Page 1,410«..1020..1,4091,4101,4111,412..1,4201,430..»

Stem Cell Treatment for Children With Spina Bifida Helps Dogs First … – UC Davis

Posted: August 26, 2017 at 6:40 pm

A pair of English bulldog puppies are the first patients to be successfully treated with a unique therapy a combination of surgery and stem cells developed at the University of California, Davis, to help preserve lower-limb function in children with spina bifida.

(Editorsnote: Photos and b-roll available.)

Because dogs with the birth defect frequently have little control of their hindquarters, they also have little hope for a future. They are typically euthanized as puppies.

At their postsurgery re-check at 4 months old, however, the siblings, named Darla and Spanky, showed off their abilities to walk, run and play to their doctor, veterinary neurosurgeon Beverly Sturges.

The initial results of the surgery are promising, as far as hind limb control, said Sturges. Both dogs seemed to have improved range of motion and control of their limbs.

The dogs have since been adopted, and continue to do well at their home in New Mexico.

Spina bifida occurs when spinal tissue improperly fuses in utero, causing a range of cognitive, mobility, urinary and bowel disabilities in about 1,500 to 2,000 children born in the U.S. each year. The dogs procedure, which involved surgical techniques developed by fetal surgeon Diana Farmer of UC Davis Health together with a cellular treatment developed by stem cell scientists Aijun Wang and Dori Borjesson, director of the universitys Veterinary Institute for Regenerative Cures, represents a major step toward curing spina bifida for both humans and dogs.

Farmer pioneered the use of surgery prior to birth to improve brain development in children with spina bifida. She later showed that prenatal surgery combined with human placenta-derived mesenchymal stromal cells (PMSCs), held in place with a cellular scaffold, helped research lambs born with the disorder walk without noticeable disability.

Sturges wanted to find out if the surgery-plus-stem-cell approach could give dogs closer-to-normal lives along with better chances of survival and adoption. At 10-weeks old, Darla and Spanky were transported from Southern California Bulldog Rescue to the UC Davis veterinary hospital, where they were the first dogs to receive the treatment, this time using canine instead of human PMSCs.

Another distinction for Darla and Spanky is that their treatment occurred after birth, since prenatal diagnosis of spina bifida is not performed on dogs, Sturges explained. The disorder becomes apparent between 1 and 2 weeks of age, when puppies show hind-end weakness, poor muscle tone, incoordination and abnormal use of their tails.

UC Davis is the only place where this type of cross-disciplinary, transformational medicine could happen, according to Farmer.

Its rare to have a combination of excellent medical and veterinary schools and strong commitment to advancing stem cell science at one institution, she said.

UC Davis is also home to the One Healthinitiative aimed at finding novel treatments like these for diseases that affect both humans and animals.

Ive often said that I have the greatest job on the planet, because I get to help kids, Farmer said. Now my job is even better, because I get to help puppies too.

With additional evaluation and U.S. Food and Drug Administration approval, Farmer and Wang hope to test the therapy in human clinical trials. Sturges and Borjesson hope to do the same with a canine clinical trial. They hope the outcomes of their work help eradicate spina bifida in dogs and humans.

In the meantime, the team wants dog breeders to send more puppies with spina bifida to UC Davis for treatment and refinements that help the researchers fix an additional hallmark of spina bifida incontinence. While Darla and Spanky are very mobile and doing well on their feet, they still require diapers.

Further analysis of their progress will determine if the surgery improves their incontinence conditions, Sturges said.

Funding for this project was provided by the Veterinary Institute for Regenerative Cures (VIRC) at the UC Davis School of Veterinary Medicine, and the Surgical Bioengineering Lab at the UC Davis School of Medicine. Private donations to the veterinary school for stem cell research also contributed to this procedure. Farmer and Wangs spina bifida research is supported by funding from the National Institutes of Health, the California Institute for Regenerative Medicine, Shriners Hospitals for Children and the March of Dimes Foundation.

Original post:
Stem Cell Treatment for Children With Spina Bifida Helps Dogs First ... - UC Davis

Posted in Stem Cell Research | Comments Off on Stem Cell Treatment for Children With Spina Bifida Helps Dogs First … – UC Davis

A new clue to hair loss: A misbehaving enzyme in follicle stem cells – STAT

Posted: August 26, 2017 at 6:40 pm

T

he roots of hair loss run deep: Its linked to hormonal balance, immune response, stem cell signaling, and now, according to new research from University of California, Los Angeles metabolism.

The study, published inNature Cell Biology, finds that the metabolism in the stem cells embedded in hair follicles is different from surrounding cells. When they tinkered with that metabolic pathway in mice, they could either halt hair growth or make it proliferate. The UCLA researchers are now testing out a duo of drugs to try and prompt that hair to grow.

This is a STAT Plus article and is only available to STAT Plus subscribers.To read the full story, subscribe to STAT Plus or log in to your account.Good news: your first 30 days are on us.

Biotech Correspondent

Meghana covers biotech and writes The Readout newsletter.

Read more:
A new clue to hair loss: A misbehaving enzyme in follicle stem cells - STAT

Posted in Stem Cell Research | Comments Off on A new clue to hair loss: A misbehaving enzyme in follicle stem cells – STAT

Stem cell research could double avo production – Fruitnet

Posted: August 26, 2017 at 6:40 pm

Researchers at the University of Queensland (UQ) have developed a stem cell multiplication method that could double the states avocado production.

The growing method could lead to 500 times more avocado plants being supplied to the industry, and could reduce the time it takes for avocado orchards to mature.

Neena Mitter from the Queensland Alliance for Agriculture & Food Innovation, said the technology would be a potential game changer the global avocado industry,which is currently experiencing a backlog of plant orders until 2020.

At present, to supply new trees, the avocado industry follows the same process they have for the last 40 years, which is to take cuttings from high quality trees and root them, Mitter said. However, this is a cumbersome, labour and resource intensive process, as it takes about 18 months from the cutting stage to having a plant for sale, which creates a huge bottleneck for nurseries across the globe in the number of trees that they can supply trees to growers."

The non-GM and environmentally friendly technology, however, can grow and root multiple avocado plants from the shoot tip of an existing plant.

[With the new technology] ten-thousand plants can be generated in a 10m2 room on a soil-less media, Mitter said.

More than 600 plants developed by the stem cell multiplication method will be tested at different sites across Australia, with the research team also looking into whether heat-adapted avocado trees can grow alongside banana plants.

The Queensland Alliance for Agriculture & Food Innovation is a UQ research institute, with funding from the Queensland government.

Link:
Stem cell research could double avo production - Fruitnet

Posted in Stem Cell Research | Comments Off on Stem cell research could double avo production – Fruitnet

Mouse model of human immune system inadequate for stem cell … – Stanford Medical Center Report

Posted: August 26, 2017 at 6:40 pm

In an ideal situation, these humanized mice would reject foreign stem cells just as a human patient would.

Wu shares senior authorship of the research, which was published Aug. 22 in Cell Reports, with Dale Greiner, PhD, professor in the Program in Molecular Medicine at the University of Massachusetts Medical School, and Leonard Shultz, PhD, professor at the Jackson Laboratory. Former postdoctoral scholars Nigel Kooreman, MD, and Patricia de Almeida, PhD, and graduate student Jonathan Stack, DVM, share lead authorship of the study.

Although these mice are fully functional in their immune response to HIV infection or after transplantation of other tissues, they are unable to completely reject the stem cells, said Kooreman. Understanding why this is, and whether we can overcome this deficiency, is a critical step in advancing stem cell therapies in humans.

Humanized mice are critical preclinical models in many biomedical fields helping to bring basic science into the clinic, but as this work shows, it is critical to frame the question properly, said Greiner. Multiple laboratories remain committed to advancing our understanding and enhancing the function of engrafted human immune systems.

Greiner and Shultz helped to pioneer the use of humanized mice in the 1990s to model human diseases and they provided the mice used in the study.

The researchers were studying pluripotent stem cells, which can become any tissue in the body. They tested the animals immune response to human embryonic stem cells, which are naturally pluripotent, and to induced pluripotent stem cells. Although iPS cells can be made from a patients own tissues, future clinical applications will likely rely on pre-screened, FDA-approved banks of stem cell-derived products developed for specific clinical situations, such as heart muscle cells to repair tissue damaged by a heart attack, or endothelial cells to stimulate new blood vessel growth. Unlike patient-specific iPS cells, these cells would be reliable and immediately available for clinical use. But because they wont genetically match each patient, its likely that they would be rejected without giving the recipients immunosuppressive drugs.

Humanized mice were first developed in the 1980s. Researchers genetically engineered the mice to be unable to develop their own immune system. They then used human immune and bone marrow precursor cells to reconstitute the animals immune system. Over the years subsequent studies have shown that the human immune cells survive better when fragments of the human thymus and liver are also implanted into the animals.

Kooreman and his colleagues found that two varieties of humanized mice were unable to completely reject unrelated human embryonic stem cells or iPS cells, despite the fact that some human immune cells homed to and were active in the transplanted stem cell grafts. In some cases, the cells not only thrived, but grew rapidly to form cancers called teratomas. In contrast, mice with unaltered immune systems quickly dispatched both forms of human pluripotent stem cells.

The researchers obtained similar results when they transplanted endothelial cells derived from the pluripotent stem cells.

To understand more about what was happening, Kooreman and his colleagues created a new mouse model similar to the humanized mice. Instead of reconstituting the animals nonexistent immune systems with human cells, however, they used immune and bone marrow cells from a different strain of mice. They then performed the same set of experiments again.

Unlike the humanized mice, these new mice robustly rejected human pluripotent stem cells as well as mouse stem cells from a genetically mismatched strain of mice. In other words, their newly acquired immune systems appeared to be in much better working order.

Although more research needs to be done to identify the cause of the discrepancy between the two types of animals, the researchers speculate it may have something to do with the complexity of the immune system and the need to further optimize the humanized mouse model to perhaps include other types of cells or signaling molecules. In the meantime, they are warning other researchers of potential pitfalls in using this model to screen for immunosuppressive drugs that could be effective after human stem cell transplants.

Many in the fields of pluripotent stem cell research and regenerative medicine are pushing the use of the humanized mice to study the human immune response, said Kooreman. But if we start to make claims using this model, assuming that these cells wont be rejected by patients, it could be worrisome. Our work clearly shows that, although there is some human immune cell activity, these animals dont fully reconstitute the human immune system.

The researchers are hopeful that recent advances may overcome some of the current models limitations.

The immune system is highly complex and there still remains much we need to learn, said Shultz. Each roadblock we identify will only serve as a landmark as we navigate the future. Already, weve seen recent improvements inhumanized mousemodels that foster enhancement of human immune function.

Wu is a member of Stanford Bio-X, the Stanford Cancer Institute and the Stanford Child Health Research Institute. He is also the Simon H. Stertzer Professor.

Additional Stanford co-authors are former research assistant Raman Nelakanti; former postdoctoral scholars Sebastian Diecke, PhD, and Veronica Sanchez-Freire, PhD; postdoctoral scholar Ning-Yi Shao, MD, PhD; instructor Elena Matsa, PhD; and associate professor of pathology Andrew Connolly, MD, PhD.

The research was funded by the California Institute of Regenerative Medicine, the National Institutes of Health (grants R01HL132875, R01HL133272, P30CA034196, UC4DK104218 and T32OD01112) and the Helmsley Charitable Trust.

Stanfords Department of Medicine also supported the work.

Original post:
Mouse model of human immune system inadequate for stem cell ... - Stanford Medical Center Report

Posted in Stem Cell Research | Comments Off on Mouse model of human immune system inadequate for stem cell … – Stanford Medical Center Report

Trial of Lung Disease Stem Cell Therapy Could Come by Year’s End – Lung Disease News

Posted: August 26, 2017 at 6:40 pm

University of North Carolina Health Careresearchers have made strides toward a stem cell treatment for lung diseases such as pulmonary fibrosis, COPD, and cystic fibrosis.

In fact, they are discussing the start of clinical trials with regulatory authorities.

The team discussed its work in two recent studies. One provedthat it is possible to isolate lung stem cells with a relatively non-invasive procedure. The other showed that stem cells reduce fibrosis in rats with pulmonary fibrosis.

The first study, in the journal Respiratory Research, was titledDerivation of therapeutic lung spheroid cells from minimally invasive transbronchial pulmonary biopsies.The second, inStem Cells Translational Medicine, was Safety and Efficacy of Allogeneic Lung Spheroid Cells in a Mismatched Rat Model of Pulmonary Fibrosis.

This is the first time anyone has generated potentially therapeutic lung stem cells from minimally invasive biopsy specimens, Dr. Jason Lobo, director of the universitys lung transplant and interstitial lung disease program,said in a press release. Hewas co-senior author of both studies.

We think the properties of these cells make them potentially therapeutic for a wide range of lung fibrosis diseases, added Dr. Ke Cheng, who led the studies with Lobo. He is anassociate professor in North Carolina State Universitys Department of Molecular Biomedical Sciences.

The research team had previously homed in on stem and support cells they could isolate from a lung tissue sample and grow in a lab. The tissue formed sphere-like structures in a lab dish, prompting the scientists to call them lung spheroid cells.

In 2015, the team showed that these cells had potent regenerative properties in animals with lung diseases. In fact, the stem cells they cultivated outperformed another type called mesenchymal stem cells.

Their latest project involved gathering lung spheroid cells from patients with various lung diseases. They used a procedure calleda transbronchial biopsy thatcan be done in a doctors office.

We snip tiny, seed-sized samples of airway tissue using a bronchoscope, Lobo said. This method involves far less risk to the patient than does a standard, chest-penetrating surgical biopsy of lung tissue.

From this tiny piece of airway, researchers gathered stem cells, then allowed them to multiply because stem cell treatments require infusions of millions of such cells.

When they injected the cells intravenously into mice, the discovered that most found their way into the animals lungs.

These cells are from the lung, and so in a sense theyre happiest, so to speak, living and working in the lung, Cheng said.

The team then tested the treatment in rats exposed to a chemical that triggers lung fibrosis. The lung spheroid cells gave rise to healthy lung cells, reducing both inflammation and fibrosis in the animals lungs.

Also, the treatment was safe and effective whether the lung spheroid cells were derived from the recipients own lungs or from the lungs of an unrelated strain of rats, Lobo said. In other words, even if the donated stem cells were foreign, they did not provoke a harmful immune reaction in the recipient animals, as transplanted tissue normally does.

The researchers said that in humans their goal would be to use patients own stem cells to minimize the risk of immune reactions. But because large quantities of cells are needed, it might be necessary to gather cells from healthy volunteers or organ donation networks as well.

Our vision is that we will eventually set up a universal cell donor bank, Cheng said.

The team is in discussions with the U.S. Food and Drug Administration aimed at starting the first human study by years end. The first trial would include a small group of pulmonary fibrosis patients. The team also hopes their spheroid stem cell therapy will help patients with other lung diseases.

Originally posted here:
Trial of Lung Disease Stem Cell Therapy Could Come by Year's End - Lung Disease News

Posted in Stem Cell Research | Comments Off on Trial of Lung Disease Stem Cell Therapy Could Come by Year’s End – Lung Disease News

Stem Cells, Fetal Tissue Research & Cloning – Oregon Right …

Posted: August 25, 2017 at 6:45 am

Stem cell research is a controversial issue that sharply divides people. There are two kinds of stem cell research: embryonic and adult. This distinction is imperative because of the ethical issues involved.

Embryonic stem cell research requires cells to be extracted from a human embryo. In the process of extracting the stem cells the embryo is destroyed and a life is ended. The embryonic stem cells are then isolated and theoretically coaxed into developing into just about any cell in the body. Embryonic stem cell transplants have not been shown concretely to have successfully helped a single patient.

Fetal tissue research requires the abortion of a living unborn child. The Center for Medical Progress, in a series of investigative videos (link to site), revealed that Planned Parenthoods affiliated clinics participate in the harvesting and sale of aborted baby body parts and placental tissue for financial gain. These are then used in research facilities around the country, including at Oregon Health and Science University. Fetal tissue research has also not successfully helped treat a single patient.

Adult stem cell research on the other hand, does not require the destruction of life. Adult stem cells are derived from sources like umbilical cords and organ tissue.

View post:
Stem Cells, Fetal Tissue Research & Cloning - Oregon Right ...

Posted in Oregon Stem Cells | Comments Off on Stem Cells, Fetal Tissue Research & Cloning – Oregon Right …

New Biomedical Engineering Grants Aim at Heart Failure and Resistant High Blood Pressure – Newswise (press release)

Posted: August 25, 2017 at 6:45 am

Newswise BIRMINGHAM, Ala. Biomedical engineering researchers will attack two banes of cardiovascular disease heart failure after heart attacks and the scourge of resistant high blood pressure with $4.8 million in National Institutes of Health grants that begin this fall.

One sign of the clinical significance of this research by the University of Alabama at Birmingham investigators are the percentile scores that Jianyi Jay Zhang, M.D., Ph.D., and Gangjian Qin, M.D., received in those two NIH grant applications.

Zhangs plan to dissect the mechanisms of electromechanical integration of a human heart-muscle patch to aid survival and stability of the patch garnered a 1 percentile score, the highest possible. Qins plan to dissect a novel molecular pathway in endothelial cells of arteries that appears to regulate contractile function and blood pressure has significant potential to improve human health from the disease and death caused by high blood pressure, NIH reviewers said, and Qin received a 2 percentile score.

Zhang, chair and professor of the UAB Department of Biomedical Engineering and holder of the T. Michael and Gillian Goodrich Endowed Chair of Engineering Leadership, will receive $2.5 million over four years. Qin, professor of biomedical engineering and director of the Molecular Cardiology Program, will receive $2.3 million over four years.

Zhang came to UAB in 2015 from the University of Minnesota Medical School with the goal of moving his work with engineered heart patches into human use within seven years. As chair of Biomedical Engineering, a joint department of the UAB School of Medicine and the UAB School of Engineering, Zhang has recruited top researchers, and he also was awarded $11 million of NIH funding in 2016 $8 million of which is shared in collaborations with University of Wisconsin and Duke University researchers.

One of the recent recruits to biomedical engineering is Qin, who serendipitously discovered a novel and fascinating line of research that may lead to new drugs for treatment-resistant high blood pressure, where existing blood pressure drugs are ineffective. People with resistant high blood pressure have increased risk of strokes, heart attacks, heart failure and arterial aneurysms, and high blood pressure is a leading cause of chronic kidney failure. Even moderately elevated arterial blood pressure shortens life expectancy.

At the time, Qin was interested in the often fatal heart failure that occurs months or years after heart attacks. He reasoned that growth of new blood vessels into the damaged heart tissue of the left ventricle could be boosted by altering the amounts of cell-cycle regulators in the E2F family of transcription factors, to speed division of cells in the endothelial tissue of arteries.

When he deleted one of the eight E2Fs that are found in mice and humans E2F2 it had no effect on cell growth. But unexpectedly, we found a striking function, Qin said. If you delete E2F2, the vessel is more contractile. It becomes rigid and hard, and this contributes to high blood pressure.

So we had a question: How does E2F2 interact with other molecules to regulate blood pressure? Qin did pull-down experiments with E2F2, where other proteins are flowed past tethered E2F2 molecules to see if any would bind. He found that a kinase enzyme called Sam68 did bind to the transcription factor.

When he knocked out the gene for Sam68 in mice, they had low blood pressure.

Ultimately, a series of experiments in Qins lab and observations of other laboratories suggested a previously unknown mechanism of blood pressure control that involves E2F2/Sam68 and the expression of endothelial converting enzyme 1b. ECE-1b affects the levels of peptides that constrict blood vessels and raise blood pressure. Dysregulation of this pathway may contribute to blood pressure disorders, especially hypertension.

Despite a strong correlation, the E2F2/Sam68-ECE-1b pathway has not explicitly been linked to blood pressure regulation, and the mechanisms of how Sam68/E2F2 signaling regulates ECE-1b expression and blood vessel function remain uncharacterized.

Qin will use his new grant to search for the link to blood pressure regulation and characterize the mechanisms. His research could provide the missing links between the results of large-scale genomewide association studies of human high blood pressure and its pathogenesis namely how dysregulation leads to refractory hypertension.

Detailed knowledge of those steps would offer new targets for potential new drugs, which are especially needed to prevent or treat resistant hypertension.

Qin says he was attracted to UAB by the strong focus of clinicians and basic scientists on solving the clinical problem of hypertension, as well as the depth and breadth of cardiovascular disease research in biomedical engineering, the UAB Department of Pathology and the UAB Division of Cardiovascular Disease. He also has great interest in Zhangs research, where Qins past work in stem cell biology and cardiovascular science can contribute.

As measured by NIH funding, the UAB Department of Biomedical Engineering is the fourth-ranked biomedical engineering department among all departments that are jointly led by schools of medicine and engineering, according to the 2016 Blue Ridge NIH database.

The joint biomedical engineering departments ahead of UAB are at Stanford University, Johns Hopkins University, and Oregon Health and Science University. Those trailing UAB in the funding ranking are at the University of North Carolina-Chapel Hill, Emory University, University of Virginia, Case Western Reserve University, University of Colorado-Denver, University of Rochester, the University of Illinois-Chicago, Wake Forest University Health Sciences and State University of New York-Stony Brook.

Read the original here:
New Biomedical Engineering Grants Aim at Heart Failure and Resistant High Blood Pressure - Newswise (press release)

Posted in Oregon Stem Cells | Comments Off on New Biomedical Engineering Grants Aim at Heart Failure and Resistant High Blood Pressure – Newswise (press release)

Gene editing used to repair diseased genes in embryos – NHSUK – NHS Choices

Posted: August 25, 2017 at 6:45 am

Deadly gene mutations removed from human embryos in landmark study, reports The Guardian. Researchers have used a gene-editing technique to repair faults in DNA that can cause an often-fatal heart condition called hypertrophic cardiomyopathy.

This inherited heart condition is caused by a genetic change (mutation) in one or more genes. Babies born with hypertrophic cardiomyopathy have diseased and stiff heart muscles, which can lead to sudden unexpected death in childhood and in young athletes.

In this latest study researchers used a technique called CRISPR-cas9 to target and then remove faulty genes. CRISPR-cas9 acts like a pair of molecular scissors, allowing scientists to cut out certain sections of DNA. The technique has attracted a great deal of excitement in the scientific community since it was released in 2014. But as yet, there have been no practical applications for human health.

The research is at an early stage and cannot legally be used as treatment to help families affected by hypertrophic cardiomyopathy. And none of the modified embryos were implanted in the womb.

While the technique showed a high degree of accuracy, its unclear whether it is safe enough to be developed as a treatment. The sperm used in the study came from just one man with faulty genes, so the study needs to be repeated using cells from other people, to be sure that the findings can be replicated.

Scientists say it is now important for society to start a discussion about the ethical and legal implications of the technology. It is currently against the law to implant genetically altered human embryos to create a pregnancy, although such embryos can be developed for research.

The study was carried out by researchers from Oregon Health and Science University and the Salk Institute for Biological Studies in the US, the Institute for Basic Science and Seoul University in Korea, and BGI-Shenzen and BGI-Quingdao in China. It was funded by Oregon Health and Science University, the Institute for Basic Science, the G. Harold and Leila Y. Mathers Charitable Foundation, the Moxie Foundation and the Leona M. and HarryB. Helmsley Charitable Trust and the Shenzhen Municipal Government of China. The study was published in the peer-reviewed journal Nature.

The Guardian carried a clear and accurate report of the study. While their reports were mostly accurate, ITV News, Sky News and The Independent over-stated the current stage of research, with Sky News and ITV News saying it could eradicate thousands of inherited conditions and the Independent claiming it opens the possibility for inherited diseases to be wiped out entirely. While this may be possible, we dont know whether other inherited diseases might be as easily targeted as this gene mutation.

Finally, the Daily Mail rolls out the arguably tired clich of the technique leading to designer babies, which seems irrelevant at this point. The CRISPR-cas9 technique is only in its infancy and (ethics aside) its simply not possible to use genetic editing to select desirable characteristics - most of which are not the result of one single, identifiable gene. No reputable scientist would attempt such a procedure.

This was a series of experiments carried out in laboratories, to test the effects of the CRISPR-Cas9 technique on human cells and embryos. This type of scientific research helps us understand more about genes and how they can be changed by technology. It doesnt tell us what the effects would be if this was used as a treatment.

Researchers carried out a series of experiments on human cells, using the CRISPR-cas9 technique first on modified skin cells, then on very early embryos, and then on eggs at the point of fertilisation by sperm. They used genetic sequencing and analysis to assess the effects of these different experiments on cells and how they developed, up to five days. They looked specifically to see what proportion of cells carrying faulty mutations could be repaired, whether the process caused other unwanted mutations, and whether the process repaired all cells in an embryo, or just some of them.

They used skin cells (which were modified into stem cells) and sperm from one man, who carried the MYBPC3 mutation in his genome, and donor eggs from women without the genetic mutation. This is the mutation known to cause hypertrophic cardiomyopathy.

Normally in such cases, roughly half the embryos would have the mutation and half would not, as theres a 50-50 chance of the embryo inheriting the male or female version of the gene.

The CRISPR-cas9 technique can be used to select and delete specific genes from a strand of DNA. When this happens, usually the cut ends of the strand join together, but this causes problems so cant be used in the treatment of humans. The scientists created a genetic template of the healthy version of the gene, which they introduced at the same time as using CRISPR-cas9 to cut the mutated gene. They hoped the DNA would repair itself with a healthy version of the gene.

One important problem with changing genetic material is the development of mosaic embryos, where some of the cells have corrected genetic material and others have the original faulty gene. If that happened, doctors would not be able to tell whether or not an embryo was healthy.

The scientists needed to test all the cells in the embryos produced in the experiment, to see whether all cells had the corrected gene or whether the technique had resulted in a mixture. They also did whole genome sequencing on some embryos, to test for unrelated genetic changes that might have been introduced accidentally during the process.

All embryos in the study were destroyed, in line with legislation about genetic research on embryos.

Researchers found that the technique worked on some of the stem cells and embryos, but worked best when used at the point of fertilisation of the egg. There were important differences between the way the repair worked on the stem cells and the egg.

Only 28% of the stem cells were affected by the CRISPR-cas9 technique. Of these, most repaired themselves by joining the ends together, and only 41% were repaired by using a corrected version of the gene.

67% of the embryos exposed to CRISPR-cas9 had only the correct version of the gene higher than the 50% that would have been expected had the technique not been used. 33% of embryos had the mutated version of the gene, either in some or all their cells.

Importantly, the embryos didnt seem to use the template injected into the zygote to carry out the repair, in the way the stem cells did. They used the female version of the healthy gene to carry out the repair, instead.

Of the embryos created using CRISPR-cas9 at the point of fertilisation, 72% had the correct version of the gene in all their cells, and 28% had the mutated version of the gene in all their cells. No embryos were mosaic a mixture of cells with different genomes.

The researchers found no evidence of mutations induced by the technique, when they examined the cells using a variety of techniques. However, they did find some evidence of gene deletions caused by DNA strands splicing (joining) themselves together without repairing the faulty gene.

The researchers say they have demonstrated how human embryos employ a different DNA damage repair system to adult stem cells, which can be used to repair breaks in DNA made using the CRISPR-cas9 gene-editing technique.

They say that targeted gene correction could potentially rescue a substantial portion of mutant human embryos, and increase the numbers available for transfer for couples using pre-implantation diagnosis during IVF treatment.

However, they caution that despite remarkable targeting efficiency, CRISPR-cas9-treated embryos would not currently be suitable for transfer. Genome editing approaches must be further optimised before clinical application can be considered, they say.

Currently, genetically-inherited conditions like hypertrophic cardiomyopathy cannot be cured, only managed to reduce the risk of sudden cardiac death. For couples where one partner carries the mutated gene, the only option to avoid passing it on to their children is pre-implantation genetic diagnosis. This involves using IVF to create embryos, then testing a cell of the embryo to see whether it carries the healthy or mutated version of the gene. Embryos with healthy versions of the gene are then selected for implantation in the womb.

Problems arise if too few or none of the embryos have the correct version of the gene. The researchers suggest their technique could be used to increase the numbers of suitable embryos. However, the research is still at an early stage and has not yet been shown to be safe or effective enough to be considered as a treatment.

The other major factor is ethics and the law. Some people worry that gene editing could lead to designer babies, where couples use the tool to select attributes like hair colour, or even intelligence. At present, gene editing could not do this. Most of our characteristics, especially something as complex as intelligence, are not the result of one single, identifiable gene, so could not be selected in this way. And its likely that, even if gene editing treatments became legally available, they would be restricted to medical conditions.

Designer babies aside, society needs to consider what is acceptable in terms of editing human genetic material in embryos. Some people think that this type of technique is "playing God" or is ethically unacceptable because it involves discarding embryos that carry faulty genes. Others think that its rational to use the scientific techniques we have developed to eliminate causes of suffering, such as inherited diseases.

This research shows that the questions of how we want to legislate for this type of technique are becoming pressing. While the technology is not there yet, it is advancing fast. This research shows just how close we are getting to making genetic editing of human embryos a reality.

Excerpt from:
Gene editing used to repair diseased genes in embryos - NHSUK - NHS Choices

Posted in Oregon Stem Cells | Comments Off on Gene editing used to repair diseased genes in embryos – NHSUK – NHS Choices

Utah family sets up campground in Rexburg to fight cancer – Rexburg Standard Journal

Posted: August 25, 2017 at 6:44 am

REXBURG A family from Sandy, Utah, is renting out space in a field across from Walmart in hopes of raising money to help their mother combat multiple myeloma cancer.

Earlier this year the Paul and Jenny Ahlstrom family went looking for a way to raise money and opted to rent out the farmland. The family started their fundraising about two years ago and so far have raised $477,000 of the $500,000 needed to fund two cancer treatments.

The campsites rent from $200 for tents to $250 for RV camping. The family advertised on various websites, and as of Friday they had rented out about 90 spaces for an estimated 400 people. They have a total of 25 acres to use should they need to open more campsites.

Each of the Ahlstroms six children worked on a portion of the project. The familys youngest opened a lemonade stand while other siblings created the eclipse camp and advertised it on social media in various ways.

Paul Ahlstrom served a Mormon mission with Rexburg farmer Roger Muir, who helped the family secure the land to transform it into a campground.

Before securing the property, the Ahlstroms needed a camping permit. That required them to get 20 portable restrooms, none of which were available in the Gem State.

Who would have thought the limiting factor on setting up a fundraiser would be access to porta-potties? We shipped them from Utah. They were $600 a porta-potty. Thats $12,000 of porta-potties. Yikes! Paul Ahlstrom said.

The joke was that the portable restrooms had been misnamed.

Instead of Honey Buckets, they should be called Gold Buckets. We pretty much bought them, he said.

Campers have been undeterred by the straw covering the field. On Friday the family received four calls about camping there. Had they been there early Friday morning, they would have had seven extra campers.

The Ahlstroms plan to set up a temporary kitchen for their campers. They also plan to host a breakfast, childrens games and the movie Galaxy Quest over the weekend for their guests.

We have central cooking tables and a couple dozen stoves everybody can cook on if they want, Paul Ahlstrom said.

Jenny Ahlstrom also visited the campground on Friday. She was diagnosed with cancer in 2010. Instead of letting the disease get her down, she created a foundation, Myeloma Crowd, and is personally working with doctors from New York to Germany to find a cure. She also hosts a weekly radio show for cancer patients.

According to the Mayo Clinic, multiple myeloma cells attack healthy bone marrow and crowd out normal blood cells.

Rather than produce helpful antibodies, the cancer cells produce abnormal proteins that can cause kidney problems, the clinic's website reported.

The Mayo Clinic reported that about three percent of Americans over the age of 50 suffer from the disease. The older a person is, the more likely it is that they will experience the illness. Men usually develop the disease, and the disease is twice as likely to be found in African-Americans.

Because Jenny Ahlstrom was well under 50, female and of European descent, the diagnosis came as a shock, she said.

I had a lot of bone damage. It hurts your kidneys. Its a plasma cancer, she said.

Jenny Ahlstrom has had two stem cell transplants to fight the disease.

This disease is weird. It always relapses. You can go a year, you can go six months, you can go 10 years, but eventually youll relapse and the treatments will stop working. Its a terminal cancer, she said.

Jenny Ahlstrom expects to return for more treatment as her remission numbers increase. Despite the ongoing treatment, she chooses to remain positive.

The fun part and the exciting part is that we decided to start this foundation, she said.

The Ahlstrom family heard about the eclipse through a friend earlier this month. The family later researched ideal locations to watch the eclipse and ways to earn money toward their foundation.

The primary goal was to help raise money for Jennys cancer foundation, Myeloma Crowd, Paul Ahlstrom said.

They later chose Rexburg because of its generally good weather this time of year and because it was rated as one of the top spots for eclipse viewing. After visiting a local hotel, they decided to set up a campground of their own.

They said the Japanese Space Agency rented their entire hotel for the August eclipse two years ago, Paul Ahlstrom said.

Thanks to Pauls long friendship with Muir, the family decided to host a campsite and earn money that way.

Originally from California, Paul Ahlstrom works as an entrepreneur and has formed various companies. Through the campsite hes currently teaching his own children how to become entrepreneurs as well.

If the family breaks even on the campground endeavor, it will be a lesson well-learned, Paul Ahlstrom said.

We dont know if its a fundraiser or a fun, expensive camping weekend, he said.

See original here:
Utah family sets up campground in Rexburg to fight cancer - Rexburg Standard Journal

Posted in Utah Stem Cells | Comments Off on Utah family sets up campground in Rexburg to fight cancer – Rexburg Standard Journal

This Chip Uses Electricity to Reprogram Cells for Healing – Singularity Hub

Posted: August 25, 2017 at 6:43 am

It sounds like science fiction: with a light zap of electricity, a tiny stamp-like device transforms your skin cells into reservoirs of blood vessels or brain cells, ready to heal you from within.

Recently, a team of medical mavericks at the Ohio State University introduced a device that does just that. The technology, dubbed tissue nanotransfection (TNT), is set to blow up the field of organ regeneration.

When zapped with a light electrical jolt, the device shoots extra bits of DNA code from its nanotube arrays directly into tiny pores in the skin. There, the DNA triggers the cells to shed their identity and reprograms them into other cell types that can be harvested to repair damaged organs.

Remarkably, the effect spreads with time. The rebooted cells release tiny membrane bubbles onto their neighboring skin cells, coaxing them to undergo transformation. Like zombies, but for good.

So far, the device has already been used to generate neurons to protect the brains of mice with experimental stroke. The team also successfully healed the legs of injured mice by turning the skin cells on their hind limbs into a forest of blood vessels.

While still a ways from human use, scientists believe future iterations of the technology could perform a myriad of medical wonders: repairing damaged organs, relieving brain degeneration, or even restoring aged tissue back to a youthful state.

By using our novel nanochip technology, injured or compromised organs can be replaced. We have shown that skin is a fertile land where we can grow the elements of any organ that is declining, says lead author Dr. Chandan Sen, who published the result in Nature Nanotechnology.

In my lab, we have ongoing research trying to understand the mechanism and do even better, adds Dr. L. James Lee, who co-led the study with Sen. So, this is the beginning, more to come.

The Ohio teams research builds on an age-old idea in regenerative medicine: that even aged bodies have the ability to produce and integrate healthy, youthful cellsgiven the right set of cues.

While some controversy remains on whether replacement cells survive in an injured body, scientistsand some rather dubious clinicsare readily exploring the potential of cell-based therapies.

All cells harbor the same set of DNA; whether they turn into heart cells, neurons, or back into stem cells depend on which genes are activated. The gatekeeper of gene expression is a set of specialized proteins. Scientists can stick the DNA code for these proteins into cells, where they hijack its DNA machinery with orders to produce the protein switchesand the cell transforms into another cell type.

The actual process works like this: scientists harvest mature cells from patients, reprogram them into stem cells inside a Petri dish, inject those cells back into the patients and wait for them to develop into the needed cell types.

Its a cumbersome process packed with landmines. Researchers often use viruses to deliver the genetic payload into cells. In some animal studies, this has led to unwanted mutations and cancer. Its also unclear whether the reprogrammed stem cells survive inside the patients. Whether they actually turn into healthy tissue is even more up for debate.

The Ohio teams device tackles many of these problems head on.

Eschewing the need for viruses, the team manufactured a stamp-sized device out of silicon that serves as a reservoir and injector for DNA. Microetched onto each device are arrays of nanochannels that connect to microscopic dents. Scientists can load DNA material into these tiny holding spots, where they sit stably until a ten-millisecond zap shoots them into the recipients tissue.

We based TNT on a bulk transfection, which is often used in the lab to deliver genes into cells, the authors explain. Like its bulk counterpart, the electrical zap opens up tiny, transient pores on the cell membrane, which allows the DNA instructions to get it.

The problem with bulk transfection is that not all genes get into each cell. Some cells may get more than they bargained for and take up more than one copy, which increases the chance of random mutations.

We found that TNT is extremely focused, with each cell receiving ample DNA, the authors say.

The device also skips an intermediary step in cell conversion: rather than turning cells back into stem cells, the team pushed mouse skin cells directly into other mature cell types using different sets of previously-discovered protein factors.

In one early experiment, the team successfully generated neurons from skin cells that seem indistinguishable from their natural counterparts: they shot off electrical pulses and had similar gene expression profiles.

Surprisingly, the team found that even non-zapped cells in the skins deeper layers transformed. Further testing found that the newly reprogrammed neurons released tiny fatty bubbles that contained the molecular instructions for transformation.

When the team harvested these bubbles and injected them into mice subjected to experimental stroke, the bubbles triggered the brain to generate new neurons and repair itself.

We dont know if the bubbles are somehow transforming other brain cell types into neurons, but they do seem to be loaded with molecules that protect the brain, the researchers say.

In an ultimate test of the devices healing potential, the researchers placed it onto the injured hind leg of a handful of mice. Three days prior, their leg arteries had been experimentally severed, whichwhen left untreatedleads to tissue decay.

The team loaded the device with factors that convert skin cells into blood vessel cells. Within a week of conversion, the team watched as new blood vessels sprouted and grew beyond the local treatment area. In the end, TNT-zapped mice had fewer signs of tissue injury and higher leg muscle metabolism compared to non-treated controls.

This is difficult to imagine, but it is achievable, successfully working about 98 percent of the time, says Sen.

A major draw of the device is that its one-touch-and-go.

There are no expensive cell isolation procedures and no finicky lab manipulations. The conversion happens right on the skin, essentially transforming patients bodies into their own prolific bioreactors.

This process only takes less than a second and is non-invasive, and then youre off. The chip does not stay with you, and the reprogramming of the cell starts,says Sen.

Because the converted cells come directly from the patient, theyre in an immune-privileged position, which reduces the chance of rejection.

This means that in the future, if the technology is used to manufacture organs immune suppression is not necessary, says Sen.

While the team plans to test the device in humans as early as next year, Sen acknowledges that theyll likely run into problems.

For one, because the device needs to be in direct contact with tissue, the skin is the only easily-accessible body part to do these conversions. Repairing deeper tissue would require surgery to insert the device into wounded areas. And to many, growing other organ cell types is a pretty creepy thought, especially because the transformation isnt completely localnon-targeted cells are also reprogrammed.

That could be because the body is trying to heal itself, the authors hypothesize. Using the chip on healthy legs didnt sprout new blood vessels, suggesting that the widespread conversion is because of injury, though (for now) there isnt much evidence supporting the idea.

For another, scientists are still working out the specialized factors required to directly convert between cell types. So far, theyve only had limited success.

But Sen and his team are optimistic.

When these things come out for the first time, its basically crossing the chasm from impossible to possible, he says. We have established feasibility.

Image Credit: Researchers demonstrate tissue nanotransfection,courtesy of The Ohio State University Wexner Medical Center.

Follow this link:
This Chip Uses Electricity to Reprogram Cells for Healing - Singularity Hub

Posted in Ohio Stem Cells | Comments Off on This Chip Uses Electricity to Reprogram Cells for Healing – Singularity Hub

Page 1,410«..1020..1,4091,4101,4111,412..1,4201,430..»