Page 42«..1020..41424344..5060..»

Archive for the ‘Skin Stem Cells’ Category

Skin cells of schizophrenia patients reveal faulty genetic pathway that began in womb – Genetic Literacy Project

The skin cells of four adults with schizophrenia have provided an unprecedented window into how the disease began while they were still in the womb, according to a recent paper in Schizophrenia Research.

The paper was publishedby researchers at the Jacobs School of Medicine and Biomedical Sciences at the University at Buffalo in collaboration with the Icahn School of Medicine at Mount Sinai. It provides what the authors call the first proof of concept for their hypothesis that a common genomic pathway lies at the root of schizophrenia.

The researchers say the work is a first step toward the design of treatments that could be administered to pregnant mothers at high risk for bearing a child with schizophrenia, potentially preventing the disease before it begins.

The authors gained insight into the early brain pathology of schizophrenia by using skin cells from four adults with schizophrenia and four adults without the disease that were reprogrammed back into induced pluripotent stem cells and then into neuronal progenitor cells.

The next step in the research is to use these induced pluripotent stem cells to further study how the genome becomes dysregulated, allowing the disease to develop.

The GLP aggregated and excerpted this blog/article to reflect the diversity of news, opinion, and analysis. Read full, original post: Faulty genomic pathway linked to schizophrenia developing in utero, study finds

See the original post here:
Skin cells of schizophrenia patients reveal faulty genetic pathway that began in womb - Genetic Literacy Project

Stem Cell Therapy An Option For ENC Patients – Public Radio East

Stem cell therapy is a quickly advancing treatment being used across the country. Now, its becoming more prevalent in eastern North Carolina to those living with chronic pain an alternative to surgery. The minimally invasive procedure is showing results in alleviating back, knee, hip and shoulder pain. Though stem cell therapy is classified by the Food and Drug Administration as experimental, patients say theyre finding relief. Meet New Bern resident and a local endodontist Dr. Donnie Luper. He was skeptical of the procedure at first.

How did you know what those stem cells were going to differentiate into? I mean was I going to grow a foot out of my shoulder or something like that?

Luper tore his rotator cuff 25 years ago during a tubing incident on the Trent River. A subsequent fall during a golf trip in 2015 sent him to a specialist.

I went to see a shoulder surgeon in Richmond. He told me that he didnt think it was a complete tear of my rotator cuff, that I could probably have a minor surgical procedure done and I asked him about stem cell.

After talking with a friend who opted for stem cell treatment for her knee pain, Luper decided to find out more.

My option was if I would have had that shoulder surgery and they had do that bicep tendon repair, I mean I would have been in a sling for six weeks and probably not working for three months.

According to the Food and Drug Administration, stem cells sometimes called the bodys master cells - have the ability to divide and develop into many different cell types. Each new cell has the potential to remain a stem cell or become another type of cell, such as a nerve cell, a skin cell, or a red blood cell. They may also help repair the body by dividing to replenish cells that are damaged by disease, injury or normal wear. Parkinsons disease, spinal cord injuries, damaged organs and cancer could all be possibly treated with the use of stem cells, but more research is needed. Dr. Angelo Tellis is the owner/physician of Aegean Medical, which provides stem cell therapy to patients in Cary, Jacksonville, Morehead City and New Bern.

The adult stem cells we call multipotent stem cells so they can only differentiate into very specific or certain kinds of tissue. Whereas the embryonic stem cells we call pluripotent and can become a variety, almost any tissue. But I only deal with adult stem cells, theyre found to be more useful in clinical applications.

Dr. Tellis says adult stem cells are more responsive to growing tissue in very specific locations. When patients go into Dr. Tellis office for the two hour procedure, he starts by numbing an area of the abdomen and performing liposuction to collect one or two syringes of body fat.

Stem cells can be found in a lot of different tissues throughout the body, but theyre actually in one of the highest concentrations in your own body fat.

The stem cell sample is combined with platelet rich plasma or PRP collected through a blood draw.

That has a lot of the chemical signals and messengers that activate stem cells. So Ill typically combine that with some of the stem cells collected from the body fat and then go under x-ray guidance and put it exactly in the targeted location where we want to create that healing process.

Soreness and stiffness can be expected immediately following the procedure and for about a week after. Dr. Tellis says the results tend to improve with time, taking about three to six months for full recovery. This was Lupers experience in 2016.

Really didnt have to take any pain medications. The joint was really sore over the weekend just because of the injection of the fluid there and after that, I had a small amount of discomfort, but nothing I really had to take medication for.

After three months, Luper says he felt 90 percent better. But he decided to get a second opinion from a shoulder surgeon.

And he told me he thought the stem cells had done a lot but that I still had one little bone spur that was rubbing against the muscle and constantly tearing the little bit of the muscle.

After surgery, Luper says his left shoulder started feeling significantly better in about a month. He was also able to return to one of his favorite pastimes golf. While surgery helped eliminate all of his pain, Luper believes stem cells helped regenerate tissue that was damaged years ago.

He said my rotator cuff muscle didnt even look like it had been torn. I actually tore that, Im sixty now, and I actually tore that when I was 34, 35 tubing on the river and I had to do physical therapy for about three months, but he said he saw absolutely no evidence that Id ever had a rotator cuff tear.

Even though some have found relief and possibly a cure through stem cell therapy, the Food and Drug Administration has not approved any stem cell-based products for use, other than HEMACORD (HE-muh-cord). According to their website, the use of stem cells raises safety concerns such as excessive cell growth, the development of tumors as well as cells migrating from the site of administration and differentiating into inappropriate cell types. And then, theres the cost of the procedure, which is not covered by insurance. The price for the treatment ranges from $2,500 to $5,000. But for those who want to avoid major surgery and the downtime associated with recovery, the risk and cost may be worth it.

If Id have surgery, my deductible would have been that because I have an out-of-pocket max. And I would want to do anything to avoid surgery, especially something that would keep me out of work for three months.

The FDA recommends that consumers interested in stem cell therapy should start a conversation with their doctor about the potential risk to benefit ratio. In addition to Aegean Medical, Advanced Health and Physical Medicine in Greenville and Regenerative Medicine Clinic of Wilmington also provide stem cell therapy in eastern North Carolina.

The rest is here:
Stem Cell Therapy An Option For ENC Patients - Public Radio East

International Stem Cell Corporation Announces Third Patient with Parkinson’s Disease in Phase I Clinical Trial – P&T Community

International Stem Cell Corporation Announces Third Patient with Parkinson's Disease in Phase I Clinical Trial
P&T Community
28, 2017 (GLOBE NEWSWIRE) -- International Stem Cell Corporation (OTCQB:ISCO), a California-based clinical stage biotechnology company developing stem cell-based therapies and biomedical products, today announced the third patient in the clinical trial ...

and more »

Read the original post:
International Stem Cell Corporation Announces Third Patient with Parkinson's Disease in Phase I Clinical Trial - P&T Community

Artificial embryo grown in a dish from two types of stem cells – New Scientist

By Andy Coghlan

Sarah Harrison and Gaelle Recher, Zernicka-Goetz Lab, University of Cambridge

Artificial mouse embryos grown from stem cells in a dish could help unlock secrets of early development and infertility that have until now evaded us.

Magdalena Zernicka-Goetz at the University of Cambridge and her team made the embryos using embryonic stem cells, the type of cells found in embryos that can mature into any type of tissue in the body.

The trick was to grow these alongside trophoblast stem cells, which normally produce the placenta. By growing these two types of cell separately and then combining them in a special gel matrix, the two mixed and started to develop together.

After around four-and-a-half days, the embryos resembled normal mouse embryos that were about to start differentiating into different body tissues and organs.

They are very similar to natural mouse embryos, says Zernicka-Goetz. We put the two types of stem cells together which has never been done before to allow them to speak to each other. We saw that the cells could self-organise themselves without our help.

This is the first time something resembling an embryo has been made from stem cells, without using an egg in some way. Techniques such as cloning, as done for Dolly the sheep and other animals, bypass the need for sperm, but still require an egg cell.

The artificial embryos are providing new insights into how embryos organise themselves and grow, says Zernicka-Goetz. The team engineered the artificial embryos so the cell types fluoresced in different colours, to reveal their movements and behaviour as the embryos go through crucial changes.

Mammal embryos were already known to start as a symmetrical ball, then elongate, form a central cavity and start developing a type of cell layer called mesoderm, which ultimately goes on to form bone and muscle.

We didnt know before how embryos form this cavity, but weve now found the mechanism for it and the sequential steps by which it forms, says Zernicka-Goetz. Its building up the foundations for the whole body plan.

The work is a great addition to the stem cell field and could be extended to human stem cell populations, says Leonard Zon at Boston Childrens Hospital, Massachusetts. Using the system, the factors that participate in embryo development could be better studied and this could help us understand early events of embryogenesis.

But Robin Lovell-Badge at the Francis Crick Institute in London says that the embryos lack two other types of cell layer required to develop the bodies organs: ectoderm, which forms skin and the central nervous system, and endoderm, which makes our internal organs.

Zernicka-Goetz hopes to see these types of cell layers develop in future experiments by adding stem cells that normally form the yolk sac, a third structure involved in embryonic development, to the mix.

If a similar feat can be achieved using human stem cells, this could tell us much about the earliest stages of our development. Current research is limited by the number of excess embryos that are donated from IVF procedures. But the new technique could produce a limitless supply, making it easier to conduct in-depth research. These artificial embryos may also be easier to tinker with, to see what effect different factors have in early embryogenesis.

Disrupting development in this way may provide new insights into the causes of abnormal embryo development and miscarriage. You would be able to understand the principles that govern each stage of development. These are not normally accessible, because they happen inside the mother, says Zernicka-Goetz.

But it is doubtful that this work could ever lead to fully grown babies in the lab. Lovell-Badge says the artificial embryos are unlikely to develop in vitro much further than shown in the study, as they would soon need the supply of nutrients and oxygen that a placenta normally channels from the mother.

Were not planning to make a mouse in the lab using stem cells, says Zernicka-Goetz. But she is hopeful that adding yolk sac stem cells will allow these artificial embryos to survive long enough to study the beginnings of organs like the heart.

Journal reference: Science, DOI: 10.1126/science.aal1810

Read more: Its time to relax the rules on growing human embryos in the lab

More on these topics:

Visit link:
Artificial embryo grown in a dish from two types of stem cells - New Scientist

Wound healing: The stem cell dynamic – Science Daily

Wound healing: The stem cell dynamic
Science Daily
It was particularly exciting to observe that the repair of the skin epidermis involves the activation of very different stem cells that react the same way to the emergency situation of the wound and have the power to completely restore the damaged ...

Follow this link:
Wound healing: The stem cell dynamic - Science Daily

Schizophrenia May Begin In The Womb, Skin Cells Suggest – ReliaWire

The skin cells of four adults with schizophrenia provide a unique insight into how the disease began before they were born.

Scientists call the findings the first proof of concept for the hypothesis that a common genomic pathway lies at the root of schizophrenia. They add the work is a step toward the design of treatments that could be administered to pregnant mothers at high risk for bearing a child with schizophrenia, potentially preventing the disease before it begins.

Michal K. Stachowiak, professor of pathology and anatomical sciences at the University at Buffalo, says:

In the last 10 years, genetic investigations into schizophrenia have been plagued by an ever-increasing number of mutations found in patients with the disease. We show for the first time that there is, indeed, a common, dysregulated gene pathway at work here.

The authors used skin cells from four adults with schizophrenia and four adults without the disease. The cells were reprogrammed back into induced pluripotent stem cells and then into neuronal progenitor cells.

By studying induced pluripotent stem cells developed from different patients, we recreated the process that takes place during early brain development in utero, thus obtaining an unprecedented view of how this disease develops, said Stachowiak. This work gives us an unprecedented insight into those processes.

Stachowiak says the research is a proof of concept for a hypothesis he and colleagues published in 2013 that proposed that a single genomic pathway, called the Integrative Nuclear FGFR 1 Signaling (INFS), is a central intersection point for multiple pathways involving more than 100 genes believed to be involved in schizophrenia.

This research shows that there is a common dysregulated gene program that may be impacting more than 1,000 genes and that the great majority of those genes are targeted by the dysregulated nuclear FGFR1, Stachowiak says.

When even one of the many schizophrenia-linked genes undergoes mutation, by affecting the INFS it throws off the development of the brain as a whole, similar to the way that an entire orchestra can be affected by a musician playing just one wrong note, he says.

The next step in the research is to use these induced pluripotent stem cells to further study how the genome becomes dysregulated, allowing the disease to develop.

We will utilize this strategy to grow cerebral organoidsmini-brains in a senseto determine how this genomic dysregulation affects early brain development and to test potential preventive or corrective treatments.

The work was funded by NYSTEM, the Patrick P. Lee Foundation, the National Science Foundation, and the National Institutes of Health.

Image: Views of a Foetus in the Womb (c. 1510 1512) by Leonardo da Vinci

Visit link:
Schizophrenia May Begin In The Womb, Skin Cells Suggest - ReliaWire

Skin cells suggest schizophrenia may start in the womb – Futurity: Research News

The skin cells of four adults with schizophrenia provide an unprecedented window into how the disease began before they were born.

Scientists call the findings the first proof of concept for the hypothesis that a common genomic pathway lies at the root of schizophreniaand say the work is a step toward the design of treatments that could be administered to pregnant mothers at high risk for bearing a child with schizophrenia, potentially preventing the disease before it begins.

We show for the first time that there is, indeed, a common, dysregulated gene pathway at work here.

In the last 10 years, genetic investigations into schizophrenia have been plagued by an ever-increasing number of mutations found in patients with the disease, says Michal K. Stachowiak, professor of pathology and anatomical sciences at the University at Buffalo. We show for the first time that there is, indeed, a common, dysregulated gene pathway at work here.

The authors gained insight into the early brain pathology of schizophrenia by using skin cells from four adults with schizophrenia and four adults without the disease. The cells were reprogrammed back into induced pluripotent stem cells and then into neuronal progenitor cells.

By studying induced pluripotent stem cells developed from different patients, we recreated the process that takes place during early brain development in utero, thus obtaining an unprecedented view of how this disease develops, said Stachowiak. This work gives us an unprecedented insight into those processes.

Stachowiak says the research, published in Schizophrenia Research, is a proof of concept for a hypothesis he and colleagues published in 2013 that proposed that a single genomic pathway, called the Integrative Nuclear FGFR 1 Signaling (INFS), is a central intersection point for multiple pathways involving more than 100 genes believed to be involved in schizophrenia.

This research shows that there is a common dysregulated gene program that may be impacting more than 1,000 genes and that the great majority of those genes are targeted by the dysregulated nuclear FGFR1, Stachowiak says.

When even one of the many schizophrenia-linked genes undergoes mutation, by affecting the INFS it throws off the development of the brain as a whole, similar to the way that an entire orchestra can be affected by a musician playing just one wrong note, he says.

The next step in the research is to use these induced pluripotent stem cells to further study how the genome becomes dysregulated, allowing the disease to develop.

We will utilize this strategy to grow cerebral organoidsmini-brains in a senseto determine how this genomic dysregulation affects early brain development and to test potential preventive or corrective treatments.

Other researchers from University at Buffalo and the Icahn School of Medicine at Mt. Sinai are coauthors of the work, which was funded by NYSTEM, the Patrick P. Lee Foundation, the National Science Foundation, and the National Institutes of Health.

Source: University at Buffalo

More here:
Skin cells suggest schizophrenia may start in the womb - Futurity: Research News

Durham scientists pioneer innovative stem cell research – Palatinate

By Jacob Dykes

In Durham, a pioneering technology has been developed which is providing a solution to fundamental issues in tissue engineering and stem cell biology. The development of new innovative technology enables the advancement of the research and discovery process and scientific thinking as a whole. For example, its hard to conceive of a biomedical sphere untouched by the blessing of PCR or DNA sequencing. Technological advancements not only offer solutions to existing obstacles, they open up new avenues of research into previously inconceivable areas.

With the current levels of excitement in the research of stem cell biology, you could be forgiven for envisaging a utopian medical scenario where a process akin to science-fiction allows us to generate complex tissues in a Petri-dish, ready for transplantation into the damaged organism. The scientific community has speculated that the nature of stem cells, in their ability to self-renew and produce cell types of any lineage will eventually provide medical solutions to some of our most vilified tissue diseases.

Transitioning speculation to reality requires time, basic research and technology development. A novel product known as Alvetex has been developed by Reinnervate, a Durham University spin-out company, which enables a new routine approach to study stem cells and their ability to form tissues in the laboratory. The product unlocks the potential of stem cell differentiation by mimicking the natural three-dimensional (3D) microenvironment cells encounter in the body, enabling the formation of 3D tissue-like structures.

Cell behaviour, in general, is guided by the complex 3D microenvironment in which they reside. Dispersal of cell-cell interactions and architectural contacts across the surface of the cell are essential for regulating gene expression, the genetic mechanism by which cells change their character and behaviour. Recreation of this microenvironment in the laboratory is essential to studying physiologically relevant behaviour, and the differentiation process by which cells form new cell types. Alvetex is a micro-engineered 3D polystyrene scaffold into which cells can be impregnated for cultivation. Cells grow within a 200-micron thick membrane of the 3D material bathed in culture medium. The microenvironment enables cells to form 3D contacts with neighbouring cells, recreating the more natural interactions found in real tissues. Overall, this affects the structure and function of the cells, enabling them to behave more like their native counterparts, which in turn improves predictive accuracy when working with advanced cell culture models.

We can take progenitor cells from the skin of donors and produce human skin We can take cell lines from the intestine and reproduce the absorptive lining of the intestine. We can take neural progenitors and recapitulate 3D neural networks.

Stefan Przyborski is a Professor of Cell Technology at Durham University and the founder of Reinnervate. He gave us an insight into his technologys applications;

We can take progenitor cells from the skin of donors and produce a full-thickness stratified human skin model (see image). We can take cell lines from the intestine and reproduce the absorptive lining of the intestine. We can take neural progenitors and recapitulate 3D neural networks to simulate aspects of nervous system function. Each of these models can be used to advance basic research, and extend our understanding of tissue development, and simulate aspects of disease.

Such technology is underpinned by well established fundamental principles such as how cellular structure is related to function, which hails way back to Da Vinci himself. It is well known that if you get the structure and the anatomy correct than the physiology will start to follow.

Alvetex technology has already been used in research that has led the publication of over 60 research papers in the field of tissue engineering and cancer biology. One particular group used the technology to successfully test drugs to prevent glioblastoma dispersal, an innovative application in brain oncology. Another has developed a 3D skin model to better study the development of metastatic melanoma, a persistently incurable invasive tumour of the skin. US scientists have used Alvetex on the International Space Station to study the formation of bone tissue in microgravity conditions.

The technology promises to be a cost-effective and ethical solution to current obstacles in cell culturing methods, producing better quality data relevant to man and reducing the need for animal models. Alvetex technology has offered a generational contribution to the process of tissue engineering research, yet the founder has higher ambitions;

What I would like to see in the next few decades is the increased complexity of the tissues that stem cells can be used to generate. If you consider the structure of an organ, the complexity, arrangement and structural organisation of those cell populations, it is far from where we are today. Advances in technology at the interface between disciplines leads to new innovative ideas to solve problems and open up new opportunities.

The development of stem cell research is an incremental process. We have to remain cautious given the potential of stem cell therapy to cause tumour formation, highlighting the need for more stringent models and controls. However, the clinical transplantation of cultured stem cells in bone and cornea repair demonstrates their enormous potential. Laboratory experiments have also demonstrated the potential of stem cells to produce kidney, pancreatic, liver, cardiac and muscle cells. It is hoped that continued research using more physiologically relevant technologies will increase the complexity of these tissues in the lab, and the diversity of their application.

Innovative technological advances play an important role in the process of biomedical science. Scientists at Durham are instrumental in the development of such new technologies that enable the process of new discoveries.

Photograph: Prof Stefan Przyborski, Durham University

See the rest here:
Durham scientists pioneer innovative stem cell research - Palatinate

Thanks to Stem Cell Therapy, Thinning Hair May Be a Thing of the Past – W Magazine

Call me a creature of habit, or just plain boring, but Ive been wearing my hair long, blonde, straight, and side-parted for more than 15 years. The only thing thats really changed is how much of it I have left. Whether the result of bleach, blowouts, stress, hormones, genetics, or all of the above, Ive been shedding like a cheap angora sweater since the age of 30. And, to make matters worse, the hair I do have is fine, fragile, and flyaway.

It wasnt always so. Flipping through old photo albums, I found evidence not only of my natural color (a long-forgotten brown) but also of the graphic, blunt bob I sported in my early 20s. I had oodles of hair back then and would smooth it to my head with pomade and push it behind my earsmuch like Guido Palau did on some of the models in Pradas spring runway show, I noted smugly.

Efforts in the ensuing years to save my ever-sparser strands have been all but futile. You name it, Ive tried it: platelet-rich plasma (PRP), treatments in which your own blood is spun down to platelets and injected into your scalp; mesotherapy (painful vitamin shots, also in the scalp); oral supplements; acupuncture; massage; herbal remedies; and high-tech hair products. Ive even resorted to wearing a silly-looking helmet that bathed my head in low-level laser light and was said to stimulate failing follicles. At this point, I would soak my mane in mares milk under the glow of a waxing supermoon if I thought it would help.

Since hair regeneration is one of the cosmetics-research worlds holiest grails (read: potential multibillion-dollar industry), Ive always hoped that a bona fide breakthrough was around the corner, and prayed it would arrive well ahead of my dotage. As it turns out, it might actually be a five-hour flight from New Yorkand around $10,000away.

It was the celebrity hairstylist Sally Hershberger who whispered the name Roberta F. Shapiro into my ear. You have to call her, she said. She is on to something, and it could be big. Shapiro, a well-respected Manhattan pain-management specialist, treats mostly chronic and acute musculoskeletal and myofascial conditions, like disc disease and degeneration, pinched nerves, meniscal tears, and postLyme disease pain syndromes. Her patient list reads like a whos who of the citys power (and pain-afflicted) elite, and her practice is so busy, she could barely find time to speak with me. According to Shapiro, a possible cure for hair loss was never on her agenda.

But thats exactly what she thinks she may have stumbled upon in the course of her work with stem cell therapy. About eight years ago, she started noticing a commonality among many of her patientsevidence of autoimmune disease with inflammatory components. Frustrated that she was merely palliating their discomfort and not addressing the underlying problems, Shapiro began to look beyond traditional treatments and drug protocols to the potential healing and regenerative benefits of stem cellsspecifically, umbilical cordderived mesenchymal stem cells, which, despite being different from the controversial embryonic stem cells, are used in the U.S. only for research purposes. After extensive vetting, she began bringing patients to the Stem Cell Institute, in Panama City, Panama, which she considers the most sophisticated, safe, and aboveboard facility of its kind. Its not a spa, or a feel-good, instant-fix kind of place, nor is it one of those bogus medical-tourism spots, she says. Lori Kanter Tritsch, a 55-year-old New York architect (and the longtime partner of Este Lauder Executive Chairman William Lauder) is a believer. She accompanied Shapiro to Panama for relief from what had become debilitating neck pain caused by disc bulges and stenosis from arthritis, and agreed to participate in this story only because she believes in the importance of a wider conversation about stem cells. If it works for hair rejuvenation, or other cosmetic purposes, great, but that was not at all my primary goal in having the treatment, Kanter Tritsch said.

While at the Stem Cell Institute, Kanter Tritsch had around 100 million stem cells administered intravenously (a five-minute process) and six intramuscular injections of umbilical cord stem cellderived growth factor (not to be confused with growth hormone, which has been linked to cancer). In the next three months, she experienced increased mobility in her neck, was able to walk better, and could sleep through the night. She also lost a substantial amount of weight (possibly due to the anti-inflammatory effect of the stem cells), and her skin looked great. Not to mention, her previously thinning hair nearly doubled in volume.

As Shapiro explains it, the process of hair loss is twofold. The first factor is decreased blood supply to hair follicles, or ischemia, which causes a slow decrease in their function. This can come from aging, genetics, or autoimmune disease. The second is inflammation. One of the reasons I think mesenchymal stem cells are working to regenerate hair is that stem cell infiltration causes angiogenesis, which is a fancy name for regrowing blood vessels, or in this case, revascularizing the hair follicles, Shapiro notes. Beyond that, she says, the cells have a very strong anti-inflammatory effect.

For clinical studies shes conducting in Panama, Shapiro will employ her proprietary technique of microfracturing, or injecting the stem cells directly into the scalp. She thinks this unique delivery method will set her procedure apart. But, she cautions, this is a growing science, and we are only at the very beginning. PRP is like bathwater compared with amniotic- or placenta-derived growth factor, or better yet, umbilical cordderived stem cells.

Realizing that not everyone has the money or inclination to fly to Panama for a treatment that might not live up to their expectations, Hershberger and Shapiro are in the process of developing Platinum Clinical, a line of hair products containing growth factor harvested from amniotic fluid and placenta. (Shapiro stresses that these are donated remnants of a live birth that would otherwise be discarded.) The products will be available later this year at Hershbergers salons.

With follicular salvation potentially within reach, I wondered if it might be time to revisit the blunt bob of my youth. I call Palau, and inquire about that sleek 1920s do he created for Prada. Fine hair can actually work better for a style like this, he says. In fact, designers often prefer models with fine hair, so the hairstyle doesnt overpower the clothing. Then he confides, Sometimes, if a girl has too much hair, we secretly braid it away. Say what? I know, its the exact opposite of what women want in the real world. But models are starting to realize that fine hair can be an asset. Look, at some point you have to embrace what you have and work with it. Wise words, perhaps, and proof that, like pretty much everything else, thick hair is wasted on the young.

From the Minimalist to the Bold, the 5 Best Hair Trends of New York Fashion Week

Watch W's most popular videos here:

View original post here:
Thanks to Stem Cell Therapy, Thinning Hair May Be a Thing of the Past - W Magazine

Girl, eight, with rare brain disorder in pioneering UCL stem cell research – Evening Standard

A girl of eight whose rare brain disorder is likely to lead to her death when she is in her teens is taking part in pioneering stem cell research in a bid to save others with same condition.

Lily Harrisss skin cells will first be turned into stem cells and then into brain cells by researchers at University College London as they seek treatments or a cure.

About 100 to 200 cases of BPAN beta-propeller protein-associated neurodegeneration are known worldwide, although this is believed to be an underestimate.

Children often suffer delayed development, sleep problems, epilepsy and lack of speech and their symptoms can be mistaken for other conditions.

Lily, from Luton, was diagnosed when she was five. She has very limited communication skills and uses a wheelchair. She wakes four or five times a night and needs drugs to control seizures.

However, she loves swimming and her father Simon said she has recently began singing on car journeys.

Shes laughed and giggled her way through everything, and shes been through a lot, he said.

Shes a beautiful little girl who can be quite naughty sometimes. Were giving her the best time we can while shes here. We have a beautiful little girl and its just so cruel.

Young people with BPAN develop abnormal muscle tone, symptoms of Parkinsons disease and dementia.

Mr Harriss and his wife Samantha, who work for an airline, know that as Lilys condition progresses she may have difficulty swallowing and require pain management.

Mr Harriss said: Lily can point to things she wants, she uses a little sign language and she can say a few words, like mummy, daddy, hello and goodbye.

Medical research like this for children is just absolutely vital.

We know we wont get a cure for Lily but, as parents, we need to be bigger than that. Other children might benefit through Lily. We are so proud of her.

The UCL study is being funded by 230,000 from childrens charity Action Medical Research and the British Paediatric Neurology Association. Lead researcher Dr Apostolos Papandreou hopes his research will lead to trials of treatments.

He said: The parents Ive met understandably feel devastated at the prospect of their children having a progressive disorder. However, theyre really keen to explore new avenues and participate in research projects.

See the original post:
Girl, eight, with rare brain disorder in pioneering UCL stem cell research - Evening Standard

Amazing medicine – The News International

If we cut off the tail of a lizard, it grows back. If we cut off the hand of a human being, it does not grow back. Why not? This question has perplexed scientists for a long time. Recently scientists at the Translational Genomics Research Institute (TGen) and Arizona State University (ASU) in the US identified three tiny RNA switches (known as microRNAs) which turn genes on and off and are responsible for the regeneration of tails in the green lizard. Now researchers are hoping that using the next generation genomic DNA and computer analysis will lead to discoveries of new therapeutic approaches to switch on similar regenerative genes in human beings.

Micro RNAs are able to control many genes at the same time. They have been compared to an orchestra conductor controlling and directing many musicians. Hundreds of genes (musicians playing the orchestra of life), controlled by a few micro RNA switches, have been identified that are responsible in the regenerative process. This may well mark the beginning of a new era in which it may be possible to regenerate cartilage in knees, repair spinal cords and amputated limbs.

Tissue regeneration has become an attractive field of science, triggered by exciting advances in stem cell technologies. Stem cells are undifferentiated biological cells that are then converted into various types of cells such as heart, kidney or skin through a process known as differentiation. They can divide into more stem cells and provide a very effective mechanism for repair of damaged tissues in the body. The developing embryo contains stem cells which are then transformed into specialised cells as the embryo develops. They can be obtained by extraction from the bone marrow, adipose tissue or blood, particularly the blood from the umblical cord after birth.

Stem cells are now finding use in a growing number of therapies. For instance leukaemia is a cancer of the white blood cells. To treat leukaemia, one approach is to get rid of the diseased white blood cells and replace them with healthy cells. This may be done by a bone marrow transplant through which the patients bone marrow stem cells are replaced with those from a healthy, matching donor. If the transplant is successful, the stem cells migrate into the patients bone marrow resulting in the production of new, healthy white blood cells that replace the abnormal cells. Stem cells can now be artificially grown and then transformed (differentiated) into the heart, kidney, nerve or other typed of cells.

The field of regenerative medicine is developing at a fast pace. It involves the replacement, engineering or regeneration of human tissues and organs so that their normal function can be restored. Tissues and organs can also be grown in the laboratory if the body cannot heal itself. If the cells of the organ being grown are derived from the patients own cells, the possibility of rejection of the transplanted organ is minimised. Stem cells may also be used to regenerate organs.

Each year about 130,000 organs, mostly kidneys, are transplanted from one human being to another. The process of growing organs artificially has been greatly accelerated by the advent of 3D bioprinting. This involves the use of 3D printing technologies through which a human organ, liver or kidney, is produced by printing it with cells, layer-by-layer. This became possible when it was discovered that human cells can be sprayed through the nozzles of an inkjet printer without destroying or damaging them. Tissues and organs can thus be produced and transplanted into humans. Joints, jaw bones and ligaments can also be produced in this manner.

Initially, the work was confined to animals when ears, bones and muscle tissues were produced by bioprinting and then successfully transplanted into animals. Even prosthetic ovaries of mice were produced and transplanted so that the recipient mice could conceive and give birth later. While gonads have not been produced by bioprinting in humans, blood vessels have already been produced by the printing process and successfully transplanted into monkeys. Considerable work is also going on in the production of human knee cartilage pads through the bioprinting process. Wear and tear of the cartilage results in difficulties in walking, particular in older age groups, and often requires knee replacement through surgeries. The development of technologies to replace the damaged cartilages with new cartilages made by bioprinting could prove to be invaluable.

Another area of active research in this field is the production of human skin by bioprinting which may be used for treating burns and ulcers. Technologies have been developed to spray stem cells derived from the patient directly on the areas of the body where the skin is needed. In this way, stem cells help skin cells regrow under suitable conditions. Similar progress is being made in generating liver, kidney and heart tissues so that the long waiting time for donors can be circumvented.

When will we be able to print entire human organs? It has been estimated that complete human kidneys and livers should become commercially available through the bioprinting process within five to seven years. Hearts will probably take longer because of their more complex internal structure. However, one thing is clear: a huge revolution is now taking place in the field of regenerative medicine, triggered by spectacular advances in stem cell research. This presents a wonderful opportunity for learning and developing expertise in this field for us in our country.

In Pakistan a number of important steps have been taken in this fast evolving field. One of them is the establishment of a first rate facility for stem cell research in the Dr Panjwani Centre for Molecular Medicine and Drug Research (PCMD) in the University of Karachi. This institution has already earned an international reputation because of its outstanding publications in this field.

A second important development is that plans to set up an Institute for Translational Regenerative Medicine at PCMD so that Pakistan remains at the cutting edge in this fast emerging field are now under way.

Such initiatives can however only contribute to the process of socio-economic development if they operate under an ecosystem that is designed to promote the establishment of a strong knowledge economy.

Pakistan spends only about 0.3 percent of its GDP on science and about two percent of its GDP on education, bringing the nations ranking to the lowest 10 countries in the world. This is largely due to the stranglehold of the feudal system over our democracy. It is only by tapping into our real wealth our children that Pakistan can emerge from the quagmire of illiteracy and poverty and stand with dignity in the comity of nations.

The writer is chairman of UN ESCAP Committee on Science Technology & Innovation and former chairman of the HEC. Email: [emailprotected]

Continue reading here:
Amazing medicine - The News International

Jonathan Pitre battles blood, lung infections before second stem cell transplant – Ottawa Sun


Ottawa Sun
Jonathan Pitre battles blood, lung infections before second stem cell transplant
Ottawa Sun
People with RDEB have a fault in the gene responsible for a specific kind of collagen that connects the outer layer of skin, the epidermis, with those below it. The clinical trial seeks a biochemical correction to that fault. If the transplant works ...

and more »

See more here:
Jonathan Pitre battles blood, lung infections before second stem cell transplant - Ottawa Sun

What are stem cells? – Craig A. Kohn | TED-Ed

Stem cells are a rapidly advancing field of biological research. Since Dr. James Thomson first cultivated human embryonic stem cells at the University of Wisconsin - Madison in the late 1990s, this field of researched has exploded with potential. The links below provide access to a curriculum developed under the supervision of Dr. Thomson as well as the co-directors and staff of the UW Stem Cell & Regenerative Medicine Center. The material has been reviewed for accuracy by the scientists actually conducting the research and was compiled and formatted by Craig Kohn, a high school teacher with research experience, for a high school audience. The PowerPoint presentation works in conjunction with the notesheet, allowing for students to work independently if preferred. More information about specific instructional practices can be found below in Teacher Notes. PowerPoint: http://bit.ly/ted-stemcells Notesheet: http://bit.ly/ted-stemcellsnotesheet Quiz: http://bit.ly/ted-stemcellsquiz Additional resources about stem cells can be found at: http://www.stemcells.wisc.edu/node/386 http://stemcells.nih.gov/Pages/Default.aspxhttp://www.stemcellschool.org/http://www.nursingdegree.net/blog/750/25-best-blogs-for-following-stem-cell-research/

See original here:
What are stem cells? - Craig A. Kohn | TED-Ed

Stem Cells and Aging | Life Code

Adult stem cell function declines with age leading to the decline in fitness

The potential therapeutic use of stem cells is a very hot topic these days. Most of the attention has focused on embryonic stem cells and induced Pluripotent Stem cells (iPS cells), which can form every tissue type in the body to regenerate failing organs. The problem is that detailed knowledge is lacking for how to stimulate the embryonic stem cells to form differentiated tissues (e.g. cells that form the heart, pancreas, muscle, and brain). Moreover, because embryonic stem cells are unlimited in their ability to form any type of tissue, the risk of cancer looms large over the therapeutic use of embryonic stem cells. For example, both embryonic and IPS stem cells can form tumors called teratomas when injected into immune-compromised mice. Enter the bodys adult stem cells, which have not generally been associated with cancer and have been used safely as therapeutics in many countries. The problem with adult stem cells is that it is difficult to get enough of them to be effective for most indications or target the harvested adult stem cells to the proper tissue. Moreover, there are scores of different types of adult stem cells in the body, so picking the best type of adult stem cell for a particular therapeutic can be challenging. Thus, adult stem cell therapeutics with all its potential to regenerate damaged organs and tissues is still a work in progress.

But what about the many populations of endogenous adult stem cells that everyone has embedded in every organ system of the body? All the organs and differing tissues of the body appear to have adult stem cells available for regenerating cells in case of injury or disease. It was recently discovered that even brain neurons and heart muscle cells (previously thought to be non-dividing and irreplaceable in adults) have their own reservoirs of adult stem cells for regeneration. Unfortunately, as we age most adult stem cell populations either decline in number and/or lose the ability to differentiate into functional tissue-specific cells. For example, cardiac muscle stem cells exist but old folks have only one half the number of cardiac stem cells found in young people. Thus, adult stem cells become more and more dysfunction with age, which progressively increases organ and tissue dysfunction with age.

There are many examples revealing the role of adult stem cells in aging. First, the outer surface of your skin continuously sloughs off dead cells, so that adult stem cells must continuously replenish the dying skin cells to maintain the skin as an effective protective barrier to the outside world. With age, there are progressively fewer functional skin stem cells, so cell turnover in the skin slows, leading to thinner, dryer skin that loses its elasticity and youthful beauty. Second, hair also thins and goes grey, as functional follicle stem cell decline and the adult stem cells generating hair color also decline. Third, the differing adult stem cells that maintain the tissues composing skeletal muscle, pancreas, heart, bone, liver, kidney, and the immune system lose functional capacity, raising the potential for decline in tissue function or outright failure with age. As a final example, the five senses of sight, hearing, smell, taste, and touch slowly wane with age, as the declining stem cell populations responsible for maintaining these functions are unable to fully replenish the sensory neurons after injury and random cell death.

If your own adult stem cells are a key factor in aging and disease, then one novel way to slow aging and disease is to stimulate your own adult stem cells to maintain their proper numbers and functional capacity to differentiate into the various tissues as needed for repair and regeneration. This makes sense, because in most, if not all, organs of the body, old cells are continually being replaced by new cells coming from the adult stem cell populations. If stem cells are not producing enough new cells, then organs slowly decline in function as you age. Thus, stimulating your own stem cells can be a winning strategy to stave off many of the disorders associated with aging.

In practice, however, stimulating adult stem cell populations in the body is not a simple task. If the proliferation of adult stem cells is over stimulated, then one may get overgrowth of tissues or a potential tumor. Alternatively, one may stimulate the stem cells to proliferate in a balanced and regulated way, but the stem cells lose functionality and cannot differentiate into the desired specialized tissues to replace senescent cells. These twin problems promoting over stimulation or dysfunctional stem cells put real limits on any proposed therapeutic for stimulating stem cells. For example, most current treatments to stimulate immunity or stem cells (nave T cells) rely on complex carbohydrates from mushrooms or microorganisms to provide antigenic material that can stimulate immunity. This will activate the immune system stem cells to make more differentiated non-stem memory T cells directed against the antigenic material, but it does nothing to stimulate more immune stem cells (nave T cells). Indeed, chronic use of such stem cell enhancers may actually lead to stem cell depletion, as more adult stem cells are exhausted from the requirement to respond to the constant presence of the polysaccharide antigen. Indeed, one theory of how the HIV virus causes a defective immune system is that it exhausts the supply of nave T cells by the repeated attacks of the mutating HIV virus.

Stem Cell 100TM is a nutraceutical supplement that improves the function of your existing stem cells rather than over stimulate stem cells to differentiate or divide. By promoting the stability and vitality of adult stem cells they have the capacity to divide when the body signals a need for more stem cells and differentiated cells. When an organ or tissue is damaged, it will send out natural signals that new cells are needed to replace old or damaged cells. Stem Cell 100TM allows the adult stem cells to respond to the damage signal by provided new differentiated cells to replace the old damaged cells and also make more adult stem cells to keep up the stem cell population. Two other compounds in Stem Cell 100TM provide further natural support for stem cells.

(Note that not everyone will experience the same effects, as conditions vary among individuals. The general expectation is that for most health measurements that are in the Normal Range for your age, Stem Cell 100TM will promote readings that you had when some 20 years younger.)

The statements above have not been reviewed by the FDA. Stem Cell 100TM is not meant as a preventive or treatment for any disease.

The rest is here:
Stem Cells and Aging | Life Code

Stem cells collected from fat may have use in anti-aging treatments – Science Daily

Stem cells collected from fat may have use in anti-aging treatments
Science Daily
Adult stem cells collected directly from human fat are more stable than other cells -- such as fibroblasts from the skin -- and have the potential for use in anti-aging treatments, according to researchers from the Perelman School of Medicine at the ...

and more »

Original post:
Stem cells collected from fat may have use in anti-aging treatments - Science Daily

Nanofiber matrix sends stem cells sprawling in all directions – Gizmag – New Atlas

Human stem cells grown on Kyoto University's "fiber-on-fiber" culturing system(Credit: Kyoto University)

Mighty promising as they are, stem cells certainly aren't easy to come by. Recent scientific advances have however given their production a much-needed boost, with a Nobel-prize winning technology that turns skin cells into embryonic-like stem cells and another that promises salamander-like regenerative abilities being just a couple of examples. The latest breakthrough in the area comes from Japanese researchers who have developed a nanofiber matrix for culturing human stem cells, that they claim improves on current techniques.

The work focuses on human pluripotent stem cells (hPSCs), which have the ability to mature into any type of adult cell, be they those of the eyes, lungs or hair follicles. But that's assuming they can be taken up successfully by the host. Working to improve the odds on this front, scientists have been exploring ways of culturing pluripotent stem cells in a way that mimics the physiological conditions of the human body, allowing them to grow in three dimensions rather than in two dimensions, as they would in a petrie dish.

Among this group is a team from Japan's Kyoto University, which has developed a 3D culturing system it says outperforms the current technologies that can only produce low quantities of low-quality stem cells. The system consists of gelatin nanofibers on a synthetic mesh made from biodegradable polyglycolic acid, resulting in what the researchers describe as a "fiber-on-fiber" (FF) matrix.

The team found that seeding human embryonic stem cells onto this type of matrix saw them adhere well, and enabled an easy exchange of growth factors and supplements. This led to what the researchers describe as robust growth, with more than 95 percent of the cells growing and forming colonies after just four days of culture.

And by designing a special gas-permeable cell culture bag, the team also demonstrated how they could scale up the approach. This is because several of the cell-loaded matrices can be folded up and placed inside the bag, with testing showing that this approach yielded larger again numbers of cells. What's more, the FF matrix could even prove useful in culturing other cell types.

"Our method offers an efficient way to expand hPSCs of high quality within a shorter term," the team writes in its research paper. "Additionally, as nanofiber matrices are advantageous for culturing other adherent cells, including hPSC-derived differentiated cells, FF matrix might be applicable to the large-scale production of differentiated functional cells for various applications."

The research was published in the journal Biomaterials.

Here is the original post:
Nanofiber matrix sends stem cells sprawling in all directions - Gizmag - New Atlas

Scientists discover an unexpected influence on dividing stem cells’ fate – ScienceBlog.com (blog)

When most cells divide, they simply make more of themselves. But stem cells, which are responsible for repairing or makingnew tissue, have a choice: They can generate more stem cells or differentiate into skin cells, liver cells, or virtually any of the bodys specialized cell types.

As reported February 3 in Science, scientists at The Rockefeller University have discovered that this pivotal decision can hinge on whether or not tiny organ-like structures, organelles, are divvied up properly within the dividing stem cell.

In order for the bodys tissues to develop properly and maintain themselves, renewal and differentiation must be carefully balanced, says senior author Elaine Fuchs, the Rebecca C. Lancefield Professor and head of the Robin Chemers Neustein Laboratory of Mammalian Cell Biology and Development. Our experiments suggest an unexpected role for the positioning and inheritance of cellular organelles, in this case enzyme-filled peroxisomes, in controlling this intricate balance.

An uneven division

The outer section of the skin, the epidermis, provides a protective barrier for the body, and stem cells reside deep within it. During development, these cells divide so that one renewing stem cell daughter remains inward while the other daughter differentiates and moves outward to become part of the epidermis outer layers. First author Amma Asare, a graduate student in the lab, wanted to know how skin cells first emerge and begin this transition.

Looking in developing mouse skin, Asare devised an approach to identify genes that help guide the balance between new cells that either stay stem-like or differentiate. One particular protein, Pex11b, caught her attention. It is associated with the membrane that surrounds the peroxisome, an organelle that helps to free energy from food.

Asare showed that the protein seems to work by making sure the organelles are in the right locations so they can be divided between the daughter cells. In cells that lacked Pex11b, peroxisomes werent divvied up evenlyin some cases, one daughter cell ended up with all of the peroxisomes and the other didnt get any at all. And for those cells whose peroxisome distribution was disrupted, cell division took longer, and the mitotic spindle, the structure that separates the daughters genetic material, didnt align correctly.

The net result of depleting skin stem cells of Pex11b, Asare found, was that fewer daughter cells were able to differentiate into mature skin cells.

A delay changes fate

The researchers next moved peroxisomes around in the cell using a sophisticated laboratory technique, and the effect was the same. If the peroxisomes are in the wrong positions during cell division, no matter how they get there, that slows down the process, Asare says.

The effect for the whole organism was dramatic: If peroxisome positioning was disrupted in the stem cells, the mice embryos could no longer form normal skin.

While some evidence already suggested the distribution of organelles, including energy-producing mitochondria, can influence the outcome of cell division, we have shown for the first time that this phenomenon is essential to the proper behavior of stem cells and formation of the tissue, says Fuchs, who is also a Howard Hughes Medical Institute Investigator.

Original post:
Scientists discover an unexpected influence on dividing stem cells' fate - ScienceBlog.com (blog)

Study implicates neural stem cell defects in smooth brain syndrome – Medical Xpress

February 13, 2017 by Nicholas Weiler

Research led by scientists at UC San Francisco and Case Western Reserve University School of Medicine has used brain "organoids"tiny 3-D models of human organs that scientists grow in a dish to study diseaseto identify root causes of Miller-Dieker Syndrome (MDS), a rare genetic disorder that causes fatal brain malformations.

MDS is caused by a deletion of a section of human chromosome 17 containing genes important for neural development. The result is a brain whose outer layer, the neocortex, which is normally folded and furrowed to fit more brain into a limited skull, instead has a smooth appearance (lissencephaly) and is often smaller than normal (microcephaly). The disease is accompanied by severe seizures and intellectual disabilities, and few infants born with MDS survive beyond childhood.

In the new studypublished online January 19, 2017 in Cell Stem Cellthe research team transformed skin cells from MDS patients and normal adults into induced pluripotent stem cells (IPSCs) and then into neural stem cells, which they placed in a 3 dimensional culture system to grow organoid models of the human neocortex with and without the genetic defect that causes MDS.

Closely observing the development of these MDS organoids over time revealed that many neural stem cells die off at early stages of development, and others exhibit defects in cell movement and cell division. These findings could help explain how the genetics of MDS leads to lissencephaly, the authors say, while also offering valuable insights into normal brain development.

"The development of cortical organoid models is a breakthrough in researchers' ability to study how human brain development can go awry, especially diseases such as MDS," said Tony Wynshaw-Boris, MD, PhD, chair of the Department of Genetics and Genome Studies at Case Western Reserve University School of Medicine, and co-senior author of the new study. "This has allowed us to identify novel cellular factors that contribute to Miller-Dieker syndrome, which has not been modeled before."

'Smooth Brain' Organoids Reveal Defects

Previous research on the causes of lissencephaly has relied on mouse models of the disease, which suggested that the main driver of the disorder was a defect in the ability of young neurons to migrate to the correct location in the brain. But Arnold Kriegstein, MD, PhD, professor of neurology, director of the Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research at UCSF, and co-senior author, says there are significant drawbacks to this approach.

"Unlike the human brain, the mouse brain is naturally smooth," Kriegstein said. "If you are studying a disease that leads to a smooth brain in humans, it's a challenge to study it in an animal that normally has a smooth brain."

The mouse brain also lacks a type of neural stem cell called outer radial glia, which were discovered by Kriegstein's group in 2010. These cells are thought to have played a crucial role in the massive expansion in size and complexity of the primate brain relative to other mammals over the course of evolution.

In order to more accurately model the progression of MDS in the embryonic human brain, study first author Marina Bershteyn, PhD, a postdoctoral researcher in the Wynshaw-Boris and Kriegstein labs, spearheaded the development of MDS cortical organoids and techniques to observe how stem cells within these organoids developed in the laboratory into the different cell types seen in first-trimester embryonic human brains.

Bershteyn and her team found using time-lapse imaging that outer radial glia cells that grew in patient-derived organoids had a defect in their ability to dividepotentially contributing to the small, smooth brains seen in MDS patients.

"There are just fundamental differences in how mouse and human brains grow and develop," said Bershteyn, who is now a scientist at Neurona Therapeutics, a company founded by Kriegstein and colleagues to develop stem cell therapies for neurological diseases. "Part of the explanation for why these observations were not made before is that outer radial glia cells are quite rare in mouse."

In addition, the team found that early neural stem cells called neuroepithelial cells which are present in both mice and humans die at surprisingly high rates in MDS organoids, and when they do survive, divide in an abnormal wayas if they are prematurely transforming into neurons, cutting short important early stages of brain development.

Consistent with prior mouse studies, time-lapse imaging also revealed that newborn neurons are unable to migrate properly through developing brain tissue, which potentially contributes to the failure of MDS brains to properly form outer brain structures.

Organoid Research Opens Doors to Studying Human Brain Diseases in Lab

Together, these observations helped the team pinpoint developmental stages and specific neural cell types that are impaired in MDS. The next step to understanding lissencephaly more broadly, the authors say, will be to test cells from patients with different genetic forms of the disease, so researchers can begin to link specific mutations with the cellular defects that drive brain malformation.

The study is also a demonstration of the utility of patient-derived brain organoids as a way to bridge the gap between animal models and human disease, the authors say. In particular, the finding that human outer radial glia cells readily grow in organoid models opens the door for scientists worldwide to study the role of these cells in both normal human brain development and disease.

"Patient-derived cortical organoids are creating a huge amount of excitement," Kriegstein said. "We are now able to study human brain development experimentally in the lab in ways that were not possible before."

Explore further: Scientists engineer gene pathway to grow brain organoids with surface folding

More information: Marina Bershteyn et al. Human iPSC-Derived Cerebral Organoids Model Cellular Features of Lissencephaly and Reveal Prolonged Mitosis of Outer Radial Glia, Cell Stem Cell (2017). DOI: 10.1016/j.stem.2016.12.007

One of the most significant ways in which the human brain is unique is the size and structure of the cerebral cortex. But what drives the growth of the human cortex, likely the foundation for our unique intellectual abilities?

Zika's hypothesized attraction to human neural stem cells may come from its ability to hijack a protein found on the surface of these cells, using it as an entryway to infection. In Cell Stem Cell on March 30, researchers ...

When you build models, whether ships or cars, you want them to be as much like the real deal as possible. This quality is even more crucial for building model organs, because disease treatments developed from these models ...

The human cerebral cortex contains 16 billion neurons, wired together into arcane, layered circuits responsible for everything from our ability to walk and talk to our sense of nostalgia and drive to dream of the future. ...

Brazilian researchers from the D'Or Institute for Research and Education (IDOR) and Federal University of Rio de Janeiro (UFRJ) have demonstrated the harmful effects of ZIKA virus (ZIKV) in human neural stem cells, neurospheres ...

The acid test for a vaccine is: "Does it protect people from infection?" Emory Vaccine Center researchers have analyzed this issue for a leading malaria vaccine called RTS,S, and their results have identified candidate signatures, ...

Patients with inflammatory bowel disease are more likely to see dramatic shifts in the make-up of the community of microbes in their gut than healthy people, according to the results of a study published online Feb. 13 in ...

Walter and Eliza Hall Institute researchers have used advanced cellular, bioinformatics and imaging technology to reveal a long-lived type of stem cell in the breast that is responsible for the growth of the mammary glands ...

Research led by scientists at UC San Francisco and Case Western Reserve University School of Medicine has used brain "organoids"tiny 3-D models of human organs that scientists grow in a dish to study diseaseto identify ...

People with hemophilia require regular infusions of clotting factor to prevent them from experiencing uncontrolled bleeding. But a significant fraction develop antibodies against the clotting factor, essentially experiencing ...

Patients with Crohn's disease, a type of inflammatory bowel disease (IBD) that causes abdominal pain and diarrhea, can also experience joint pain. In Crohn's disease, which affects about 800,000 Americans, the immune system ...

Please sign in to add a comment. Registration is free, and takes less than a minute. Read more

Read more:
Study implicates neural stem cell defects in smooth brain syndrome - Medical Xpress

Can banking baby teeth treat diabetes? – Fox News

When she was just 11 months old, Billie Sue Wozniaks daughter Juno was diagnosed with type 1 diabetes, an autoimmune disease that affects 1.25 million people and approximately 200,000 children under age 20 in the United States.

The disease had affected several members of Billie Sues family, including her uncle, who passed away at the age of 30.

My first thought was, Her life is going to be short, the 38-year-old from Reno, Nevada recalled. The more that I learned, the more I found that many people with type 1 live longer and the treatment advances are really exciting.

While looking for treatments, Wozniak learned about encapsulation therapy, in which an encapsulated device containing insulin-producing islet cells derived from stem cells is implanted under the skin. The encapsulation device is designed to protect the cells from an autoimmune attack and may help people produce their own insulin.

After learning of the therapy through JDRF, Wozniak saw an ad on Facebook for Store-A-Tooth, a company that offers dental stem cell banking. She decided to move forward with the stem cell banking, just in case the encapsulation device became an option for Juno.

In March 2016, a dentist extracted four of Junos teeth, and sent them to a lab so her stem cells could be cryopreserved. Wozniak plans to bank the stem cells from Junos molars as well.

Its a riskI dont know for sure if it will work out, Wozniak said.

Dental stem cells: a future of possibilities

For years, stem cells from umbilical cord blood and bone marrow have been used to treat blood and bone marrow diseases, blood cancers and metabolic and immune disorders.

Although there is the potential for dental stem cells to be used in the same way, researchers are only beginning to delve into the possibilities.

Dental stem cells are not science fiction, said Dr. Jade Miller, president of the American Academy of Pediatric Dentistry. I think at some point in time, were going to see dental stem cells used by dentistson a daily practice.

Dental stem cells have the potential to produce dental tissue, bone, cartilage and muscle. They may be used to repair cavities, fix a tooth damaged from periodontal disease or bone loss, or even grow a tooth instead of using dental implants.

In fact, stem cells can be used to repair cracks in teeth and cavities, according to a recent mouse study published in the journal Scientific Reports.

Theres also some evidence that dental stem cells can produce nerve tissue, which might eliminate the need for root canals. A recent study out of Tufts University found that a collagen-based biomaterial used to deliver stem cells to the inside of damaged teeth can regenerate dental pulp-like tissues.

Dental stem cells may even be able to treat neurological disorders, spinal cord and traumatic brain injuries.

I believe those are the kinds of applications that will be the first uses of these cells, said Dr. Peter Verlander, Chief Scientific Officer for Store-A-Tooth.

When it comes to treating diseases like type 1 diabetes, dental stem cells also show promise. In fact, a study in the Journal of Dental Research found that dental stem cells were able to form islet-like aggregates that produce insulin.

Unlike umbilical cord blood where theres one chance to collect stem cells, dental stem cells can be collected from several teeth. Also, gathering stem cells from bone marrow requires invasive surgery and risk, and it can be painful and costly.

The stem cells found in baby teeth, known as mesenchymal cells, are similar to those found in other parts of the body, but not identical.

There are differences in these cells, depending on where they come from, Verlander said.

Whats more, mesenchymal stem cells themselves differ from hematopoietic, or blood-forming stem cells. Unlike hematopoietic stem cells, mesenchymal stem cells can expand.

From one tooth, we expect to generate hundreds of billions of cells, Verlander said.

Yet the use of dental stem cells is not without risks. For example, theres evidence that tumors can develop when stem cells are transplanted. Theres also a chance of an immune rejection, but this is less likely if a person uses his own stem cells, Miller said.

The process for banking stem cells from baby teeth is relatively simple. A dentist extracts the childs teeth when one-third of the root remains and the stem cells are still viable. Once the teeth are shipped and received, the cells are extracted, grown and cryopreserved.

Store-A-Tooths fees include a one-time payment of $1,749 and $120 per year for storage, in addition to the dentists fees for extraction.

For families who are interested in banking dental stem cells, they should know that theyre not necessarily a replacement for cord blood banking or bone marrow stem cells.

Theyre not interchangeable, we think of them as complementary, Verlander said.

Although the future is unclear for Junowho was born in 2008her mom is optimistic that shell be able to use the stem cells for herself and if not, someone else.

Ultimately, however, Wozniak hopes that if dental stem cells arent the answer, there will be a biological cure for type 1 diabetes.

I hold out hope that somewhere, someone is going to crack the code, she said.

Julie Revelant is a health journalist and a consultant who provides content marketing and copywriting services for the healthcare industry. She's also a mom of two. Learn more about Julie at revelantwriting.com.

Here is the original post:
Can banking baby teeth treat diabetes? - Fox News

Scientists discover an unexpected influence on dividing stem cells … – Science Daily


Science Daily
Scientists discover an unexpected influence on dividing stem cells ...
Science Daily
When it divides, a stem cell has a choice: produce more stem cells or turn into the specific types of cells that compose skin, muscle, brain, or other tissue.

and more »

See the original post here:
Scientists discover an unexpected influence on dividing stem cells ... - Science Daily

Induced pluripotent stem cells don’t increase genetic mutations – Science Daily

Induced pluripotent stem cells don't increase genetic mutations
Science Daily
Using skin cells from the same donor, they created genetically identical copies of the cells using both the iPSC and the subcloning techniques. They then sequenced the DNA of the skin cells as well as the iPSCs and the subcloned cells and determined ...

See the rest here:
Induced pluripotent stem cells don't increase genetic mutations - Science Daily

Celebrate Your Skin with Champagne & Phyto-StemCells – PR Newswire (press release)

PARIS and STOCKHOLM, Feb. 9, 2017 /PRNewswire/ -- Laboratoire Fleur de Sants new Champagne Collection uses Extrait de Champagne, fueled by grape seed Phyto-StemCell's Resveratrol, for the ultimate antioxidant protection and photo-aging prevention. By reinforcing the skin's structural matrix (collagen and elastin) and stimulating its natural regeneration process, this powerful antioxidant postpones skin aging and leaves it smooth and even toned. One more reason to love Champagne!

"Antioxidant rich, Champagne extract is used in our products because it's incredibly effective at protecting and nourishing your skin. We believe that beautiful, healthy skin is worth celebrating every day," says Mathias Tonnesson, CEO of Laboratoire Fleur de Sant.

Champagne takes on a whole new meaning in skin care

The most famous sparkling wine in the world isn't just for drinking any more.

Fleur de Sant has captured its essence for the ultimate global anti-aging range of products. Extremely rich in antioxidants (Resveratrol), Champagne is one of the most beneficial ingredients protecting skin from free radicals and stress to which we are exposed every day by breathing in pollution or being unprotected from UV light.

By counteracting these factors, Champagne extract reduces the damaging marks photo-aging leaves on your skin (wrinkles, sagging skin, dark spots). It works by restoring the skin's structural tissue collagen and elastin to make it more resistant to various environmental aggressors. Antioxidants, which Champagne owes to grape seed extract, are of the highest potency, being at least 20 times more powerful than Vitamin C or E. In Fleur de Sant products, the exclusive Extrait de Champagne is further enhanced by grape seed Phyto-StemCell Infusion, which together deliver tremendously strong anti-aging force.

For more information about Fleur de Sant Champagne Collection, visit http://www.fleurdesante.com/products/

What makes phyto-stem cells so special?

Phyto-stem cells counteract the negative effect of the UV light, help maintain skin stem cell's functions and reinforce their capacity to grow, which in turn slows down the skin aging process. On top of this, they accelerate regeneration and the tissue building functions of skin, resulting in restoration of firmness and wrinkle reduction.

About Laboratoire Fleur de Sant

Fleur de Sant was founded in 1980, with the distinction of being the only brand in the world to utilize Swedish and French medicinal flowers in their beneficial formulations. The tradition continues as the brand is experiencing a re-birth with CEO Mathias Tonnesson. His passion to create skin care with "every detail considered" sees the latest clinically proven collections containing antioxidant-rich Champagne extract, plant stem cell-boosted flowers, and airless packaging that makes every formulation more effective. 95% natural and never tested on animals, Fleur de Sant is more than premium skin care it is the result of one man's passion to create products made from love.

Visit: http://www.fleurdesante.com

Contact: Mathias Tonnesson CEO, Laboratoire Fleur de Sant +1 (646) 893-4100Ext: 100 145363@email4pr.com

To view the original version on PR Newswire, visit:http://www.prnewswire.com/news-releases/celebrate-your-skin-with-champagne--phyto-stemcells-300404181.html

SOURCE Laboratoire Fleur de Sante

Home

Excerpt from:
Celebrate Your Skin with Champagne & Phyto-StemCells - PR Newswire (press release)

Could we one day make babies from only skin cells? – CNN

In 2016, scientists in Japan revealed the birth of mice from eggs made from a parent's skin cells, and many researchers believe the technique could one day be applied to humans.

The process, called in vitro gametogenesis, allows eggs and sperm to be created in a culture dish in the lab.

Though most scientists agree we're still a long way off from doing it clinically, it's a promising technology that has the potential to replace traditional in vitro fertilization to treat infertility.

If and when this process is successful in humans, the implications would be immense, but scientists are now raising legal and ethical questions that need to be addressed before the technology becomes a reality.

In vitro gametogenesis, or IVG, is similar to IVF -- in vitro fertilization -- in that the joining of egg and sperm takes place in a culture dish.

Trounson believes IVG can provide hope for couples when IVF is not an option.

This procedure can "help men or women who have no gametes -- no sperm or eggs," said Trounson, a renowned stem cell scientist best known for developing human IVF with Carl Wood in 1977.

Another potential benefit with IVG is that there is no need for a woman to receive high doses of fertility drugs to retrieve her eggs, as with traditional IVF.

In addition, same-sex couples would be able to have biological children, and people who lost their gametes through cancer treatments, for instance, would have a chance at having biological children.

In theory, a single woman could also conceive on her own, a concept that Sonia M. Suter, professor of law at George Washington University, calls "solo IVG." She points out that it comes with some risk, as there will be less genetic variety among the babies.

She added that the risk is even greater than with cloning and although you could use genetic diagnosis to find disease in embryos before implantation, it wouldn't fully reduce the risk.

This all contributes to the fact that IVG is much more complicated than one might think, and experts add that the process will be even more complex in humans than in mice.

"It's a much tougher prospect to do this in a human -- much, much tougher. It's like climbing a few stairs versus climbing a mountain," Trounson said.

"Gametogenesis (in a mouse) is much faster. Everything is much faster and less complicated than you have in a human. So you've got to make sure there's very long intervals to get you the right outcome. ... Life, gametogenesis, everything, is much, much briefer than it is in a human."

Most scientists are reluctant to commit to an exact time frame, but it's probably safe to say they're many years away.

Knoepfler used the example of an unapproved and, he says, potentially dangerous three-person baby produced in Mexico in 2016 by a US doctor without FDA approval.

Creating a three-person baby involves a process known as pronuclear transfer, in which an embryo is created using genetic material from three people -- the intended mother and father and an egg donor -- to remove the risk of genetic diseases caused by DNA in a mother's mitochondria. The mitochondria are parts of a cell used to create energy but also carry DNA that is passed on only through the maternal line.

This process recently received approval in the UK, but it remains illegal in many countries, including the US.

"Because it seems rogue biomedical endeavors are on the increase, someone could try IVG without sufficient data or governmental approval in the next five to 10 years," Knoepfler said.

"IVG takes us into uncharted territory, so it's hard to say legal issues that might come up," he said, adding that "even other more extreme technologies, such as cloning, of the reproductive kind are not technically prohibited in the US."

For IVG to be researched further, it will be necessary to perform IVF using the derived gametes and then to study the embryos in ways that would involve their destruction. "At a minimum, federal funding could not be used for such work, but what other laws might come into play is less clear," Knoepler said.

In several countries, the implantation of a fertilized egg is not allowed if it's been maintained longer than 14 days.

Dr. Mahendra Rao, scientific adviser with the New York Stem Cell Foundation, explained that in the US, scientists can legally make sperm and oocytes (immature eggs) from other cells and perform IVF. But they would not be able to perform implantation, even in animals.

He said there needs to be clarity that this rule doesn't apply to "synthetic" embryos scientists are building in culture, where there's no intention of implanting them.

Daley and his co-authors highlight concerns over "embryo farming" and the consequence of parents choosing an embryo with preferred traits.

"IVG could, depending on its ultimate financial cost, greatly increase the number of embryos from which to select, thus exacerbating concerns about parents selecting for their 'ideal' future child," they write.

With a large number of eggs available through IVG, the process might exacerbate concerns about the devaluation of human life, the authors say.

Also worrying is the potential for someone to get hold of your genetic material -- such as sloughed-off skin cells -- without your permission. The authors raise questions about the legal ramifications and how they would be handled in court.

"Should the law consider the source of the skin cells to be a legal parent to the child, or should it distinguish an individual's genetic and legal parentage?" they ask.

As new forms of assisted reproductive technology stretch our ideas about identity, parentage and existing laws and regulations around stem cell research, researchers highlight the need to address these thoughts and have answers in place before making IVG an option.

See the article here:
Could we one day make babies from only skin cells? - CNN

Stem Cells – SciTechStory

Stem cells are often in the news. These days its usually about some advance in research. Sometimes the controversy about using embryonic stem cells resurfaces. Despite all the coverage (pro or con) stem cells are not well understood. What are they and why are they important?

In more ways than one, its the potential of stem cells that makes them important. At the moment most of the work with stem cells is still in the laboratory; but thats changing. Within the next few years stem cells, in one form or another, will be at work in medical applications such as repairing a damaged pancreas or a heart. In fact, stem cells will be used to repair or even re-grow tissues all over the body skin, liver, lungs, bone marrow. The production of stem cells, their delivery, and procedures for using them will become the basis of an industry. In the not too distant future stem cells, or the knowledge we gain from working with them, will be used in sophisticated repair of the brain and as part of the development of replacement organs. The potential is enormous.

What are stem cells?

Stem cells are found in most multicellular creatures and come in different varieties; all have an important ability: They can fully reproduce themselves almost indefinitely. For example, in mammals like human beings, blood stem cells (hematopoietic stem cells) are active all our lives in the marrow of bones, where they continually produce the many different kinds of blood cells. Therein is another key property for most stem cells; they can become other kinds of cells. The word for this process is differentiate; blood stem cells can differentiate into red blood cells, white blood cells, blood platelets and so forth. The ability to produce different kinds of cells is why stem cells may be used, for example, to repair or replace damaged heart cells something mature heart cells cannot do on their own.

Stem cell jargon

When you read about stem cells, there are a number of words that jump out jargon, yes, but still descriptive. Stem cells are classified by their potency, that is, what other kinds of cells they can become, or put another way, their ability to differentiate into other cells. There is a rank order from more to less potent:

Totipotent sometimes also called omnipotent stem cells can construct a complete and viable organism. In short, they are the same as a cell created by the fusion of the egg and a sperm (an embryonic cell). Totipotent cells can become any type of cell.

Pluripotent stem cells are derived from totipotent cells and are nearly as versatile. They can become any type of cell, except embryonic.

Multipotent stem cells can become a wide variety of cells, but only those of a close family, for example blood stem cells (hematopoietic cells) can become any of the blood cells, but not other kinds of cells.

Oligopotent stem cells are limited to becoming specific types of cells, such as endoderm, ectoderm, and mesoderm.

Unipotent stem cells can only produce cells of their own type, for example skin cells. They can renew themselves (replicate indefinitely), which distinguishes them from non-stem cells.

To a certain extent the potency of a stem cell relates to its usefulness. In one view of an ideal (lab) world, only totipotent stem cells would be used because they can become any other kind of cell. The real world (lab or otherwise) doesnt work that way. For one thing, stem cells of lesser versatility than totipotent cells are valuable for use in specific applications. Even unipotent stem cells, lowest on the potency poll, are arguably better suited for some targeted uses than more generic stem cells. Most importantly, for many uses, especially for medical purposes, pluripotent stem cells are extremely versatile and less controversial.

Avoiding embryonic stem cells

The true totipotent stem cell is a fertilized egg one embryonic cell. To obtain it means detecting and collecting the cell shortly after fertilization and before it begins to divide. Collecting embryonic stem cells one at a time is very difficult and very expensive. Also, in some parts of the world, using embryonic stem cells is highly controversial, usually on religious grounds. Collecting embryonic stem cells can be considered abortion, since the procedure means the cell(s) will not become an embryo. The label abortion is also applied to collecting embryonic stem cells (by gastrulation) shortly after the first fertilized cell begins to divide. These cells, obviously more numerous, are pluripotent and have been the mainstay of stem cell research.

The history of opposition to the use of embryonic stem cells goes back to the 1990s, when stem cell research was in its own infancy. At that time the only source of viable laboratory stem cells was from in vitro living donors. Most of these were harvested from fertilization clinics. They were so difficult to acquire that only a few stem cell lines (painstakingly cultivated generations of embryonic stem cells) were available. Even those were controversial. The United States banned the taking of embryonic stem cells except for 23 grandfathered lines. (This ban was lifted in 2009.)

The controversy over embryonic stem cells can be avoided primarily in two ways. One way is to use adult stem cells. The word adult is a bit misleading since the cells may be derived from fetuses, newborns, and children, which is why theyre sometimes called somatic stem cells. It means that these stem cells come from relatively mature tissue, cells that are already differentiated to a certain degree. Thats why adult stem cells are almost always classified as multipotent, oligopotent, or unipotent. The other way is to transform adult stem cells into pluripotent stem cells. Many approaches to this transformation are being explored in labs all over the world. Some approaches are derived from fetal/newborn substances such as amniotic fluid and placental or umbilical tissue. Other approaches use mature (differentiated) stem cells, such as those from skin, and genetically modify them until they become pluripotent. Such cells are called induced pluripotent stem cells, often abbreviated as iPSC.

At the moment, it is not possible to say which approaches to stem cell production and application will be the most effective. Even some that seem unlikely (stem cells from skin cells?) may turn out to be the most economical and useful. Still, this is where the payoff for stem cell research lies both in terms of scientific knowledge and in profits for medical applications. Consequently the amount of research work in progress is substantial, and often competitive.

Stem Cell Tourism

Because experimental medical techniques and human desperation can add up to big money, there is a developing market for stem cell applications for a variety of medical disorders. Unfortunately, at least for now, with the exception of blood cell transplants and skin cell treatments, most of these applications are either fraudulent or based on shaky experimental results. In general, most stem cell treatments are at best unethical and often illegal; however, their status around the world is a patchwork quilt of laws and regulations (or their absence). It is a near ideal situation for scam artists to lure desperate people into traveling long distances for stem cell treatment that is illegal in their own country. Hence the name: stem cell tourism.

Tracking the Impact of Stem Cell Research

In relative terms, stem cell research is just getting started. Researchers have been at it since the 1950s; but one of the most important discoveries so far induced pluripotent stem cells dates back to only 2006. This means that stem cells are: a. Not yet well understood and b. Their use in medicine is largely experimental and tentative. Heres a useful listing of what the National Institute of Health (U.S. NIH) considers some of the major open questions about adult stem cells:

How many kinds of adult stem cells exist, and in which tissues do they exist? How do adult stem cells evolve during development and how are they maintained in the adult? Are they leftover embryonic stem cells, or do they arise in some other way? Why do stem cells remain in an undifferentiated state when all the cells around them have differentiated? What are the characteristics of their niche that controls their behavior? Do adult stem cells have the capacity to transdifferentiate, and is it possible to control this process to improve its reliability and efficiency? If the beneficial effect of adult stem cell transplantation is a trophic effect, what are the mechanisms? Is donor cell-recipient cell contact required, secretion of factors by the donor cell, or both? What are the factors that control adult stem cell proliferation and differentiation? What are the factors that stimulate stem cells to relocate to sites of injury or damage, and how can this process be enhanced for better healing? [Source: U.S. National Institute of Health]

SciTechStory Impact Area: Stem Cells

Theres not much debate on the importance of stem cell research. It has already had major impact on our understanding of cell biology, and it will provide more. It is just beginning to have an impact on medicine, with much more to come. In fact, news about stem cell research already occurs once or twice a week (on average) that pace is likely to increase. As a matter of keeping up, its necessary to attempt sorting lab work from practical application, which is to say sorting promise from delivery. Even at that it will be difficult to select which stem cell stories are significant.

More:
Stem Cells - SciTechStory

Stem-cell-derived cells flag a possible new treatment for rare blood disorder – Medical Xpress

February 8, 2017 Blood stem cells from patients with Diamond-Blackfan anemia dont mature properly (right two columns). Credit: Doulatov et al., Science Translational Medicine (2017)

Researchers at Boston Children's Hospital's Stem Cell Research Program were able, for the first time, to use patients' own cells to create cells similar to those in bone marrow, and then use them to identify potential treatments for a blood disorder. The work was published today by Science Translational Medicine.

The team derived the so-called blood progenitor cells from two patients with Diamond Blackfan anemia (DBA), a rare, severe blood disorder in which the bone marrow cannot make enough oxygen-carrying red blood cells. The researchers first converted some of the patients' skin cells into induced pluripotent stem (iPS) cells. They then got the iPS cells to make blood progenitor cells, which they loaded into a high-throughput drug screening system. Testing a library of 1,440 chemicals, the team found several that showed promise in a dish. One compound, SMER28, was able to get live mice and zebrafish to start churning out red blood cells.

The study marks an important advance in the stem cell field. iPS cells, theoretically capable of making virtually any cell type, were first created in the lab in 2006 from skin cells treated with genetic reprogramming factors. Specialized cells generated by iPS cells have been used to look for drugs for a variety of diseasesexcept for blood disorders, because of technical problems in getting iPS cells to make blood cells.

"iPS cells have been hard to instruct when it comes to making blood," says Sergei Doulatov, PhD, co-first author on the paper with Linda Vo and Elizabeth Macari, PhD. "This is the first time iPS cells have been used to identify a drug to treat a blood disorder."

DBA currently is treated with steroids, but these drugs help only about half of patients, and some of them eventually stop responding. When steroids fail, patients must receive lifelong blood transfusions and quality of life for many patients is poor. The researchers believe SMER28 or a similar compound might offer another option.

"It is very satisfying as physician scientists to find new potential treatments for rare blood diseases such as Diamond Blackfan anemia," says Leonard Zon, MD, director of Boston Children's Stem Cell Research Program and co-corresponding author on the paper with George Q. Daley, MD, PhD. "This work illustrates a wonderful triumph," says Daley, associate director of the Stem Cell Research Program and also dean of Harvard Medical School.

Making red blood cells

As in DBA itself, the patient-derived blood progenitor cells, studied in a dish, failed to generate the precursors of red blood cells, known as erythroid cells. The same was true when the cells were transplanted into mice. But the chemical screen got several "hits": in wells loaded with these chemicals, erythroid cells began appearing.

Because of its especially strong effect, SMER28 was put through additional testing. When used to treat the marrow in zebrafish and mouse models of DBA, the animals made erythroid progenitor cells that in turn made red blood cells, reversing or stabilizing anemia. The same was true in cells from DBA patients transplanted into mice. The higher the dose of SMER28, the more red blood cells were produced, and no ill effects were found. (Formal toxicity studies have not yet been conducted.)

Circumventing a roadblock

Previous researchers have tried for years to isolate blood stem cells from patients. They have sometimes succeeded, but the cells are very rare and cannot create enough copies of themselves to be useful for research. Attempts to get iPS cells to make blood stem cells have also failed.

The Boston Children's researchers were able to circumvent these problems by instead transforming iPS cells into blood progenitor cells using a combination of five reprogramming factors. Blood progenitor cells share many properties with blood stem cells and are readily multiplied in a dish.

"Drug screens are usually done in duplicate, in tens of thousands of wells, so you need a lot of cells," says Doulatov, who now heads a lab at the University of Washington. "Although blood progenitor cells aren't bona fide stem cells, they are multipotent and they made red cells just fine."

SMER28 has been tested preclinically for some neurodegenerative diseases. It activates a so-called autophagy pathway that recycles damaged cellular components. In DBA, SMER28 appears to turn on autophagy in erythroid progenitors. Doulatov plans to further explore how this interferes with red blood cell production.

Explore further: Scientists find that persistent infections in mice exhaust progenitors of all blood cells

More information: "Drug discovery for Diamond-Blackfan anemia using reprogrammed hematopoietic progenitors," Science Translational Medicine stm.sciencemag.org/lookup/doi/10.1126/scitranslmed.aah5645

Researchers at Boston Children's Hospital's Stem Cell Research Program were able, for the first time, to use patients' own cells to create cells similar to those in bone marrow, and then use them to identify potential treatments ...

Methicillin-resistant Staphylococcus aureus (MRSA) infections are caused by a type of staph bacteria that has become resistant to the antibiotics used to treat ordinary staph infections. The rise of MRSA infections is limiting ...

Adding just the right mixture of signaling moleculesproteins involved in developmentto human stem cells can coax them to resemble somites, which are groups of cells that give rise to skeletal muscles, bones, and cartilage ...

What is hearing? For Valeriy Shafiro, PhD, that question is fundamental, even though it's one that most people who hear well may probably never think about.

Massachusetts General Hospital (MGH) investigators have developed a novel approach to analyze brainwaves during sleep, which promises to give a more detailed and accurate depiction of neurophysiological changes than provided ...

An expectant mother's exposure to the endocrine-disrupting chemical bisphenol A (BPA) can raise her offspring's risk of obesity by reducing sensitivity to a hormone responsible for controlling appetite, according to a mouse ...

Please sign in to add a comment. Registration is free, and takes less than a minute. Read more

Link:
Stem-cell-derived cells flag a possible new treatment for rare blood disorder - Medical Xpress

Archives