotent stem cells readily develop into this exact tissue (you can easily spot a patch of darkly pigmented cells in the dish), macular degeneration has long been a favourite. Clinical trials are now underway in the United States, United Kingdom and Japan to determine whether replacing faulty RPE cells with those made in the lab from either human embryonic stem cells or induced pluripotent stem cells could help.

At this early stage, safety is a key concern. The surgical technique to deliver the cells carries the risk of detaching the retina and causing further vision loss. In May 2018, the London Project to Cure Blindness announced that two patients with macular degeneration specifically whats called the wet form due to extensive blood vessel growth under the retina had improved their vision with no significant side-effects after participating in a clinical trial.

Another early entrant in the race to the clinic is type 1 diabetes. Its a disease caused by friendly fire: the immune system seeks and destroys the beta cells of the pancreas. These remarkable cells can both sense rising blood sugar levels and release the exact amount of insulin needed to lower glucose levels to normal. When these cells are destroyed, which often occurs in childhood, the person is no longer able to control their blood sugar levels.

More than 120,000 Australians manage the disease with regular injections of insulin. But they cant regulate their blood sugar levels as precisely as beta cells do. And there are consequences: high blood sugar levels can damage the blood vessels in the heart, eyes and kidneys, while low levels can be fatal. Some patients have been lucky enough to receive a whole pancreas transplant or tissues containing beta cells from cadavers. But there are two problems. First, transplant donors are in short supply. Second, the donated tissue will likely suffer the fate of the original: attack by the immune system.

Enter pluripotent stem cells. Supply is no longer a problem. After two decades of trying, scientists are now able to make large quantities of fully functional beta cells in the lab. And as far as keeping the immune system at bay, several start-up companies have come up with the tea-bag approach. They encase the beta cells in a porous capsule. Like tea leaves, the beta cells are netted in but soluble factors easily move in and out across the net, including insulin and blood-borne glucose as well as other nutrients. Crucially, the net also stops marauding immune cells from getting to the beta cells.

The Californian company, Viacyte, is trialling a teabag about the size and shape of a credit card. Made of surgical-grade polymer, the capsule encases immature beta cells (theyre more robust if they mature inside the body), and is inserted just under the patients skin.

The key challenge, so far, is providing intimate contact with surrounding blood vessels so that the transplanted cells increase in number and survive. In June this year, the company reported its results at a meeting of the American Diabetes Association. Overall, they said there was a low rate of survival, but when cells did survive they produced insulin.

The company is now evaluating a second device that allows the patients blood vessels to grow through the walls of the capsule.

Jeffrey Phillips

A strong stayer in the race to the clinic is Parkinsons disease (PD). Predominantly a disease of ageing, around 1% of people over the age of 60 suffer from it.

The disease results from the death of brain neurons that release the neurotransmitter dopamine. Like a conductor, dopamine ensures different parts of the brain act in synchrony to execute routine movements. Without dopamine, patients have trouble controlling their walking and experience tremors in their hands and other parts of their bodies. Could replacing the faulty dopamine-producing neurons with healthy ones provide a way to combat PD?

More than 20 years ago, a few different research groups around the world gave it a try. Using human foetal tissue, they dissected out the dopamine-producing cells, and surgically implanted these into the brains of patients, specifically in a region called the striatum.

Some patients improved, but others reported significant side effects, particularly uncontrollable jerky movements known as dyskinesia. Questions were asked about whether the correct types of cells were being transferred to the correct part of the brain and further experiments were put on hold. A key question was whether pluripotent stem cells could offer a more precise and reliable source of dopamine-producing cells.

Jump forward to 2018 and several groups are on the cusp of testing new types of replacement cells for PD in a series of clinical trials. Years of research has shown that ES cells and iPS cells can be directed to develop into the correct type of neurons and that sufficiently large numbers can be generated.

When tested in animals, the dopamine-producing cells corrected movement disorders and did not form tumours.

This time around, rather than working in silos, different groups of researchers in Japan, Sweden, UK and US have banded together in a coalition called G-Force PD. Although each group is using a slightly different approach for their clinical trial, by sharing their results and expertise they hope to bring a cell-based therapy for PD closer to reality.

Jeffrey Phillips

Skin stem cells have long been solid performers for growing skin grafts to treat severe burns. But in November 2017, headlines ran hot with a report that a seven-year-old refugee Syrian boy, on the verge of death from a genetic skin condition, had been saved by a graft of skin stem cells corrected by gene therapy.

Hassan, now living with his family in Germany, suffered from a severe form of Epidermolysis Bullosa (EB). Its been referred to as the worst disease youve never heard of. It affects about 500,000 people worldwide, and can be caused by mutations to 18 different genes. In each case, the mutation disrupts the anchoring of the skins upper layer, the epidermis, to the underlying dermis. The result is skin that tears as easily as a butterflys wing. The only treatment is painful bandaging and re-bandaging.

Hassans skin had started blistering from birth but by the time he was seven, a bacterial infection had robbed him of 80% of his skin cover. In a last ditch effort to save his life, his German doctors contacted veteran stem cell researcher Michele De Luca at the University of Modena and Reggio Emilia in Italy. In 2006, De Luca had used skin grafts corrected by gene therapy to treat a leg wound of a woman who suffered from the same form of EB that Hassan suffered from. It was caused by a mutation to a gene called LAMB3.

De Lucas team took a tiny patch of skin containing stem cells from Hassans groin. They also spliced a copy of the LAMB3 gene into a benign virus. Then they infected the skin cells with the virus which ferried the LAMB3 gene into their DNA. The genetically corrected skin grew into a sheet which was grafted onto Hassans body. Five months after the first graft, Hassan was discharged. A month later he was back at school and playing soccer. Thanks to the genetically corrected stem cells, his grafted skin no longer blisters or shreds. The executive director of the Dystrophic Epidermolysis Bullosa Research Association of America dubbed Hassans treatment a sea change to the world of EB. Besides de Lucas group, Peter Marinkovich and Jean Tang at Stanford University School of Medicine, United States, are also trialling genetically-corrected skin grafts for a different type of EB.

Jeffrey Phillips

One of the front runners at the start of the stem cell race was spinal cord injury. Perhaps you remember the actor Christopher Reeve, aka Superman? Following a horse riding accident that left him a quadriplegic, he campaigned tirelessly for researchers to be allowed to use human embryonic stem cells to treat spinal cord injury which claims about 180,000 new cases each year. Perhaps thanks to his efforts in 2010, the world saw the first clinical trial using cells made from human ES cells.

Conducted by the California based biotech company Geron, the researchers had directed ES cells to develop into precursors of oligodendrocytes. These octopus-like cells wind their arms around neurons in the spinal cord to provide electrical insulation as well as nurturing factors. With a spinal cord injury, these important support cells can be lost. Four patients were injected with stem cell-derived oligodendrocyte precursors soon after their injury.

Controversially, Geron discontinued the study in 2011 to refocus their business. Asterias Biotherapeutics picked up the baton and last July, in a company press release, reported the results of an early clinical trial on 25 additional patients who were all injected with oligodendrocyte precursors three to six weeks post-injury. They reported no serious adverse events and that four patients recovered a degree of motor function that may increase their ability to lead an independent life. However, we have to wait to see the peer reviewed published results before we can assess the state of progress.

Beyond replacing oligodendrocytes made from ES cells, other clinical trials are testing different types of cells ranging from neurons obtained from donated foetal tissue to using the patients own cells obtained from the back of the nose where they play an important role in supporting the regeneration of the olfactory neurons. Some types of transplanted cells may act as paramedics, helping damaged motor neurons to recover. Others are designed to directly replace spinal cord neurons.

It remains too early to tell which approach will result in long-term improvements. While many with spinal cord injury are eager for even small improvements such as bladder or bowel control, patients should be careful about trying marketed experimental procedures outside well-conducted clinical trials as they may cause further harm. In a chilling example, one young woman who sought treatment using olfactory cells developed a large, painful mucus-secreting tumour in her spine and no improvement of her paraplegia. Unfortunately, many stem cell cures promoted online, especially for spinal cord injury, lack credibility.

Seeking advice from your medical specialist is the best way to find out more. If they dont know about a trial or claimed treatment, it is probably a mirage.

Jeffrey Phillips

Marked as a long shot for many years, stem cell research is starting to pay dividends for kidney disease. Though its not ready to provide transplants, it is already helping to discover new treatments.

Kidneys are the bodys vital cleansing and balancing system. They filter waste products and toxins from our blood into urine, maintain the bodys water balance and also make hormones important for regulating blood pressure and the production of red blood cells.

Kidney disease, which affects one in 10 Australians, damages the filtration units called nephrons. The major causes are diabetes and high blood pressure. Once gone, the nephrons cannot regenerate. But waiting for a donated kidney can take years; close to 1,000 Australians are currently on the waiting list for a transplant. This health crisis has catapulted researchers into trying to recreate kidney tissue from pluripotent stem cells an immense challenge as these are complex biological machines composed of many interacting parts.

Melissa Littles group, based at the Murdoch Childrens Research Institute in Melbourne, have pioneered this research. In 2015, they successfully grew tiny kidney-like structures that were showcased on the cover of Nature with the headline: Kidney in a dish. While their mini-kidneys possess many of the working parts of a mature kidney, theres a long way to go before they can be used as transplants. The plumbing for example bringing blood in and taking waste out is not yet functional. Also they are tiny, smaller than the tip of your finger.

Nevertheless, these mini-kidneys are already making a difference to our understanding of how kidneys develop and what goes awry in kidney disease, especially the hereditary form. For example, researchers were recently able to make mini-kidneys from a child suffering from a rare genetic condition that can cause end-stage kidney disease. They did it by first generating iPS cells from the childs skin. In the lab they were able to observe structural abnormalities in the childs cells and also showed that when the genetic mutation was corrected, the structural defect was corrected. This provides a new insight into inherited kidney disease where previously we knew very little about how these conditions develop.

Jeffrey Phillips

This article appeared in Cosmos 80 - Spring 2018 under the headline "The stem cell race"

More:
Which spare body parts will stem cells deliver first? | Cosmos

Storing Stem Cells For Life – Smart Cells

One of the bravest moves in that direction has come from stem cell research and therapy. Stem cell therapy is currently being used to successfully treat more than 80 diseases, but the field is rapidly evolving backed by prestigious research and clinical trials.

Smart Cells is the first private UK stem cell storage company to have released stored stem cell units for use in the treatment of children with life-threatening illnesses. We have released the greatest number of samples for use in transplants from the UK.

We believe with the development of technology in the future we will be able to treat even more illnesses.

We believe our customers deserve the best service available and we run our state of the art facility with leading professionals in the field.

We believe that storing your childs stem cells at birth can be a crucial part of treating or curing an unexpected illness.

We believe that in the future this service should be available to every parent, child and family.

We are a company that is for life.

Continued here:
Storing Stem Cells For Life - Smart Cells

Stem Cells – MedicineNet

Stem cell facts

What are stem cells?

Stem cells are cells that have the potential to develop into many different or specialized cell types. Stem cells can be thought of as primitive, "unspecialized" cells that are able to divide and become specialized cells of the body such as liver cells, muscle cells, blood cells, and other cells with specific functions. Stem cells are referred to as "undifferentiated" cells because they have not yet committed to a developmental path that will form a specific tissue or organ. The process of changing into a specific cell type is known as differentiation. In some areas of the body, stem cells divide regularly to renew and repair the existing tissue. The bone marrow and gastrointestinal tract are examples of areas in which stem cells function to renew and repair tissue.

The best and most readily understood example of a stem cell in humans is that of the fertilized egg, or zygote. A zygote is a single cell that is formed by the union of a sperm and ovum. The sperm and the ovum each carry half of the genetic material required to form a new individual. Once that single cell or zygote starts dividing, it is known as an embryo. One cell becomes two, two become four, four become eight, eight become sixteen, and so on, doubling rapidly until it ultimately grows into an entire sophisticated organism composed of many different kinds of specialized cells. That organism, a person, is an immensely complicated structure consisting of many, many, billions of cells with functions as diverse as those of your eyes, your heart, your immune system, the color of your skin, your brain, etc. All of the specialized cells that make up these body systems are descendants of the original zygote, a stem cell with the potential to ultimately develop into all kinds of body cells. The cells of a zygote are totipotent, meaning that they have the capacity to develop into any type of cell in the body.

The process by which stem cells commit to become differentiated, or specialized, cells is complex and involves the regulation of gene expression. Research is ongoing to further understand the molecular events and controls necessary for stem cells to become specialized cell types.

Stem Cells:One of the human body's master cells, with the ability to grow into any one of the body's more than 200 cell types.

All stem cells are unspecialized (undifferentiated) cells that are characteristically of the same family type (lineage). They retain the ability to divide throughout life and give rise to cells that can become highly specialized and take the place of cells that die or are lost.

Stem cells contribute to the body's ability to renew and repair its tissues. Unlike mature cells, which are permanently committed to their fate, stem cells can both renew themselves as well as create new cells of whatever tissue they belong to (and other tissues).

Why are stem cells important?

Stem cells represent an exciting area in medicine because of their potential to regenerate and repair damaged tissue. Some current therapies, such as bone marrow transplantation, already make use of stem cells and their potential for regeneration of damaged tissues. Other therapies that are under investigation involve transplanting stem cells into a damaged body part and directing them to grow and differentiate into healthy tissue.

Embryonic stem cells

During the early stages of embryonic development the cells remain relatively undifferentiated (immature) and appear to possess the ability to become, or differentiate, into almost any tissue within the body. For example, cells taken from one section of an embryo that might have become part of the eye can be transferred into another section of the embryo and could develop into blood, muscle, nerve, or liver cells.

Cells in the early embryonic stage are totipotent (see above) and can differentiate to become any type of body cell. After about seven days, the zygote forms a structure known as a blastocyst, which contains a mass of cells that eventually become the fetus, as well as trophoblastic tissue that eventually becomes the placenta. If cells are taken from the blastocyst at this stage, they are known as pluripotent, meaning that they have the capacity to become many different types of human cells. Cells at this stage are often referred to as blastocyst embryonic stem cells. When any type of embryonic stem cells is grown in culture in the laboratory, they can divide and grow indefinitely. These cells are then known as embryonic stem cell lines.

Fetal stem cells

The embryo is referred to as a fetus after the eighth week of development. The fetus contains stem cells that are pluripotent and eventually develop into the different body tissues in the fetus.

Adult stem cells

Adult stem cells are present in all humans in small numbers. The adult stem cell is one of the class of cells that we have been able to manipulate quite effectively in the bone marrow transplant arena over the past 30 years. These are stem cells that are largely tissue-specific in their location. Rather than typically giving rise to all of the cells of the body, these cells are capable of giving rise only to a few types of cells that develop into a specific tissue or organ. They are therefore known as multipotent stem cells. Adult stem cells are sometimes referred to as somatic stem cells.

The best characterized example of an adult stem cell is the blood stem cell (the hematopoietic stem cell). When we refer to a bone marrow transplant, a stem cell transplant, or a blood transplant, the cell being transplanted is the hematopoietic stem cell, or blood stem cell. This cell is a very rare cell that is found primarily within the bone marrow of the adult.

One of the exciting discoveries of the last years has been the overturning of a long-held scientific belief that an adult stem cell was a completely committed stem cell. It was previously believed that a hematopoietic, or blood-forming stem cell, could only create other blood cells and could never become another type of stem cell. There is now evidence that some of these apparently committed adult stem cells are able to change direction to become a stem cell in a different organ. For example, there are some models of bone marrow transplantation in rats with damaged livers in which the liver partially re-grows with cells that are derived from transplanted bone marrow. Similar studies can be done showing that many different cell types can be derived from each other. It appears that heart cells can be grown from bone marrow stem cells, that bone marrow cells can be grown from stem cells derived from muscle, and that brain stem cells can turn into many types of cells.

Peripheral blood stem cells

Most blood stem cells are present in the bone marrow, but a few are present in the bloodstream. This means that these so-called peripheral blood stem cells (PBSCs) can be isolated from a drawn blood sample. The blood stem cell is capable of giving rise to a very large number of very different cells that make up the blood and immune system, including red blood cells, platelets, granulocytes, and lymphocytes.

All of these very different cells with very different functions are derived from a common, ancestral, committed blood-forming (hematopoietic), stem cell.

Umbilical cord stem cells

Blood from the umbilical cord contains some stem cells that are genetically identical to the newborn. Like adult stem cells, these are multipotent stem cells that are able to differentiate into certain, but not all, cell types. For this reason, umbilical cord blood is often banked, or stored, for possible future use should the individual require stem cell therapy.

Induced pluripotent stem cells

Induced pluripotent stem cells (iPSCs) were first created from human cells in 2007. These are adult cells that have been genetically converted to an embryonic stem celllike state. In animal studies, iPSCs have been shown to possess characteristics of pluripotent stem cells. Human iPSCs can differentiate and become multiple different fetal cell types. iPSCs are valuable aids in the study of disease development and drug treatment, and they may have future uses in transplantation medicine. Further research is needed regarding the development and use of these cells.

Why is there controversy surrounding the use of stem cells?

Embryonic stem cells and embryonic stem cell lines have received much public attention concerning the ethics of their use or non-use. Clearly, there is hope that a large number of treatment advances could occur as a result of growing and differentiating these embryonic stem cells in the laboratory. It is equally clear that each embryonic stem cell line has been derived from a human embryo created through in-vitro fertilization (IVF) or through cloning technologies, with all the attendant ethical, religious, and philosophical problems, depending upon one's perspective.

What are some stem cell therapies that are currently available?

Routine use of stem cells in therapy has been limited to blood-forming stem cells (hematopoietic stem cells) derived from bone marrow, peripheral blood, or umbilical cord blood. Bone marrow transplantation is the most familiar form of stem cell therapy and the only instance of stem cell therapy in common use. It is used to treat cancers of the blood cells (leukemias) and other disorders of the blood and bone marrow.

In bone marrow transplantation, the patient's existing white blood cells and bone marrow are destroyed using chemotherapy and radiation therapy. Then, a sample of bone marrow (containing stem cells) from a healthy, immunologically matched donor is injected into the patient. The transplanted stem cells populate the recipient's bone marrow and begin producing new, healthy blood cells.

Umbilical cord blood stem cells and peripheral blood stem cells can also be used instead of bone marrow samples to repopulate the bone marrow in the process of bone marrow transplantation.

In 2009, the California-based company Geron received clearance from the U. S. Food and Drug Administration (FDA) to begin the first human clinical trial of cells derived from human embryonic stem cells in the treatment of patients with acute spinal cord injury.

What are experimental treatments using stem cells and possible future directions for stem cell therapy?

Stem cell therapy is an exciting and active field of biomedical research. Scientists and physicians are investigating the use of stem cells in therapies to treat a wide variety of diseases and injuries. For a stem cell therapy to be successful, a number of factors must be considered. The appropriate type of stem cell must be chosen, and the stem cells must be matched to the recipient so that they are not destroyed by the recipient's immune system. It is also critical to develop a system for effective delivery of the stem cells to the desired location in the body. Finally, devising methods to "switch on" and control the differentiation of stem cells and ensure that they develop into the desired tissue type is critical for the success of any stem cell therapy.

Researchers are currently examining the use of stem cells to regenerate damaged or diseased tissue in many conditions, including those listed below.

References

REFERENCE:

"Stem Cell Information." National Institutes of Health.

Originally posted here:
Stem Cells - MedicineNet

Embryonic stem cell – Wikipedia

Embryonic stem cells (ES cells or ESCs) are pluripotent stem cells derived from the inner cell mass of a blastocyst, an early-stage pre-implantation embryo.[1][2] Human embryos reach the blastocyst stage 45 days post fertilization, at which time they consist of 50150 cells. Isolating the embryoblast, or inner cell mass (ICM) results in destruction of the blastocyst, a process which raises ethical issues, including whether or not embryos at the pre-implantation stage should have the same moral considerations as embryos in the post-implantation stage of development.[3][4] Researchers are currently focusing heavily on the therapeutic potential of embryonic stem cells, with clinical use being the goal for many labs. These cells are being studied to be used as clinical therapies, models of genetic disorders, and cellular/DNA repair. However, adverse effects in the research and clinical processes have also been reported.

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

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

Under defined conditions, embryonic stem cells are capable of propagating indefinitely in an undifferentiated state. Conditions must either prevent the cells from clumping, or maintain an environment that supports an unspecialized state.[2] While being able to remain undifferentiated, ESCs also have the capacity, when provided with the appropriate signals, to differentiate (presumably via the initial formation of precursor cells) into nearly all mature cell phenotypes.[6]

Due to their plasticity and potentially unlimited capacity for self-renewal, embryonic stem cell therapies have been proposed for regenerative medicine and tissue replacement after injury or disease. Pluripotent stem cells have shown potential in treating a number of varying conditions, including but not limited to: spinal cord injuries, age related macular degeneration, diabetes, neurodegenerative disorders (such as Parkinson's disease), AIDS, etc.[7] In addition to their potential in regenerative medicine, embryonic stem cells provide an alternative source of tissue/organs which serves as a possible solution to the donor shortage dilemma. Not only that, but tissue/organs derived from ESCs can be made immunocompatible with the recipient. Aside from these uses, embryonic stem cells can also serve as tools for the investigation of early human development, study of genetic disease and as in vitro systems for toxicology testing.[5]

According to a 2002 article in PNAS, "Human embryonic stem cells have the potential to differentiate into various cell types, and, thus, may be useful as a source of cells for transplantation or tissue engineering."[8]

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

Current research focuses on differentiating ESCs into a variety of cell types for eventual use as cell replacement therapies (CRTs). Some of the cell types that have or are currently being developed include cardiomyocytes (CM), neurons, hepatocytes, bone marrow cells, islet cells and endothelial cells.[9] However, the derivation of such cell types from ESCs is not without obstacles, therefore current research is focused on overcoming these barriers. For example, studies are underway to differentiate ESCs in to tissue specific CMs and to eradicate their immature properties that distinguish them from adult CMs.[10]

Besides becoming an important alternative to organ transplants, ESCs are also being used in field of toxicology and as cellular screens to uncover new chemical entities (NCEs) that can be developed as small molecule drugs. Studies have shown that cardiomyocytes derived from ESCs are validated in vitro models to test drug responses and predict toxicity profiles.[9] ES derived cardiomyocytes have been shown to respond to pharmacological stimuli and hence can be used to assess cardiotoxicity like Torsades de Pointes.[17]

ESC-derived hepatocytes are also useful models that could be used in the preclinical stages of drug discovery. However, the development of hepatocytes from ESCs has proven to be challenging and this hinders the ability to test drug metabolism. Therefore, current research is focusing on establishing fully functional ESC-derived hepatocytes with stable phase I and II enzyme activity.[18]

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

Yury Verlinsky, a Russian-American medical researcher who specialized in embryo and cellular genetics (genetic cytology), developed prenatal diagnosis testing methods to determine genetic and chromosomal disorders a month and a half earlier than standard amniocentesis. The techniques are now used by many pregnant women and prospective parents, especially couples who have a history of genetic abnormalities or where the woman is over the age of 35 (when the risk of genetically related disorders is higher). In addition, by allowing parents to select an embryo without genetic disorders, they have the potential of saving the lives of siblings that already had similar disorders and diseases using cells from the disease free offspring.[19]

Differentiated somatic cells and ES cells use different strategies for dealing with DNA damage. For instance, human foreskin fibroblasts, one type of somatic cell, use non-homologous end joining (NHEJ), an error prone DNA repair process, as the primary pathway for repairing double-strand breaks (DSBs) during all cell cycle stages.[20] Because of its error-prone nature, NHEJ tends to produce mutations in a cells clonal descendants.

ES cells use a different strategy to deal with DSBs.[21] Because ES cells give rise to all of the cell types of an organism including the cells of the germ line, mutations arising in ES cells due to faulty DNA repair are a more serious problem than in differentiated somatic cells. Consequently, robust mechanisms are needed in ES cells to repair DNA damages accurately, and if repair fails, to remove those cells with un-repaired DNA damages. Thus, mouse ES cells predominantly use high fidelity homologous recombinational repair (HRR) to repair DSBs.[21] This type of repair depends on the interaction of the two sister chromosomes formed during S phase and present together during the G2 phase of the cell cycle. HRR can accurately repair DSBs in one sister chromosome by using intact information from the other sister chromosome. Cells in the G1 phase of the cell cycle (i.e. after metaphase/cell division but prior the next round of replication) have only one copy of each chromosome (i.e. sister chromosomes arent present). Mouse ES cells lack a G1 checkpoint and do not undergo cell cycle arrest upon acquiring DNA damage.[22] Rather they undergo programmed cell death (apoptosis) in response to DNA damage.[23] Apoptosis can be used as a fail-safe strategy to remove cells with un-repaired DNA damages in order to avoid mutation and progression to cancer.[24] Consistent with this strategy, mouse ES stem cells have a mutation frequency about 100-fold lower than that of isogenic mouse somatic cells.[25]

On January 23, 2009, Phase I clinical trials for transplantation of oligodendrocytes (a cell type of the brain and spinal cord) derived from human ES cells into spinal cord-injured individuals received approval from the U.S. Food and Drug Administration (FDA), marking it the world's first human ES cell human trial.[26] The study leading to this scientific advancement was conducted by Hans Keirstead and colleagues at the University of California, Irvine and supported by Geron Corporation of Menlo Park, CA, founded by Michael D. West, PhD. A previous experiment had shown an improvement in locomotor recovery in spinal cord-injured rats after a 7-day delayed transplantation of human ES cells that had been pushed into an oligodendrocytic lineage.[27] The phase I clinical study was designed to enroll about eight to ten paraplegics who have had their injuries no longer than two weeks before the trial begins, since the cells must be injected before scar tissue is able to form. The researchers emphasized that the injections were not expected to fully cure the patients and restore all mobility. Based on the results of the rodent trials, researchers speculated that restoration of myelin sheathes and an increase in mobility might occur. This first trial was primarily designed to test the safety of these procedures and if everything went well, it was hoped that it would lead to future studies that involve people with more severe disabilities.[28] The trial was put on hold in August 2009 due to FDA concerns regarding a small number of microscopic cysts found in several treated rat models but the hold was lifted on July 30, 2010.[29]

In October 2010 researchers enrolled and administered ESTs to the first patient at Shepherd Center in Atlanta.[30] The makers of the stem cell therapy, Geron Corporation, estimated that it would take several months for the stem cells to replicate and for the GRNOPC1 therapy to be evaluated for success or failure.

In November 2011 Geron announced it was halting the trial and dropping out of stem cell research for financial reasons, but would continue to monitor existing patients, and was attempting to find a partner that could continue their research.[31] In 2013 BioTime (AMEX:BTX), led by CEO Dr. Michael D. West, acquired all of Geron's stem cell assets, with the stated intention of restarting Geron's embryonic stem cell-based clinical trial for spinal cord injury research.[32]

BioTime company Asterias Biotherapeutics (NYSE MKT: AST) was granted a $14.3 million Strategic Partnership Award by the California Institute for Regenerative Medicine (CIRM) to re-initiate the worlds first embryonic stem cell-based human clinical trial, for spinal cord injury. Supported by California public funds, CIRM is the largest funder of stem cell-related research and development in the world.[33]

The award provides funding for Asterias to reinitiate clinical development of AST-OPC1 in subjects with spinal cord injury and to expand clinical testing of escalating doses in the target population intended for future pivotal trials.[33]

AST-OPC1 is a population of cells derived from human embryonic stem cells (hESCs) that contains oligodendrocyte progenitor cells (OPCs). OPCs and their mature derivatives called oligodendrocytes provide critical functional support for nerve cells in the spinal cord and brain. Asterias recently presented the results from phase 1 clinical trial testing of a low dose of AST-OPC1 in patients with neurologically-complete thoracic spinal cord injury. The results showed that AST-OPC1 was successfully delivered to the injured spinal cord site. Patients followed 23 years after AST-OPC1 administration showed no evidence of serious adverse events associated with the cells in detailed follow-up assessments including frequent neurological exams and MRIs. Immune monitoring of subjects through one year post-transplantation showed no evidence of antibody-based or cellular immune responses to AST-OPC1. In four of the five subjects, serial MRI scans performed throughout the 23 year follow-up period indicate that reduced spinal cord cavitation may have occurred and that AST-OPC1 may have had some positive effects in reducing spinal cord tissue deterioration. There was no unexpected neurological degeneration or improvement in the five subjects in the trial as evaluated by the International Standards for Neurological Classification of Spinal Cord Injury (ISNCSCI) exam.[33]

The Strategic Partnership III grant from CIRM will provide funding to Asterias to support the next clinical trial of AST-OPC1 in subjects with spinal cord injury, and for Asterias product development efforts to refine and scale manufacturing methods to support later-stage trials and eventually commercialization. CIRM funding will be conditional on FDA approval for the trial, completion of a definitive agreement between Asterias and CIRM, and Asterias continued progress toward the achievement of certain pre-defined project milestones.[33]

The major concern with the possible transplantation of ESC into patients as therapies is their ability to form tumors including teratoma.[34] Safety issues prompted the FDA to place a hold on the first ESC clinical trial, however no tumors were observed.

The main strategy to enhance the safety of ESC for potential clinical use is to differentiate the ESC into specific cell types (e.g. neurons, muscle, liver cells) that have reduced or eliminated ability to cause tumors. Following differentiation, the cells are subjected to sorting by flow cytometry for further purification. ESC are predicted to be inherently safer than IPS cells created with genetically-integrating viral vectors because they are not genetically modified with genes such as c-Myc that are linked to cancer. Nonetheless, ESC express very high levels of the iPS inducing genes and these genes including Myc are essential for ESC self-renewal and pluripotency,[35] and potential strategies to improve safety by eliminating c-Myc expression are unlikely to preserve the cells' "stemness". However, N-myc and L-myc have been identified to induce iPS cells instead of c-myc with similar efficiency.[36]More recent protocols to induce pluripotency bypass these problems completely by using non-integrating RNA viral vectors such as sendai virus or mRNA transfection.

Due to the nature of embryonic stem cell research, there is a lot of controversial opinions on the topic. Since harvesting embryonic stem cells necessitates destroying the embryo from which those cells are obtained, the moral status of the embryo comes into question. Scientists argue that the 5-day old mass of cells is too young to achieve personhood or that the embryo, if donated from an IVF clinic (which is where labs typically acquire embryos from), would otherwise go to medical waste anyway. Opponents of ESC research counter that any embryo has the potential to become a human, therefore destroying it is murder and the embryo must be protected under the same ethical view as a developed human being.[37]

In vitro fertilization generates multiple embryos. The surplus of embryos is not clinically used or is unsuitable for implantation into the patient, and therefore may be donated by the donor with consent. Human embryonic stem cells can be derived from these donated embryos or additionally they can also be extracted from cloned embryos using a cell from a patient and a donated egg.[49] The inner cell mass (cells of interest), from the blastocyst stage of the embryo, is separated from the trophectoderm, the cells that would differentiate into extra-embryonic tissue. Immunosurgery, the process in which antibodies are bound to the trophectoderm and removed by another solution, and mechanical dissection are performed to achieve separation. The resulting inner cell mass cells are plated onto cells that will supply support. The inner cell mass cells attach and expand further to form a human embryonic cell line, which are undifferentiated. These cells are fed daily and are enzymatically or mechanically separated every four to seven days. For differentiation to occur, the human embryonic stem cell line is removed from the supporting cells to form embryoid bodies, is co-cultured with a serum containing necessary signals, or is grafted in a three-dimensional scaffold to result.[50]

Embryonic stem cells are derived from the inner cell mass of the early embryo, which are harvested from the donor mother animal. Martin Evans and Matthew Kaufman reported a technique that delays embryo implantation, allowing the inner cell mass to increase. This process includes removing the donor mother's ovaries and dosing her with progesterone, changing the hormone environment, which causes the embryos to remain free in the uterus. After 46 days of this intrauterine culture, the embryos are harvested and grown in in vitro culture until the inner cell mass forms egg cylinder-like structures, which are dissociated into single cells, and plated on fibroblasts treated with mitomycin-c (to prevent fibroblast mitosis). Clonal cell lines are created by growing up a single cell. Evans and Kaufman showed that the cells grown out from these cultures could form teratomas and embryoid bodies, and differentiate in vitro, all of which indicating that the cells are pluripotent.[41]

Gail Martin derived and cultured her ES cells differently. She removed the embryos from the donor mother at approximately 76 hours after copulation and cultured them overnight in a medium containing serum. The following day, she removed the inner cell mass from the late blastocyst using microsurgery. The extracted inner cell mass was cultured on fibroblasts treated with mitomycin-c in a medium containing serum and conditioned by ES cells. After approximately one week, colonies of cells grew out. These cells grew in culture and demonstrated pluripotent characteristics, as demonstrated by the ability to form teratomas, differentiate in vitro, and form embryoid bodies. Martin referred to these cells as ES cells.[42]

It is now known that the feeder cells provide leukemia inhibitory factor (LIF) and serum provides bone morphogenetic proteins (BMPs) that are necessary to prevent ES cells from differentiating.[51][52] These factors are extremely important for the efficiency of deriving ES cells. Furthermore, it has been demonstrated that different mouse strains have different efficiencies for isolating ES cells.[53] Current uses for mouse ES cells include the generation of transgenic mice, including knockout mice. For human treatment, there is a need for patient specific pluripotent cells. Generation of human ES cells is more difficult and faces ethical issues. So, in addition to human ES cell research, many groups are focused on the generation of induced pluripotent stem cells (iPS cells).[54]

On August 23, 2006, the online edition of Nature scientific journal published a letter by Dr. Robert Lanza (medical director of Advanced Cell Technology in Worcester, MA) stating that his team had found a way to extract embryonic stem cells without destroying the actual embryo.[55] This technical achievement would potentially enable scientists to work with new lines of embryonic stem cells derived using public funding in the USA, where federal funding was at the time limited to research using embryonic stem cell lines derived prior to August 2001. In March, 2009, the limitation was lifted.[56]

The iPSC technology was pioneered by Shinya Yamanakas lab in Kyoto, Japan, who showed in 2006 that the introduction of four specific genes encoding transcription factors could convert adult cells into pluripotent stem cells.[57] He was awarded the 2012 Nobel Prize along with Sir John Gurdon "for the discovery that mature cells can be reprogrammed to become pluripotent." [58]

In 2007 it was shown that pluripotent stem cells highly similar to embryonic stem cells can be generated by the delivery of three genes (Oct4, Sox2, and Klf4) to differentiated cells.[59] The delivery of these genes "reprograms" differentiated cells into pluripotent stem cells, allowing for the generation of pluripotent stem cells without the embryo. Because ethical concerns regarding embryonic stem cells typically are about their derivation from terminated embryos, it is believed that reprogramming to these "induced pluripotent stem cells" (iPS cells) may be less controversial. Both human and mouse cells can be reprogrammed by this methodology, generating both human pluripotent stem cells and mouse pluripotent stem cells without an embryo.[60]

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

However, as a first indication that the induced pluripotent stem cell (iPS) cell technology can in rapid succession lead to new cures, it was used by a research team headed by Rudolf Jaenisch of the Whitehead Institute for Biomedical Research in Cambridge, Massachusetts, to cure mice of sickle cell anemia, as reported by Science journal's online edition on December 6, 2007.[61][62]

On January 16, 2008, a California-based company, Stemagen, announced that they had created the first mature cloned human embryos from single skin cells taken from adults. These embryos can be harvested for patient matching embryonic stem cells.[63]

The online edition of Nature Medicine published a study on January 24, 2005, which stated that the human embryonic stem cells available for federally funded research are contaminated with non-human molecules from the culture medium used to grow the cells.[64] It is a common technique to use mouse cells and other animal cells to maintain the pluripotency of actively dividing stem cells. The problem was discovered when non-human sialic acid in the growth medium was found to compromise the potential uses of the embryonic stem cells in humans, according to scientists at the University of California, San Diego.[65]

However, a study published in the online edition of Lancet Medical Journal on March 8, 2005 detailed information about a new stem cell line that was derived from human embryos under completely cell- and serum-free conditions. After more than 6 months of undifferentiated proliferation, these cells demonstrated the potential to form derivatives of all three embryonic germ layers both in vitro and in teratomas. These properties were also successfully maintained (for more than 30 passages) with the established stem cell lines.[66]

Read the rest here:
Embryonic stem cell - Wikipedia

New CRISPR Approach Converts Skin Cells into Pluripotent …

Scientists say that for the first time they have been able to convertskin cells into pluripotent stem cells by activating the cells' own genes. The team reportedly used a type of CRISPRa gene-editing technology that does not cut DNA and can activate gene expression without mutating the genome.Up till now, reprogramming has only been possible by introducing the critical genes for the conversion, called Yamanaka factors, artificially into skin cells where they are not normally active.

The study (Human Pluripotent Reprogramming with CRISPR Activators)is published in Nature Communications.

CRISPR-Cas9-based gene activation (CRISPRa) is an attractive tool for cellular reprogramming applications due to its high multiplexing capacity and direct targeting of endogenous loci. Here we present the reprogramming of primary human skin fibroblasts into induced pluripotent stem cells (iPSCs) using CRISPRa, targeting endogenousOCT4,SOX2,KLF4,MYC, andLIN28Apromoters. The low basal reprogramming efficiency can be improved by an order of magnitude by additionally targeting a conserved Alu-motif enriched near genes involved in embryo genome activation (EEA-motif). This effect is mediated in part by more efficient activation ofNANOGandREX1,write the investigators.

These data demonstrate that human somatic cells can be reprogrammed into iPSCs using only CRISPRa. Furthermore, the results unravel the involvement of EEA [EGA-enriched Alu-motif]-motif-associated mechanisms in cellular reprogramming.

"CRISPR/Cas9 can be used to activate genes. This is an attractive possibility for cellular reprogramming because multiple genes can be targeted at the same time. Reprogramming based on activation of endogenous genes rather than overexpression of transgenes is also theoretically a more physiological way of controlling cell fate and may result in more normal cells. In this study, we show that it is possible to engineer a CRISPR activator system that allows robust reprogramming of iPSCs, saysTimo Otonkoski, M.D., Ph.D., at the University of Helsinki.

An important key for success was also activating a critical genetic element that was earlier found to regulate the earliest steps of human embryo development after fertilization. "Using this technology, pluripotent stem cells were obtained that resembled very closely typical early embryonal cells, addsJuha Kere, M.D., Ph.D., at the Karolinska Institute and King's College London.

The discovery also suggests that it might be possible to improve many other reprogramming tasks by addressing genetic elements typical of the intended target cell type.

The technology may find practical use in biobanking and many other tissue technology applications, notes doctoral student Jere Weltner, the first author of the article."In addition, the study opens up new insights into the mechanisms controlling early embryonic gene activation."

Read more here:
New CRISPR Approach Converts Skin Cells into Pluripotent ...

Skin & Human Stem Cells – BareFacedTruth.com

We have a lot of knowledge to share with you about stem cells and their value in skin care. We thought we would start with a current review of ongoing work in human stem cell science to give you some context. In the next few days we will be getting a lot more specific about wound healing, anti-aging, and related applications.

Human Stem Cells: Introduction

Future advances in many medical fields are thought to be dependent on continued progress in stem cell research. In this section, BTF briefly looks at the future of stem cell based therapies in the treatment of traumatic injury, degenerative diseases, and other ailments, and concludes with a review of current cell based therapies (stem cell and non-stem cell) in the field of skin care.

While the possible indications for stem cell based therapies are numerous,the field of stem cell science is young and years (or decades) may pass before todays promising laboratory results translate into useful clinical treatments. Only time will tell whether successes evolve or remain frustratingly elusive. We do know that success is possible.

The first stem cell therapy was bone marrow transplantation, originally accomplished in the mid 1960s. Last year, there were more than 50,000 such transplants worldwide. In earlier years, infusion of filtered bone marrow cells was performed with stem cells comprising but a very small part of the volume. Newer techniques have made it possible to separate cellular types to enable use of much higher concentrations of stem cells.

Much progress has been made in characterizing stem cells and understanding how they function. There is much more to the story than differentiation into tissue specific cells. Recent research shows that perhaps even more important is the fact that stem cells, especially certain types of stem cells, communicate with the cells around them by producing cellular signals called cytokines, of which there are hundreds.

Cytokines trigger specific receptors on cell membranes that result in precise responses. This phenomenon is considered an essential element in the healing response of all tissues. Identifying and characterizing the large number of cytokines is an important part of stem cell research.

Not every induced response is necessarily beneficial. It is the symphony of responses that is important. How to promote helpful responses while inhibiting non-beneficial ones is a continuing focus of cellular biochemical research as well as the basis upon which drug companies spend huge resources developing drugs to either trigger or block particular cytokine receptors. Good examples in the field of dermatology are EGFR (epidermal growth factor receptor) blocking compounds for use in treating susceptible cells, most notably cancers stimulated by EGF.

Potential Treatments

Stem cell therapies hold potential to treat many conditions and diseases that affect millions of people in the U.S.

From the Laboratory to the Bedside

Going from the research laboratory to the bedside takes time. Only one month ago, the FDA granted marketing approval for the first licensed stem cell product. Derived from donated umbilical cord blood, the product contains stem cells that can restore a recipients blood cell levels and function. In the chart below, the type of cells recovered from umbilical cord blood are those designated as HSC cell. They are the exact cells responsible for the success of bone marrow transplantation.

Of particular note are the cells designated in the chart as MSC or mesenchymal stem cells. MSC cells are the focus of intense research in the treatment of a number of conditions because this type of stem cell can differentiate into a variety of cell types including bone, cartilage, muscles, nerve, and skin (fibroblast.)

Recent announcements about stem cells being used to fabricate replacement parts (bone, cartilage, heart muscle) are based on MSC research. They truly are the duct tape of the bodys repair tool box; a phrase coined because of their importance in the healing of injuries.

Research has shown MSC cells reside in a number of tissues, including the bone marrow. Through precise chemical signaling that originate from sites of injury, MSC cells have the ability to become mobile, enter the blood stream and travel through the circulation to the injury. Upon arrival, MSCs orchestrate the healing response. Local resident stem cells are also called into action, to produce more stem cells or to produce needed tissue specific cells. In large part, MSCs accomplish their tasks bio-chemically.

Secreted cytokines have been identified as themajormechanism by which MSCs perform their important reparative functions. There are hundreds of cytokines identified thus far. The healing response is an intricate and balanced process in which many cytokines participate.

Despite their inherent ability to differentiate into essentially any type of cell, embryonic stem cells are unlikely to be a major research focus in the foreseeable future. Ethical and political considerations limit the acceptability of their use. Federal regulations permit research only on existing cell lines which are few in number. It is difficult to see how this prohibition will end any time soon.

Getting Closer butNot There Yet

MSC (mesenchymal stem cell) therapies include use ofcellsanduse of MSC factors, the cytokines or chemical messengers mentioned above. Methods of administration will likely include intravenous infusion, injections into tissues or body spaces, or development of drugs that activate or block certain cytokine effects. Drugs already in development include epidermal growth factor receptor (EGFR) blockers for use in cancer treatment.

Stem Cells and Skin Health

From fetal life to death, the numbers and activity of stem cells diminish. The chart at left shows how the population of mesenchymal stem cells in the bone marrow dwindles with age.

Knowing that stem cells are important in producing differentiated daughter cells (such as fibroblasts within the dermis) and are instrumental in orchestrating the bodys response to injury, it is easy to understand how skin damage from sun exposure, gravity, smoking, trauma, toxins, even repetitive facial movement, accumulates over time.

This is one line of evidence (we will look at others) that mesenchymal stem cells (or more specifically the relative lack of same) has a lot to do with aging. Skin aging included.

Products Claiming to Activate Skin Stem Cells

The number of skin products claiming to activate human skin stem cells is large and growing. As discussed previously on BFT, a whole slew of plant derived stem cell products are being marketing, NONE of which can actually or theoretically activate anything, especially not a human stem cell.

Other products claim to have essential nutrients or antioxidants or some other magical ingredient that will suddenly make stem cells take notice and unleash their regenerative power. It is highly unlikely, except in the most extreme case of malnourishment, that any nutrient or antioxidant is deficient enough to cause a cell not to function.

These and the botanical stem cell products are marketing ploys. Human stem cells deep within the dermis will never know whether or not these substances are applied. Moisturizers and other recognized ingredients in these products can be beneficial to skin appearancebut not because a stem cell is involved.

This is worse than junk science. This is scamming.

View original post here:
Skin & Human Stem Cells - BareFacedTruth.com

Stem Cell Basics – ISSCR

The human body comprises more than 200 types of cells, and every one of these cell types arises from the zygote, the single cell that forms when an egg is fertilized by a sperm. Within a few days, that single cell divides over and over again until it forms a blastocyst, a hollow ball of 150 to 200 cells that give rise to every single cell type a human body needs to survive, including the umbilical cord and the placenta that nourishes the developing fetus.

Each cell type has its own size and structure appropriate for its job. Skin cells, for example, are small and compact, while nerve cells that enable you to wiggle your toes have long, branching nerve fibers called axons that conduct electrical impulses.

Cells with similar functionality form tissues, and tissues organize to form organs. Each cell has its own job within the tissue in which it is found, and all of the cells in a tissue and organ work together to make sure the organ functions properly.

Regardless of their size or structure, all human cells start with these things in common:

Stem cells are the foundation of development in plants, animals and humans. In humans, there are many different types of stem cells that come from different places in the body or are formed at different times in our lives. These include embryonic stem cells that exist only at the earliest stages of development and various types of tissue-specific (or adult) stem cells that appear during fetal development and remain in our bodies throughout life.

Stem cells are defined by two characteristics:

Beyond these two things, though, stem cells differ a great deal in their behaviors and capabilities.

Embryonic stem cells are pluripotent, meaning they can generate all of the bodys cell types but cannot generate support structures like the placenta and umbilical cord.

Other cells are multipotent, meaning they can generate a few different cell types, generally in a specific tissue or organ.

As the body develops and ages, the number and type of stem cells changes. Totipotent cells are no longer present after dividing into the cells that generate the placenta and umbilical cord. Pluripotent cells give rise to the specialized cells that make up the bodys organs and tissues. The stem cells that stay in your body throughout your life are tissue-specific, and there is evidence that these cells change as you age, too your skin stem cells at age 20 wont be exactly the same as your skin stem cells at age 80.

Go here to read the rest:
Stem Cell Basics - ISSCR

Stem Cells Can Create Skin For Burn Victims | IFLScience

When burn victims need a skin graft they typically have to grow skin on other parts of their bodies - a process that can take weeks. A new technique uses stem cells derived from the umbilical cord to generate new skin much more quickly. The results were published in Stem Cells Translational Medicine by lead author Ingrid Garzn from the University of Granadas Department of Histology.

Not only can the stem cells develop artificial skin more quickly than regular normal skin growth, but the skin can also be stored so it is ready right when it is needed. Tens of thousands of grafts are performed each year for burn victims, cosmetic surgery patients, and for people with large wounds having difficulty healing. Traditionally, this involves taking a large patch of skin (typically from the thigh) and removing the dermis and epidermis to transplant elsewhere on the body.

The artificial skin requires the use of Wharton's jelly mesenchymal stem cells. As the name implies, Whartons jelly is a gelatinous tissue in the umbilical cord that contains uncommitted mesenchymal stemcells (MSC). The MSC is then combined with agarose(a polysaccharide polymer) and fibrin (the fibrous protein that aids in blood clotting). This yielded two results: skin and the mucosal lining of the mouth. The researchers are very pleased to have found two new uses for the stem cells of Whartons jelly, which have not previously been researched for epithelial applications.

Once the epithelial tissues have been created, researchers can store it in tissue banks. If someone is brought into the hospital following a devastating burn or accident, the tissue is ready to graft immediately; not in a few weeks. However, the stem-cell skin is not able to fully differentiate in vitro. After the graft, it has complete cell-cell junctions and will develop all of the necessary layers of normal epithelial tissue.

The MSCs are taken from the umbilical cord after the baby has been born, which poses no risk to either the mother or the child. This method is relatively inexpensive and has been shown to be more efficient than stem cells derived from bone marrow.

Read more here:
Stem Cells Can Create Skin For Burn Victims | IFLScience

Printing Skin Cells on Burn Wounds – Wake Forest School of …

Skin is the body's largest organ. Loss of the skin barrierresults in fluid and heat loss and the risk of infection. Thetraditional treatment for deep burns is to cover them with healthyskin harvested from another part of the body. But in cases ofextensive burns, there often isn't enough healthy skin toharvest.

During phase I of AFIRM, WFIRM scientists designed, built andtested a printer designed to print skin cells onto burn wounds. The"ink" is actually different kinds of skin cells. A scanner is usedto determine wound size and depth. Different kinds of skin cellsare found at different depths. This data guides the printer as itapplies layers of the correct type of cells to cover the wound. Youonly need a patch of skin one-tenth the size of the burn to growenough skin cells for skin printing.

During Phase II of AFIRM, the WFIRM team will explore whether atype of stem cell found in amniotic fluid and placenta (afterbirth)is effective at healing wounds. The goal of the project is to bringthe technology to soldiers who need it within the next 5 years.

This video -- with a mock hand and burn -- demonstrates the process.

More here:
Printing Skin Cells on Burn Wounds - Wake Forest School of ...

Genetically modified skin grown from stem cells saved a 7 …

Scientists reported Wednesday that they genetically modified stem cells to grow skinthat they successfully grafted over nearly all of a child's body a remarkable achievement thatcouldrevolutionize treatment of burn victims and people with skin diseases.

The research, published in the journalNature, involved a 7-year-old boy who suffers from a genetic disease known as junctional epidermolysis bullosa (JEB)that makes skin so fragile that minor friction such as rubbing causes the skin to blister or come apart.

By the time the boy arrived at Children's Hospital of Ruhr-University in Germany in 2015, he wasgravely ill.Doctors noted that hehad complete epidural loss on about 60 percent of his body surface area, was in so much pain that he was on morphine, and fighting off a systemic staph infection. The doctors triedeverything they could think of: antibiotics, changing dressings, grafting skin donated by his father. But nothing worked, and they told his parents to prepare for the worst.

We had a lot of problems in the first days keeping this kid alive, Tobias Hirsch, one of the treating physicians, recalled in a conference call with reporters this week.

Gene therapy to treat a skin disease. (Nature News & Views)

Hirsch and his colleague Tobias Rothoeft began to scour the medical literature foranything that might help and came acrossanarticle describing a highlyexperimental procedure to genetically engineer skin cells.They contacted the author, Michele De Luca, of the Center for Regenerative Medicine at the University of Modena and Reggio Emilia in Italy. De Luca flew out right away.

Using a technique he had used only twice before and even then only on small parts of the body,De Luca harvested cells froma four-square-centimeter patch of skin on anunaffected part of the boy's body and brought them into the lab. There, he genetically modified them so that they no longer contained the mutated form of a gene known to cause the disease and grew the cells into patches of genetically modified epidermis. They discovered, the researchers reported, that the human epidermis is sustained by a limited number of long-lived stem cells which are able to extensively self-renew.

In three surgeries, the child's doctorstook that lab-grownskin and used it to cover nearly 80 percent ofthe boy's body mostly on the limbs and on his back, which had suffered the most damage. The procedure was permitted under a compassionate useexception that allows researchers under certain dire circumstances to make a treatment available even though it is not approved by regulators for general use. Then, over the course of the nexteight months while thechild was in the intensive care unit, they watched and waited.

The boy'srecovery was stunning.

The regenerated epidermis firmly adhered to the underlying dermis, the researchers reported. Hair follicles grew out of some areas. And even bumps and bruises healed normally. Unlike traditional skin grafts that requireointmentonce or twice a day to remain functional, the boy's new skin was fine with the normal amount of washing and moisturizing.

The epidermis looks basically normal. There is no big difference, De Luca said. He said he expects the skin to last basically the life of the patient.

In an analysis accompanying themain article in Nature, Mariacelest Aragona and Cedric Blanpain wrote that this therapy appears to be one of the few examples of trulyeffective stem-cell therapies. The study demonstrates the feasibility and safety of replacing the entire epidermis using combined stem-cell and gene therapy, and also provides important insights into how different types of cellswork together to help ourskin renew itself.

They said there are still many other lingering questions, including whether such procedures might work better in children than adults and whether there would be longer-term adverseconsequences, such as the development ofcancer.

There are also manychallenges to translating this research to treating wounds sustained in fires or other violent ways. In the skin disease that was treated in the boy, the epidermis is damaged but the layer beneath it, the dermis, is intact. The dermis is what the researchers called an ideal receiving bed for the lab-grown skin. But if deeper layers of the skin are burned or torn off, it's possible that the artificial skin would not adhere as well.

No matter how you prepare, its a bad situation, De Luca said. For the time being, he says he's continuingto study the procedure in two clinical trials that involve genetic diseases.

Meanwhile, Hirsch and Rothoeft report that the boy is continuing to do well and is not on any medication for the first time in many years. Doctors are carefully monitoring the child for any signs that there may be some cells that were not corrected and that the disease may reemerge, but right now that does not appear to be happening in the transplanted areas. However, the child does have some blisteringin about 2 to 3 percent of his body in non-grafted areas, and they are considering whether to replace that skin as well.

But for now, they are giving the boy time to be a boy, Rothoeft said: The kid is now back to school and plays soccer and spends other days with the children.

Read more:

New evidence of brain damage from West Nile virus years after bite, scientists say

She signed up to be a surrogate mother and unwittingly gave away her own child

Toddler hospitalized after his fathers arrest postponed kidney donation

Read the original here:
Genetically modified skin grown from stem cells saved a 7 ...

Adult Stem Cells and Gene Therapy Save a Young Boy With …

When people talk about something that saved their skin, they usually mean that it helped them out of a difficult situation. But a young boy in Germany has literally had his skinand his lifesaved through the use of genetically-engineered adult stem cells.

The boy suffered from a condition called junctional epidermolysis bullosa, a severe and often lethal disease in which a mutation leaves the skin cells unable to interconnect and maintain epidermal integrity. The skin blisters and falls off, and the slightest touch or abrasion can leave a patch of skin gone and a painful, difficult-to-heal wound behind. There is no cure for the disease and little other than palliative care available for sufferers of the most severe forms.

Now researchers have combined use of adult stem cells with genetic engineering to successfully treat the young boys life-threatening condition. The boys doctors in Germany called on Dr. Michele De Luca at the University of Modena and Reggio Emilia in Italy to use a technique he has developed to correct the genetic problem and grow new skin.

Over many years, Dr. De Luca has developed a method to grow skin from a patients own epidermal adult stem cells, correct the genetic mutation in the laboratory, and use the genetically-engineered adult stem cells to grow healthy new skin. Dr. De Luca and his team took a tiny patch of skin from the boy, isolated the epidermal stem cells and corrected the genetic problem in stem cell culture. Then they grew sheets of genetically-corrected skin and transplanted them onto the boy.

Reports called the boys recovery stunning, with successful replacement of 80 percent of his skin. Before the procedure, the boys doctors tried several treatments to no avail. One doctor even said, We had a lot of problems in the first days keeping this kid alive. Yet within six months of starting the process, the boy was back in school.

PRO-LIFE COLLEGE STUDENT? LifeNews is looking for interns interested in writing, social media, or video creation. Contact us today.

His skin has remained healthy and completely blister-free. According to the published reports now 21 months after the boys transplant, he loves to show off his new skin and is enjoying school, playing soccer, and being a normal kid. The research has also taught scientists much about the possibilities of using adult stem cells in combination with gene therapy for treatment of diseases.

LifeNews Note: File photo.

Here is the original post:
Adult Stem Cells and Gene Therapy Save a Young Boy With ...

Archives