Page 51«..1020..50515253..6070..»

Archive for the ‘Skin Stem Cells’ Category

Pain and Itch in a Dish

Contact Information

Available for logged-in reporters only

Newswise LA JOLLA, CANovember 24, 2014A team led by scientists from The Scripps Research Institute (TSRI) has found a simple method to convert human skin cells into the specialized neurons that detect pain, itch, touch and other bodily sensations. These neurons are also affected by spinal cord injury and involved in Friedreichs ataxia, a devastating and currently incurable neurodegenerative disease that largely strikes children.

The discovery allows this broad class of human neurons and their sensory mechanisms to be studied relatively easily in the laboratory. The induced sensory neurons generated by this method should also be useful in the testing of potential new therapies for pain, itch and related conditions.

Following on the work of TSRI Professor Ardem Patapoutian, who has identified many of the genes that endow these neurons with selective responses to temperature, pain and pressure, we have found a way to produce induced sensory neurons from humans where these genes can be expressed in their normal cellular environment, said Associate Professor Kristin K. Baldwin, an investigator in TSRIs Dorris Neuroscience Center. This method is rapid, robust and scalable. Therefore we hope that these induced sensory neurons will allow our group and others to identify new compounds that block pain and itch and to better understand and treat neurodegenerative disease and spinal cord injury.

The report by Baldwins team appears as an advance online publication in Nature Neuroscience on November 24, 2014.

In Search of a Better Model

The neurons that can be made with the new technique normally reside in clusters called dorsal root ganglia (DRG) along the outer spine. DRG sensory neurons extend their nerve fibers into the skin, muscle and joints all over the body, where they variously detect gentle touch, painful touch, heat, cold, wounds and inflammation, itch-inducing substances, chemical irritants, vibrations, the fullness of the bladder and colon, and even information about how the body and its limbs are positioned. Recently these neurons have also been linked to aging and to autoimmune disease.

Because of the difficulties involved in harvesting and culturing adult human neurons, most research on DRG neurons has been done in mice. But mice are of limited use in understanding the human version of this broad somatosensory system.

Mouse models dont represent the full diversity of the human response, said Joel W. Blanchard, a PhD candidate in the Baldwin laboratory who was co-lead author of the study with Research Associate Kevin T. Eade.

See more here:
Pain and Itch in a Dish

Pain-Sensing Neurons Created From Human, Mouse Skin Cells

November 25, 2014

Chuck Bednar for redOrbit.com Your Universe Online

Two different teams of researchers, one led by scientists from The Scripps Research Institute (TSRI) and the other involving members of the Harvard Stem Cell Institute (HSCI) have discovered ways to create the neurons that detect pain, itch and other sensations in laboratory conditions out of human and mouse skin cells.

The TSRI study, which was published online Monday in the journal Nature Neuroscience, used what the authors referred to as a simple technique to create neurons that normally reside in clusters called dorsal root ganglia (DRG) along the outer spine. Those neurons are often affected by spinal cord injuries and a neurodegenerative condition known as Friedreichs ataxia.

According to the researchers, DRG sensory neurons extend their nerve fibers into skin, muscle and joints located throughout the body. The neurons are capable of alternately detecting gentle touch, painful contact, heat, cold, wounds, inflammation, chemical irritants, itch-inducing agents and fullness of the bowels and bladder. They also relay information about the position of the body and limbs, and have been linked to aging and autoimmune disease.

Due to the difficulties involved in culturing adult human neurons, most research relating to DRG neurons has been done in mice. However, the rodents are of limited use in understanding the human version of this somatosensory system, TSRI explained. The new discovery will allow this type of human neurons and their associated sensory mechanisms to be studied with relative ease in laboratory conditions, according to the study authors.

We have found a way to produce induced sensory neurons from humans where these genes can be expressed in their normal cellular environment, associate professor Kristin K. Baldwin, an investigator in TSRIs Dorris Neuroscience Center, said in a statement. This method is rapid, robust and scalable. Therefore we hope that these induced sensory neurons will allow our group and others to identify new compounds that block pain and itch and to better understand and treat neurodegenerative disease and spinal cord injury.

Similarly, the HSCI-led study, which included experts from Boston Childrens Hospital (BCH) and Harvards Department of Stem Cell and Regenerative Biology (HSCRB), was able to successfully convert mouse and human skin cells into pain-sensing neurons that responded to several different types of stimuli responsible for causing both acute and inflammatory pain.

The authors of this study, which also appeared in Wednesdays online edition of Nature Neuroscience, said that their research could help scientists better understand the different types of pain that we experience, as well as better identify why people respond to pain in different ways and why some individuals are more or less likely to develop chronic pain. It could also result in the development of improved pain-relieving medications.

The six-year project resulted in the creation of neuronal pain receptors that respond to both the types of intense stimuli triggered by a physical injury, and the more subtle stimuli triggered by inflammation which results in pain tenderness. The researchers report that the fact the neurons can respond to both the gross and fine forms of stimulation which produce separate types of pain in humans confirm that they are functionally normal.

Read the original post:
Pain-Sensing Neurons Created From Human, Mouse Skin Cells

Stem Cells for Skin Tissue Engineering and Wound Healing

1. Keratinocytes

There are two approaches to commit ES cells and adult stem cells (of non-epidermal origin) to the keratinocyte lineage in vitro. One approach would be to expose the cells to a cocktail of exogenous cytokines, growth factors, chemicals, and extracellular matrix (ECM) substrata over a prolonged duration of in vitro culture. Only a fraction of the stem cells would be expected to undergo commitment to the keratinocyte lineage, because many of these cytokines, growth factors, chemicals, and ECM substrata would exert non-specific pleitropic effects on stem cell differentiation into multiple lineages. At best, the cocktail combination of various cytokines, growth factors, chemicals, and ECM substrata can be optimized by trial and error, to maximize the proportion of stem cells committing to the keratinocyte lineage, while at the same time yielding a large number of other undesired lineages. Hence, extensive selection/purification and proliferation of the commited keratinocyte progenitors is likely to be required.

By using such an approach, Coraux et al.54 managed to achieve commitment and subsequent differentiation of murine ES cells into the keratinocyte lineage, in the presence of a cocktail combination of bone morphogenetic protein-4 (BMP-4), ascorbate, and ECM derived from human normal fibroblasts (HNFs) and murine NIH-3T3 fibroblasts. Nevertheless, it must be noted that the study of Coraux et al.54 also reported a high degree (approximately 80%) of non-specific differentiation into multiple uncharacterized lineages, and no attempt was made to purify differentiated keratinocytes or keratinocyte progenitors from the mixture of lineages derived from murine ES cells. Bagutti et al.61 reported that coculture with human dermal fibroblasts (HDFs) as well as HDF-conditioned media could induce beta integrin- deficient murine ES cells to commit and differentiate into the keratinocyte lineage. However, as with the study of Coraux et al.,54 the keratinocytes were interspersed with differentiated cells of other lineages. Recently, differentiation of human ES cells into the keratinocyte lineage was also reported by Green et al.62 However, this study was based on in vivo teratoma formation within a SCID mouse model, and to date, there are no parallel in vitro studies that have been reported.

With adult stem cells of non-epidermal origin, there are also few studies 63, 64 which have successfully achieved re-commitment and trans-differentiation to the keratinocyte lineage. Even so, these studies were based primarily on the transplantation of undifferentiated stem cells in vivo, with the observed trans-differentiation occurring sporadically and at extremely low frequencies. Moreover, the validity of the experimental data may be clouded by controversy over the artifact of stem cell fusion in vivo.65 To date, there are no parallel in vitro studies that have achieved recommitment and trans-differentiation of non-epidermal adult stem cells to the keratinocyte lineage. It can therefore be surmised that the use of exogenous cytokines, growth factors, chemicals, and ECM substrata to induce ES cell and nonepidermal adult stem cell commitment to the keratinocyte lineage is a relatively inefficient, time-consuming, and labor-intensive process that would require extensive selection and purification of the committed keratinocyte progenitors. Hence, it would be technically challenging to apply this to the clinical situation.

The other approach for inducing ES cell and non-epidermal adult stem cell commitment to the keratinocyte lineage is through genetic modulation. This may be achieved by transfecting stem cells with recombinant DNA constructs encoding for the expression of signaling proteins that promote commitment to the keratinocyte lineage. Of particular interest are the Lef-1/Tcf family of Wnt regulated transcription factors that act in concert with b-catenin,66, 67 c-myc which is a downstream target of the Wnt-signaling pathway,68, 69 and the transactivation domain containing isoform of transcription factor p63 (Tap63).70, 71 Interestingly, the transcription factor GATA-3, which is well known to be a key regulator of T-cell lineage determination, has also been shown to be essential for stem cell lineage determination in skin, where it is expressed at the onset of epidermal stratification and Inner Root Sheath (IRS) specification in follicles.72 Recombinant overexpression of p6373 and c-Myc74 has been reported to promote commitment and differentiation to the keratinocyte lineage.

The disadvantage of directing differentiation through genetic modulation is the potential risks associated with utilizing recombinant DNA technology in human clinical therapy. For example, the overexpression of any one particular protein within transfected stem cells would certainly have unpredictable physiological effects upon transplantation in vivo. This problem may be overcome by placing the recombinant expression of the particular protein under the control of switchable promoters, several of which have been developed for expression in eukaryotic systems. Such switchable promoters could be responsive to exogenous chemicals,75 heat shock,76 or even light.77 Genetically modified stem cells may also run the risk of becoming malignant within the transplanted recipient. Moreover, there are overriding safety concerns with regard to the use of recombinant viral based vectors in the genetic manipulation of stem cells.78 It remains uncertain as to whether legislation would ultimately permit the use of genetically modified stem cells for human clinical therapy. At present, the potential detrimental effects of transplanting genetically modified stem cells in vivo are not well studied. More research needs to be carried out on animal models to address the safety aspects of such an approach.

More recently, there is emerging evidence that some transcription factors (which are commonly thought of as cytosolic proteins) have the ability to function as paracrine cell to cell signaling molecules.79 This is based on intercellular transfer of transcription factors through atypical secretion and internalization pathways.79 Hence, there is an exciting possibility that transcription factors implicated in commitment to the keratinocyte lineage may in the future be genetically engineered to incorporate domains that enable them to participate in novel paracrine signaling mechanisms. This in turn would have tremendous potential for inducing the commitment of ES cells and non-epidermal adult stem cells to the keratinocyte lineage.

Skin appendages, including hair follicles, sebaceous glands and sweat glands, are linked to the epidermis but project deep into the dermal layer. The skin epidermis and its appendages provide a protective barrier that is impermeable to harmful microbes and also prevents dehydration. To perform their functions while being confronted with the physicochemical traumas of the environment, these tissues undergo continual rejuvenation through homeostasis, and, in addition, they must be primed to undergo wound repair in response to injury. The skins elixir for maintaining tissue homeostasis, regenerating hair, and repairing the epidermis after injury is its stem cells.

The hair follicle is composed of an outer root sheath that is contiguous with the epidermis, an inner root sheath and the hair shaft. The matrix surrounding the dermal papilla, in the hair root, contains actively dividing, relatively undifferentiated cells and is therefore a pocket of MSCs that are essential for follicle formation. The lower segment of each hair follicle cycles through periods of active growth (anagen), destruction (catagen) and quiescence (telogen).80 A specialized region of the outer root sheath of the hair follicle, known as the bulge, is located below the sebaceous gland, which is also the attachment site of the arrector pili muscle, receiving inputs from sensory nerve endings and blood vessels. Furthermore, the hair follicle bulge is a reservoir of slow-cycling multipotent stem cells.81, 82 Subsets of these follicle-derived multipotent stem cells can be activated and migrate out of hair follicles to the site of a wound to repair the damaged epithelium; however, they contribute little to the intact epidermis. These hair follicle stem cells can also contribute to the growth of follicles themselves and the sebaceous gland. For example, in the absence of hair follicle stem cells, hair follicle and sebaceous gland morphogenesis is blocked, and epidermal wound repair is compromised.83 In addition to containing follicle epidermal stem cells, the bulge contains melanocyte stem cells.84 Recent studies show that nestin, a marker for neural progenitor cells, is selectively expressed in cells of the hair follicle bulge and that these stem cells can differentiate into neurons,85 glia, keratinocytes, smooth muscle cells, melanocytes and even blood vessels.86, 87 Examination of close developmental and anatomical parallels between epithelial tissue and dermal tissue in skin and hair follicles has revealed dermal tissue to have stem cells. Paus et al. indicated that hair follicle dermal sheath cells might represent a source of dermal stem cells that not only incorporate into the hair-supporting papilla, low down in the follicle, but also move up and out from the follicle dermal sheath into the dermis of adjoining skin.88 Hair follicle dermal sheath cells taken from the human scalp can form new dermal papilla, induce the formation of hair follicles, and produce hair shafts when transplanted onto skin.89 There is also a clear transition from dermal sheath to dermal papilla cells.90 When the follicle dermal cells are implanted into skin wounds, they can be incorporated into the new dermis in a manner similar to that of skin wound-healing fibroblasts.91 However, these cell populations still lack specific markers for purifying and distinguishing the stem cells from their progeny. Furthermore, of prime importance is improving our understanding of the relation between bulge cells and interfollicular epidermal stem cells and between bulge cells and other stem cells inhabiting the skin and the mechanisms of hair growth.

Recently, cell replacement therapy has offered a novel and powerful medical technology for skin repair and regeneration: a new population of stem cell, called a neural crest stem cell, from adult hair follicles, was discovered to have the ability to differentiate in vitro to keratinocytes, neurons, cartilage/bone cells, smooth muscle cells, melanocytes, glial cells, and adipocytes.9296 In mammalian skin, skin-derived neural progenitors were isolated and expanded from the dermis of rodent skin and adult human scalp and could differentiate into both neural and mesodermal progeny.97, 98 Skin-derived neural progenitor cells were isolated based on the sphere formation of floating cells after 37 days of culture in uncoated flasks with epidermal growth factor and fibroblast growth factor, and characterized by the production of nestin and fibronectin, markers of neural precursors. In addition, skin-derived neural progenitor cells were identified as neural crest derived by the use of Wnt1 promoter driving LacZ expression in the mouse. Some of the LacZ-positive cells were found in the skin of the face, as well as in the dermis and dermal papilla of murine whisker.99 These skin derived neural crest cells have already shown promising results in regenerative medicine such as the promotion of regenerative axonal growth after transplantation into injured adult mouse sciatic nerves 95 or spinal cord repair,100 resulting in the recovery of peripheral nerve function. This new study marks an important first step in the development of real stem-cell-based therapies and skin tissue regeneration.

Read more here:
Stem Cells for Skin Tissue Engineering and Wound Healing

Platelet-like nanoparticles improve on nature to stem the blood flow

The skin is the body's first line of defense against infection. And when this barrier is broken, or an internal organ is ruptured, it is the process of coagulation, or clotting, which relies largely on blood cells called platelets, that seals the breach and stems the flow of blood. Researchers at UC Santa Barbara (UCSB) have now synthesized nanoparticles that mimic the form and function of platelets, but can do more than just accelerate the body's natural healing processes.

Platelets are nucleus-free blood cells that are essentially the building blocks for any blood clot, binding together at the edge of a wound as well as changing shape and secreting chemical messengers to call more platelets to the scene of the injury to assist in the healing process. However, coagulation can be hampered if an injury is too severe, or if the injured person is taking anti-coagulation medication or suffers from a platelet disorder.

In such cases, the platelet-like nanoparticles (PLNs) synthesized at UCSB could save the day. Developed by researchers in UCSB's Department of Chemical Engineering and its Center for Bioengineering, the synthetic platelets behave just like their naturally-occurring counterparts, mimicking their shape, flexibility and surface biology.

In this way, their creators say they could be added to a patient's own natural blood supply or augment the patient's own platelet supply and accelerate the healing process for both internal and external injuries. Not only will the PLNs congregate at the site of an injury, but like natural platelets, they'll also call other platelets to the site and bind to them. Then, once their task has been completed and the wound has been plugged, the PLNs will dissolve into the blood.

Furthermore, the researchers claim to have improved on nature, with their nanoscale synthetic platelets outperforming micron-sized natural ones in tests. The synthetic platelets are also able to be customized with medications that match the needs of patients with a specific condition. They could also be used to carry antibiotics to certain parts of the body, such as across the blood-brain barrier, to provide targeted therapy and improved diagnostics.

"This technology could address a plethora of clinical challenges," said Dr. Scott Hammond, director of UCSBs Translational Medicine Research Laboratories. "One of the biggest challenges in clinical medicine right now which also costs a lot of money is that were living longer and people are more likely to end up on blood thinners." The researchers say PLNs would help in the treatment of such elderly patients, allowing the PLNs to be targeted at the site of an injury without triggering unwanted bleeding.

Importantly, the researchers say the synthetic platelets have a longer shelf life and are cheaper relatively speaking than another person's platelets both important factors at times of emergencies and natural disasters when blood products are in highest demand.

These aren't the first synthetic platelets weve seen that improve on nature. In 2009, researchers at Case Western University reported the development of artificial platelets made of biodegradable polymers that reduced clotting times in animal models and attracted the interest of the military.

The team will now look at how well the production of the PLNs scales up and asses practical clinical concerns, such as manufacturing, storage, sterility ahead of pre-clinical and clinical testing.

Results of the researchers current findings appear in the journal ACS Nano.

Excerpt from:
Platelet-like nanoparticles improve on nature to stem the blood flow

Nail stem cells prove more versatile than press ons

PUBLIC RELEASE DATE:

20-Nov-2014

Contact: Cristy Lytal lytal@med.usc.edu 323-442-2172 University of Southern California - Health Sciences

There are plenty of body parts that don't grow back when you lose them. Nails are an exception, and a new study published in the Proceedings of the National Academy of Sciences (PNAS) reveals some of the reasons why.

A team of USC Stem Cell researchers led by principal investigator Krzysztof Kobielak and co-first authors Yvonne Leung and Eve Kandyba has identified a new population of nail stem cells, which have the ability to either self-renew or undergo specialization or differentiation into multiple tissues.

To find these elusive stem cells, the team used a sophisticated system to attach fluorescent proteins and other visible "labels" to mouse nail cells. Many of these cells repeatedly divided, diluting the fluorescence and labels among their increasingly dim progeny. However, a few cells located in the soft tissue attached to the base of the nail retained strong fluorescence and labels because they either did not divide or divided slowly -- a known property of many stem cells.

The researchers then discovered that these slow-dividing stem cells have the flexibility to perform dual roles. Under normal circumstances, the stem cells contribute to the growth of both the nails and the adjacent skin. However, if the nail is injured or lost, a protein called "Bone Morphogenic Protein," or BMP, signals to the stem cells to shift their function exclusively to nail repair.

The researchers are now wondering whether or not the right signals or environmental cues could induce these nail stem cells to generate additional types of tissue -- potentially aiding in the repair of everything from nail and finger defects to severe skin injuries and amputations.

"That was very surprising discovery, since the dual characteristic of these nail stem cells to regenerate both the nail and skin under certain physiological conditions is quite unique and different from other skin stem cells, such as those of the hair follicle or sweat gland," said Kobielak.

###

Read the original post:
Nail stem cells prove more versatile than press ons

Health Beat: Stem cells to repair broken chromosomes

CLEVELAND -

Our bodies contain 23 pairs of them, 46 total, but if chromosomes are damaged, they can cause birth defects, disabilities, growth problems, even death.

Case Western Reserve University scientist Anthony Wynshaw-Boris is studying how to repair damaged chromosomes with the help of a recent discovery. He's taking skin cells and reprogramming them to work like embryonic stem cells, which can grow into different cell types.

"You're taking adult or a child's skin cells. You're not causing any loss of an embryo, and you're taking those skin cells to make a stem cell," said Wynshaw-Boris.

Scientists studied patients with a specific defective chromosome that was shaped like a ring. They took the patients' skin cells and reprogrammed them into embryonic-like cells in the lab. They found this process caused the damaged "ring" chromosomes to be replaced by normal chromosomes.

"It at least raises the possibility that ring chromosomes will be lost in stem cells," said Wynshaw-Boris.

While this research was only conducted in lab cultures on the rare ring-shaped chromosomes, scientists hope it will work in patients with common abnormalities like Down syndrome.

"What we're hoping happens is we might be able to use, modify, what we did, to rescue cell lines from any patient that has any severe chromosome defect," Wynshaw-Boris explained.

It's research that could one day repair faulty chromosomes and stop genetic diseases in their tracks.

The reprogramming technique that transforms skin cells to stem cells was so groundbreaking that a Japanese physician won the Nobel Prize in medicine in 2012 for developing it.

See the rest here:
Health Beat: Stem cells to repair broken chromosomes

Fat and Bone Marrow-Derived Stem Cells Combo Shows Promise in Preventing Transplant Rejection

Durham, NC (PRWEB) November 20, 2014

With more soldiers returning from combat suffering devastating injuries, doctors are turning to a reconstructive surgery that uses tissue transplantation along with immuno-suppression therapy. This approach has had encouraging results; however, rejection of transplanted tissue from an unmatched donor remains a critical complication. A new study found in the latest issue of STEM CELLS Translational Medicine reports that researchers may have found a way around that.

We demonstrated in mice that a single infusion of adipose-derived stromal cells (ASC) which are stem cells taken from fat in a minimally invasive procedure from an unmatched donor combined with an extremely low dose of bone marrow cells resulted in stable long-term tolerance of the skin graft without undo consequences such as graft versus host disease, said Thomas Davis, Ph.D., a contractor from the Henry M. Jackson Foundation who is working at the Naval Medical Research Centers Regenerative Medicine Department. Dr. Davis is lead author of the study.

He added, As we move forward, we are cautiously optimistic, appreciating that the transition from these laboratory models to proof-of-principle preclinical studies is challenging and not straightforward. If successful, the technology has diverse therapeutic applications in clinical transplantation in both military and civilian settings.

The research team wanted to try using ASCs because these cells have proven to be more potent than bone marrow and cord-blood derived stem cells when it comes to inhibiting the bodys rejection of transplantations from an unmatched donor. They conducted the study by doing skin grafts in mice. One group of grafted mice received no stem cell transfusions; one group received human-derived ASCs after the graft occurred; and another group received a combination of human ASCs and stem cells harvested from the mouses own bone marrow, also after placement of the graft.

More than 200 days later, the combination of human ASC and limited numbers of blood marrow stem cells effectively prevented rejection, with no evidence of graft versus host disease, Dr. Davis reported.

Navy Capt. Eric A. Elster, M.D., professor and chair of the surgery department at Uniformed Services University of the Health Sciences, helped lead the study. ASC constitutively produced high levels of anti-inflammatory/immunoregulatory factors, he said. While further work is needed to validate this approach in other laboratory models before clinical trials can begin, the ability to use ASC, which are non-donor specific and clinically feasible, to induce tolerance opens a new horizon in transplantation.

The implications of this research are broad, said Anthony Atala, MD, editor of STEM CELLS Translational Medicine and director of the Wake Forest Institute for Regenerative Medicine. If these findings are duplicated in additional models and in human trials, there is potential to apply this strategy to many areas of transplantation.

###

This article, Adipose-derived Stromal Cells Promote Allograft Tolerance Induction, and more can be accessed at http://www.stemcellsTM.com.

Go here to see the original:
Fat and Bone Marrow-Derived Stem Cells Combo Shows Promise in Preventing Transplant Rejection

Mount Sinai researchers awarded grant to find new stem cell therapies for vision recovery

PUBLIC RELEASE DATE:

20-Nov-2014

Contact: Jessica Mikulski jmikulski@nyee.edu 212-979-4274 The Mount Sinai Hospital / Mount Sinai School of Medicine @mountsinainyc

The National Eye Institute (NEI), a division of the National Institutes of Health, has awarded researchers at the Icahn School of Medicine at Mount Sinai a five-year grant totaling $1 million that will support an effort to re-create a patients' ocular stem cells and restore vision in those blinded by corneal disease.

About six million people worldwide have been blinded by burns, trauma, infection, genetic diseases, and chronic inflammation that result in corneal stem cell death and corneal scarring.

There are currently no treatments for related vision loss that are effective over the long term. Corneal stem cell transplantation is an option in the short term, but availability of donor corneas is limited, and patients must take medications that suppress their immune systems for the rest of their lives to prevent rejection of the transplanted tissue.

A newer proposed treatment option is the replacement of corneal stem cells to restore vision. The grant from the NEI will fund Mount Sinai research to re-create a patient's own stem cells and restore vision in those blinded by corneal disease. Technological advances in recent years have enabled researchers to take mature cells, in this case eyelid or oral skin cells, and coax them backward along the development pathways to become stem cells again. These eye-specific stem cells would then be redirected down pathways that become needed replacements for damaged cells in the cornea, in theory restoring vision.

"Our findings will allow the creation of transplantable eye tissue that can restore the ocular surface," said Albert Y. Wu, MD, PhD, Assistant Professor, Department of Ophthalmology at the Icahn School of Medicine at Mount Sinai and principle investigator for the grant-funded effort. "In the future, we will be able to re-create a patient's own corneal stem cells to restore vision after being blind," added Dr. Wu, also Director of the Ophthalmic Plastic and Reconstructive Surgery, Stem Cell and Regenerative Medicine Laboratory in the Department of Ophthalmology and a member of the Black Family Stem Cell Institute at Icahn School of Medicine. "Since the stem cells are their own, patient's will not require immunosuppressive drugs, which would greatly improve their quality of life."

Specifically, the grant will support efforts to discover new stem cell therapies for ocular surface disease and make regenerative medicine a reality for people who have lost their vision. The research team will investigate the most viable stem cell sources, seek to create ocular stem cells from eyelid or oral skin cells, explore the molecular pathways involved in ocular and orbital development, and develop cutting-edge biomaterials to engraft a patient's own stem cells and restore vision.

###

See the rest here:
Mount Sinai researchers awarded grant to find new stem cell therapies for vision recovery

Mount Sinai Researchers Awarded $1 Million Grant to Find New Stem Cell Therapies for Vision Recovery

Contact Information

Available for logged-in reporters only

Newswise NEW YORK November 20, 2014 The National Eye Institute (NEI), a division of the National Institutes of Health, has awarded researchers at the Icahn School of Medicine at Mount Sinai a five-year grant that will support an effort to re-create a patients ocular stem cells and restore vision in those blinded by corneal disease.

About six million people worldwide have been blinded by burns, trauma, infection, genetic diseases, and chronic inflammation that result in corneal stem cell death and corneal scarring. There are currently no treatments for related vision loss that are effective over the long term. Corneal stem cell transplantation is an option in the short term, but availability of donor corneas is limited, and patients must take medications that suppress their immune systems for the rest of their lives to prevent rejection of the transplanted tissue.

A newer proposed treatment option is the replacement of corneal stem cells to restore vision. The grant from the NEI will fund Mount Sinai research to re-create a patients own stem cells and restore vision in those blinded by corneal disease. Technological advances in recent years have enabled researchers to take mature cells, in this case eyelid or oral skin cells, and coax them backward along the development pathways to become stem cells again. These eye-specific stem cells would then be redirected down pathways that become needed replacements for damaged cells in the cornea, in theory restoring vision.

Our findings will allow the creation of transplantable eye tissue that can restore the ocular surface, said Albert Y. Wu, MD, PhD, Assistant Professor, Department of Ophthalmology at the Icahn School of Medicine at Mount Sinai and principle investigator for the grant-funded effort. In the future, we will be able to re-create a patients own corneal stem cells to restore vision after being blind, added Dr. Wu, also Director of the Ophthalmic Plastic and Reconstructive Surgery, Stem Cell and Regenerative Medicine Laboratory in the Department of Ophthalmology and a member of the Black Family Stem Cell Institute at Icahn School of Medicine. Since the stem cells are their own, patients will not require immunosuppressive drugs, which would greatly improve their quality of life.

Specifically, the grant will support efforts to discover new stem cell therapies for ocular surface disease and make regenerative medicine a reality for people who have lost their vision. The research team will investigate the most viable stem cell sources, seek to create ocular stem cells from eyelid or oral skin cells, explore the molecular pathways involved in ocular and orbital development, and develop cutting-edge biomaterials to engraft a patients own stem cells and restore vision.

Other investigators from Mount Sinai include Ihor Lemischka, PhD, Director, Black Family Stem Cell Institute and J. Mario Wolosin, PhD, Professor of Ophthalmology. The research is supported by NEI grant EY023997.

###

View original post here:
Mount Sinai Researchers Awarded $1 Million Grant to Find New Stem Cell Therapies for Vision Recovery

Researchers Convert Skin Cells To Replace HD-Damaged Brain Cells

By Estel Grace Masangkay

A team of researchers at the Washington University School of Medicine in St. Louis reported that they have discovered a way to directly convert human skin cells into a type of brain cell that has been damaged by Huntingtons disease.

The team chose to produce a certain type of brain cell known as medium spiny neurons, which play a key part in controlling movement. Medium spiny neurons are the cells most affected by Huntingtons disease, a neurodegenerative disorder characterized by involuntary muscle movements and cognitive decline. The disease symptoms typically begin showing in mid-adulthood, and they steadily worsen over time.

For their experiment, the scientists used adult human skin cells instead of the typical mouse cells or embryonic human cells. The team placed the skin cells in an environment similar to the environment of brain cells and then exposed them to two small molecules of RNA named miR-9 and miR-124. In their past research, the scientists have discovered that these microRNAs turn skin cells into a mix of various neuron types. Dr. Yoo and his colleagues fine-tuned the chemical signals by further exposing the cells to transcription factors they knew are found in the part of the brain where medium spiny neurons thrive. Results show that the converted cells survived for at least six months after they were injected into mices brains. The cells also behaved in a similar fashion to native brain cells.

Not only did these transplanted cells survive in the mouse brain, they showed functional properties similar to those of native cells. These cells are known to extend projections into certain brain regions. And we found the human transplanted cells also connected to these distant targets in the mouse brain. That's a landmark point about this paper, said Dr. Andrew S. Yoo, assistant professor of developmental biology in Washington University School of Medicine and senior author of the study.

The new process differs from other techniques in that it does not need to undergo a stem cell phase, thereby avoiding production of multiple cell types. The scientists added that using adult human cells offers the opportunity to use the patients own cells in future procedures, which would radically minimize the risk of rejection by the patients immune system. Dr. Yoos team is now preparing to test skin cells taken from patients with Huntingtons disease using the approach. They also intend to inject healthy reprogrammed human cells into mice models of Huntingtons disease to check whether these have any effect on the diseases symptoms.

The researchers work was published in the previous months issue of the journal Neuron.

Read more:
Researchers Convert Skin Cells To Replace HD-Damaged Brain Cells

NR Skin Launches Anti-Aging Product Line

Woodland Hills, CA (PRWEB) November 19, 2014

Longtime skincare industry professional Nancy Ryan announces the launch of NR Skin, featuring a line of efficacious products that deliver various skin rejuvenation and age repair benefits for all skin types.

According to Dr. Lisa Benest, Board-certified dermatologist, Burbank, CA, the NR Skin line offers a range of daily skincare and skin rejuvenation products distinguished by high concentrations of powerhouse ingredients that are known for their anti-aging properties, such as antioxidant vitamins and minerals, plant stem cells, lipids, as well as peptides. Dr. Benest notes that NR Skin products offer pure, clean ingredients that feel great on the skin and deliver visible results.

Backed by more than 20 years of skincare industry experience and expertise, NR Skin Founder and CEO Nancy Ryan comments, the creation of NR Skin is a culmination of my lifes work and lifelong passion for excellence in skincare. Im thrilled to help people improve their quality of life by achieving healthy, beautiful skin through such pure and effective products.

Before establishing NR Skin in 2014, Ms. Ryan led Pro-Med Consulting, Inc. for 21 years, which was built upon the core mission of giving dermatologists, plastic surgeons and medical spas a viable way to build their own brand equity and expand their businesses with private label, medical-grade skin care products. Over the years, she developed numerous relationships with leading physicians, whose businesses grew significantly by offering patients her high-performance products that bore each doctors name.

Prior to this successful venture, she worked for two pioneering skin care companies, Ortho Dermatologics, (makers of Retin-A Micro/Renova) and NeoStrata, where she had the opportunity to learn about skin care chemistry and the most effective ways to treat various skin conditions with specific product ingredients.

The NR Skin product line consists of: the following clinically tested products: Age-defying Peptide Cream; Citrus Stem Cell Fusion Cream, Neuro-Peptide Serum. Retinol Complex Treatment Super Antioxidant Cream, Super C Serum Treatment, Comfort Cleanser, Lash Teez Eyelash Growth Serum and Sunscreen Lotion SPF30.

To view products and recommended regimens, visit: http://www.nrskin.com Follow us on Facebook: http://www.facebook.com/nrskin and on Twitter: @nrskincompany

Visit link:
NR Skin Launches Anti-Aging Product Line

Bubble Boy Disease Cured With Stem Cells

TIME Health medicine Bubble Boy Disease Cured With Stem Cells Alysia Padilla-Vacarro and daughter Evangelina on the day of her gene therapy treatment. Evangelina, now two years old, has had her immune system restored and lives a healthy and normal life. Courtesy of UCLA Researchers have treated more than two dozen patients with a treatment made from their own bone marrow cells

Alysia Padilla-Vaccaro and Christian Vaccaro owe their daughters life to stem cells. Evangelina, now two, is alive today because she saved herself with her own bone marrow cells.

Evangelina, a twin, was born with a severe immune disorder caused by a genetic aberration that makes her vulnerable to any and all bacteria and viruses; even a simple cold could be fatal. But doctors at University of California Los Angeles (UCLA) Broad Stem Cell Research Center gave her a new treatment, using her own stem cells, that has essentially cured her disease. Shes one of 18 children who have been treated with the cutting-edge therapy, and the studys leader, Dr. Donald Kohn, says that the strategy could also be used to treat other gene-based disorders such as sickle cell anemia.

Known to doctors as adenosine deaminase (ADA)-deficient severe combined immunodeficiency (SCID), its better known as bubble boy disease, since children born with the genetic disorder have immune systems so weak that they need to stay in relatively clean and germ-free environments. Until Evangelina and her sister Annabella were 11 months old, We were gowned and masked and did not go outside, says their mother Alysia Padilla-Vaccaro. Our children did not physically see our mouths until then because we were masked all the time. We couldnt take them outside to take a breath of fresh air, because there is fungus in the air, and that could kill her.

Both parents wore masks at work to lower the chances they would be exposed to germs that they might bring back home. And they took showers and changed clothes as soon as they entered the house.

MORE: Gene-Therapy Trial Shows Promise Fighting Bubble Boy Syndrome

SCID is caused by a genetic mutation in the ADA gene, which normally produces the white blood cells that are the front lines of the bodys defense against bacteria and viruses. The Vaccaros decided to treat Annabella in the same way that they cared for Evangelina; They were crawling and playing with each other, and every toy they sucked on, they stuck in each others hands and each others mouth, so we couldnt take one outside to have a grand old time and potentially bring something back that could harm her sister, says Padilla-Vaccaro.

The only treatments for SCID are bone marrow transplants from healthy people, ideally a matched sibling; the unaffected cells can then repopulate the immune system of the baby with SCID. But despite being her twin, Annabella wasnt a blood match for her sister, nor were her parents. Padilla-Vaccaro and her husband, Christian, were considering unrelated donors but were concerned about the risk of rejection. We would be trying to fix one problem and getting another, she says.

MORE: Stem Cells Allow Nearly Blind Patients to See

Thats when the doctors at the Childrens Hospital at Orange County, where Evangelina was diagnosed, told her parents about a stem cell trial for SCID babies at UCLA, led by Dr. Donald Kohn. As soon as they said trial, I thought, my kid is dead, says Padilla-Vaccaro of the last resort option. But a dozen children born with other forms of SCIDin which different mutations caused the same weak immune systemswho were successfully treated by Kohn convinced the couple that the therapy was worth trying. Kohn had one spot left in the trial and was willing to hold it for Evangelina until she matured more. Born premature, she was diagnosed at six weeks old and needed more time for what was left of her immune system to catch up to weather the procedure.

Follow this link:
Bubble Boy Disease Cured With Stem Cells

What is a Stem Cell Support Serum? | RG Cell | Agerite Solutions – Video


What is a Stem Cell Support Serum? | RG Cell | Agerite Solutions
What is a Stem Cell Support Serum? Paloma: And I suppose my next question would be what is a stem cell support serum? Dean: Well, in skin care, serums are co...

By: BusinessTrendsTV

Visit link:
What is a Stem Cell Support Serum? | RG Cell | Agerite Solutions - Video

Anti Aging Stem Cell Serums Renew Skin – Life Line Skin Care

Stem cells are the building blocks of your skin. They have a unique ability to replace damaged and diseased cells. As they divide, they can proliferate for long periods into millions of new skin cells.

As we age, our stem cells lose their potency. Your skin's ability to repair itself just isn't what it used to be. The result can be fine lines, wrinkles, age spots, and sagging skin. But non-embryonic stem cells -- the same stem cells active early in life -- are highly potent. Lifeline anti-aging stem cell serums tap into the potency of these stem cells to help renew your skin.

Scientists at Lifeline Skin Care discovered that human non-embryonic stem cell extracts can help renew skin -- by replacing old cells with healthy new ones. These stem cell extracts help stimulate your own skin's abilities to repair itself. And Lifeline anti-aging stem cell serums were born.

Where Stem Cells in Anti Aging Products Come From

The first types of human stem cells to be studied by researchers were embryonic stem cells, donated from in vitro fertilization labs. But because creating embryonic stem cells involves the destruction of a fertilized human embryo, many people have ethical concerns about the use of such cells.

Lifeline Skin Care (through its parent company, ISCO) is the first company in the world to discover how to create human non-embryonic stem cells -- and how to take extracts from them. As a result, you need never be concerned that a viable human embryo was damaged or destroyed to create these anti-aging products.

The non-embryonic stem cells in Lifeline stem cell serums are derived from unfertilized human oocytes (eggs) which are donated to ISCO from in vitro fertilization labs and clinics.

Lifeline Anti Aging Stem Cell Serums are Based in Science

Lifeline Skin Care's exclusive anti-aging products are a combination of several discoveries and unique high-technology, patent-pending formulations.

See the original post here:
Anti Aging Stem Cell Serums Renew Skin - Life Line Skin Care

A mature approach

AP Glowing: Model Kate Moss

For someone who has mature skin, it is annoying watching 20-somethings with blemish-less complexions worrying about wrinkles and crowfeet. No one seems to be concerned about those over 40 who, for the better part of their 20s and 30s, were busy cooking, cleaning and washing up instead of pandering to their skin.

So when Page 3 Salon, in Race Course, issued an invitation to try its new award-winning skin therapy for mature skin all the way from Spain, one jumped at it. It is supposed to be the same treatment celebrities such as Elton John, Penelope Cruz, Jemima Khan and Kate Moss use to keep their skin glowing and young.

If you have mature skin, this treatment is the one for you, says a spokesperson for the brand Skeyndor (meaning golden skin), who was in Coimbatore recently to promote the product and train the beauty therapists at Page 3 the correct way to use the products. Skeyndor has over 200 products to suit other skin types, too. R&D is the companys strength and it is also one of the first in the cosmetic industry to use nano-technology for skin care.

She says, comfortingly, that my skin is still not too bad and sits me down to explain what I can do to keep it from aging too fast.

The special facial that is recommended for me uses products that are gentle on the skin and one particular treatment that has the same effect as Botox. And, without being invasive at all. Before the facial commences a photograph is taken focussing on the problem areas of the skin. And once the hour-plus, soothing treatment is done (so soothing that I fall asleep), it is time for another photograph. And strangely enough, even the sceptic in me has to admit there is a discernible difference in skin texture. It felt more elastic and from the photographs I could tell there were fewer few lines.

The beautician recommends a series of facials at regular intervals (depending on the condition of the skin and the amount of repair it needs). The salon will make a schedule for you and remind you when it is time to come for a treatment. The salon also provides you with tips on home care. Page 3 offers two high-end special facials. Both sound tempting: One is called Revisit your youth and the other Turn Back Time.

Anyone who is 35 plus can go for the Turn Back Time facial, says Shan of Page 3. This treatment is supposed to arrest ageing and promote the production of epidermal stem cells. It holds off the fine lines, wrinkles and sagging. Revisit Your Youth on the other hand is corrective. It promises to reduce the crows feet and lines that have already appeared on your face. The products in this line work like Botox without being invasive, says Shan.

The products are available at the salon. For appointments and details call:0422-4393333/4223331

What is mature skin?

Visit link:
A mature approach

The Binding of Isaac: Rebirth Unlimited Fart Sound Glitch – Video


The Binding of Isaac: Rebirth Unlimited Fart Sound Glitch
I played the FART SNDS seed and a bug happened, ED, or tyrone... Tyrone is a black name but he is Hispanic but appears to be white skin... Stem Cells The Bin...

By: djsponge10

Continued here:
The Binding of Isaac: Rebirth Unlimited Fart Sound Glitch - Video

Stem cells to repair broken chromosomes

(Ivanhoe Newswire) CLEVELAND, Ohio -- In 1990 the Human Genome Project started. It was a massive scientific undertaking that aimed to identify and map out the body's complete set of DNA. This research has paved the way for new genetic discoveries; one of those has allowed scientists to study how to fix bad chromosomes.

Our bodies contain 23 pairs of them, 46 total. But if chromosomes are damaged, they can cause birth defects, disabilities, growth problems, even death.

Case Western scientist Anthony Wynshaw-Boris is studying how to repair damaged chromosomes with the help of a recent discovery. He's taking skin cells and reprogramming them to work like embryonic stem cells, which can grow into different cell types.

You're taking adult or a child's skin cells. You're not causing any loss of an embryo, and you're taking those skin cells to make a stem cell. Anthony Wynshaw-Boris, M.D., PhD, of Case Western Reserve University, School of Medicine told Ivanhoe.

Scientists studied patients with a specific defective chromosome that was shaped like a ring. They took the patients' skin cells and reprogrammed them into embryonic-like cells in the lab. They found this process caused the damaged ring chromosomes to be replaced by normal chromosomes.

It at least raises the possibility that ring chromosomes will be lost in stem cells, said Dr. Wynshaw-Boris.

While this research was only conducted in lab cultures on the rare ring-shaped chromosomes, scientists hope it will work in patients with common abnormalities like Down syndrome.

What we're hoping happens is we might be able to use, modify, what we did, to rescue cell lines from any patient that has any severe chromosome defect, Dr. Wynshaw-Boris explained.

It's research that could one day repair faulty chromosomes and stop genetic diseases in their tracks.

The reprogramming technique that transforms skin cells to stem cells was so ground-breaking that a Japanese physician won the Nobel Prize in medicine in 2012 for developing it.

Link:
Stem cells to repair broken chromosomes

RGN Regenerative Skin Solution by Hansderma – Video


RGN Regenerative Skin Solution by Hansderma
Courtney Lee, our RGN user made a nice introduction video how to use RGN and her experience! RGN is made in USA and not a Korean brand. RGN Skincare is committed to providing world class ...

By: Hansderma, Inc.

See the article here:
RGN Regenerative Skin Solution by Hansderma - Video

Naturopathica: Soothing Winter Skin

By Emily J. Weitz

As a winter chill creeps into the air, we have to armor ourselves with extra layers of clothing, and make sure our homes are well-insulated. This carries over, too, to the way we care for our skin. Naturopathica, a business that started in East Hampton whose products can now be found in 350 spas across North America, offers a wide array of healing elixirs specifically formulated to balance and protect skin.

In New York, the winters tend to be cold and windy, said Barbara Close, Founder and CEO of Naturopathica. Cold air, wind, and artificial heat pull moisture from the skin and attack our skin barrier, which can lead to dry, irritated, sensitized and devitalized skin.

This can lead to skin losing its summer glow, and also to longer term issues like wrinkles and spotting. But having a consistent, healthy skincare regimen is a way to combat these issues.

If you consistently promote healthy skin with ingredients that target your concerns, said Ms. Close, you will experience the best possible result.

Other problems, like environmental pollutants, poor nutrition, and stress affect skin health.

Using ingredients with antioxidant and protective benefits on a daily basis helps to prevent premature aging and other visible concerns.

But Ms. Close says that even more important is how a skincare regimen can protect against disease. Specific ingredients to look for in your skincare products include Micronized Zinc Oxide, which reflects UVA/UVB rays. This compound is not absorbed into the bloodstream, so it remains on top of the skin like a shield. She also recommends seed oils, like carrot seed oil or rosehip seed oil.

Read this article:
Naturopathica: Soothing Winter Skin

Stem Cell Skin Care – BlueHorizonSkinCare.com

Blue Horizon's Special Skin Serum Stem Product Fact Sheet

Our Stem Cell Skin Care is a potent anti-aging innovation derived from non-embryonic human stem cell research. Blue Horizon International has infused medicines most promising clinical advances into this powerful skin care product.

Cytokine action, epidermal growth factors (EGFs), short and long-chained hyaluronic acid and ceramides combat the effects of aging and deliver unique skin benefits without surgery.

Our formulation is safe, having passed toxicology tests in accordance with European Union regulation 1223/2009/EC.

Patents are pending.

Our skin care is derived from what stem cell scientists call a conditioned medium. Here, human stem cells from placentas and umbilical cords condition the culture medium by releasing cytokines and other skin regenerating proteins that become available for skin repair. We stabilize the liberated cytokines, rendering them safe and accessible for aesthetic skin improvement. The conditioned medium is the base for our stem cell skin care products.

An independent skin test on twenty individuals aged 46 to 81 found a 23% reduction in skin roughness, including a decrease in the appearance of fine lines, wrinkles and scars.

Cytokines are one of todays most exciting captured biological processes, because they govern so many regenerative functions. The cytokine group of chemical regulators includes a diverse assortment of interleukins, interferons and growth factors that control anti-aging and activate the bodys immune system.

Cytokines stimulate, propagate and regulate new cell production in human skin. These messaging molecules mobilize cell division to help heal age related damage. Cytokines have powerful influence over skin texture and quality because they regulate cell shape, metabolism and migration from one location to another.

Several stem cell skin care ranges claim cytokine-style benefits. However, human stem cell cytokines are more biologically compatible with human skin than cytokine proteins from other sources.

See original here:
Stem Cell Skin Care - BlueHorizonSkinCare.com

Identifying the source of stem cells

7 hours ago Amy Ralston, MSU biochemist and molecular biologist, has identified a possible source of stem cells, which can advance regenerative and fertility research. Credit: G.L. Kohuth

When most animals begin life, cells immediately begin accepting assignments to become a head, tail or a vital organ. However, mammals, including humans, are special. The cells of mammalian embryos get to make a different first choice to become the protective placenta or to commit to forming the baby.

It's during this critical first step that research from Michigan State University has revealed key discoveries. The results, published in the current issue of PLOS Genetics, provide insights into where stem cells come from, and could advance research in regenerative medicine. And since these events occur during the first four or five days of human pregnancy, the stage in which the highest percentage of pregnancies are lost, the study also has significant implications for fertility research.

Pluripotent stem cells can become any cell in the body and can be created in two ways. First, they can be produced when scientists reprogram mature adult cells. Second, they are created by embryos during this crucial four-day window of a mammalian pregnancy. In fact, this window is uniquely mammalian, said Amy Ralston, MSU assistant professor of biochemistry and molecular biology, and lead author on the study.

"Embryos make pluripotent stem cells with 100 percent efficiency," she said. "The process of reprogramming cells, manipulating our own cells to become stem cells, is merely 1 percent efficient. Embryos have it figured out, and we need to learn how they're doing it."

The researchers' first discovery is that in mouse embryos, the gene, Sox2, appears to be acting ahead of other genes traditionally identified as playing crucial roles in stem cell formation. Simply put, this gene could determine the source of stem cells in mammals. Now researchers are trying to decipher why Sox2 is taking the lead role.

"Now we know Sox2 is the first indicator that a cell is pluripotent," Ralston said. "In fact, Sox2 may be the pre-pluripotent gene. We show that Sox2 is detectable in just one or two cells of the embryo earlier than previously thought, and earlier than other known stem cell genes."

The second discovery is that Sox2 has broader influence than initially thought. The gene appears to help coordinate the cells that make the fetus and the other cells that establish the pregnancy and nurture the fetus.

Future research will focus on studying exactly why Sox2 is playing this role. The team has strong insights, but they want to go deeper, Ralston said.

"Reprogramming is amazing, but it's inefficient," she said. "What we've learned from the embryo is how to improve efficiency, a process that could someday lead to generating stem cells for clinical purposes with a much higher success rate."

See the original post:
Identifying the source of stem cells

Cellular alchemy turns skin cells into brain cells

Move over stem cells. A different kind of cellular alchemy is allowing cells to be converted directly into other tissues to treat disease or mend injuries.

Stem cells have long been touted as the future of regenerative medicine as they can multiply indefinitely and be turned into many different cell types. Ideally, this would take a personal approach a patient's own cells would be converted into whatever type of cell is required to fix their injury or treat their symptoms. Earlier this year, for instance, people with age-related macular degeneration, the most common cause of blindness in the West, had retinal cells made from their own stem cells injected into their eyes.

Mature cells can be converted into stem cells by exposing them to a cocktail of chemicals that reverts them back to an embryonic-like state. Another set of chemicals is then used to turn the cells into the desired tissue type.

Skipping the stem cell stage would be more efficient, says Andrew Yoo of Washington University in St Louis, Missouri, and would reduce the chance that the new tissue could grow into a tumour a risk with stem cells because of their capacity to regenerate.

Yoo has now managed to do just that, using a process known as "transdifferentiation". His team have turned human skin cells into medium spiny neurons, the cells that go wrong in Huntington's disease.

To the skin cells, the team added two short snippets of genetic material called microRNAs. MicroRNAs are signalling molecules and the two they picked turn on genes in brain cells during embryonic development. They also added four transcription factors another kind of signalling molecule to turn on genes normally active in medium spiny neurons.

Within four weeks the skin cells had changed into MSNs. When put into the brains of mice, the cells survived for at least six months and made connections with the native tissue. "This is a very cool result," says Ronald McKay of the Lieber Institute for Brain Development in Baltimore.

The team's next step is to transplant the cells into mice with a version of Huntington's to see if the new neurons reduce their symptoms.

"Being able to produce cells with medium spiny neuron characteristics directly without first having to generate stem cells is impressive," says Edward Wild of University College London. "Using this offers the tantalising prospect of cell replacement treatments."

Wild points out, however, that before this approach can be used on people with Huntington's, researchers would first have to correct the faulty genetic mutation in their skin cells. And while medium spiny neurons are the first to degenerate in the disease, other brain cells may also be affected. "When it comes to cell replacement we should probably be aiming for a cocktail of cells," says Wild.

See the rest here:
Cellular alchemy turns skin cells into brain cells

WashU researchers tease out brain cells from skin cells

Researchers at Washington University have figured out how to scratch off a tiny piece of flesh from a persons forearm, isolate the skin cells and turn those cells into neurons the brain cells that transmit information in the form of electrical signals throughout the body.

The research is significant because it could put researchers on a path to treating certain diseases of the brain, including Alzheimers, ALS, Parkinsons and others.

Washington Universitys research, published Oct. 22 in the journal Neuron, relates specifically to Huntingtons disease, an inherited illness that affects an estimated 30,000 people in the U.S.

Huntingtons is caused when neurons in the brain start to malfunction and die. Initially, sufferers often experience problems with coordination and learning new information. As more neurons die, symptoms worsen and can be fatal.

Andrew Yoo, an assistant professor of developmental biology, explained that skin cells have the same DNA as brain cells.

To get skin cells to begin acting like brain cells, Yoos team at the Washington University medical school was able to create a process in which skin cells were reprogrammed into brain cells.

Basically we were able to pull a genetic trick to generate neurons, Yoo said.

The reprogrammed cells are a specific type that play a large role in controlling movement. They are typically the cells affected by Huntingtons disease.

The next step for researchers, Yoo said, is to re-create the conditions for Huntingtons in a Petri dish to study healthy brain cells alongside malfunctioning ones to get a better understanding of what causes the disease.

From there, Yoo said its possible that researchers develop a way to treat the illness.

Read more:
WashU researchers tease out brain cells from skin cells

Mini-Stomachs Let Scientists Study Ulcers in a Lab Dish

Scientists have grown miniature stomachs in a lab dish using stem cells, and are already using them to study stomach cancer. They hope they can grow patches to fix ulcers, find new drugs to treat and even prevent stomach cancer, and perhaps even grow replacement stomachs some day.

They discovered that the bacteria that cause stomach cancer begin doing their dirty work almost immediately, attaching to the stomach lining and causing tumors to start growing in response. Helicobacter pylori causes many, if not most, cases of stomach cancer, which affects more than 22,000 Americans a year and kills half of them. Stomach cancer is a major killer globally, affecting close to a million people a year and killing more than 70 percent of them.

And the team grew their mini-stomachs using two different types of stem cells human embryonic stem cells, grown from very early human embryos, but also induced pluripotent stem cells or iPS cells, which are made by tricking bits of skin or other tissue into acting like a stem cell.

In our hands they worked exactly the same, James Wells of Cincinnati Childrens Hospital Medical Center, who led the research. Both were able to generate, in a petri dish, human stomach tissue.

Immunofluorescent image of human stomach tissue made using stem cells

Stem cells are the body's master cells. Embryonic stem cells and iPS cells are both pluripotent meaning they can give rise to any tissue in the body. They've been used to grow miniature human livers, retinas, brain tissue and have been injected into eyes to treat eye disease.

Growing anything close to a real stomach or even a patch for an ulcer is a long way off. The gastric organoids Wellss team made the name up are just about the size of a BB bullet.

Its not easy getting stem cells to do what you want them to do. Wells and his team, including graduate student Kyle McCracken, had to use various growth factors and chemicals, each introduced at precisely the right time, to coax the cells into becoming three-dimensional blobs of stomach tissue. The stomach is a complex organ, with layers of muscle cells, cells that make up the stomach lining and glands that secrete proteins and acid to digest food.

"The bacteria immediately know what to do and they behaved as if they were in the stomach.

But the process worked, and the mini-stomachs look just like stomach tissue, the team reports in this weeks issue of the journal Nature.

See original here:
Mini-Stomachs Let Scientists Study Ulcers in a Lab Dish

Best Stem Cell Skin Care Beauty Creams and Serums

5-4-3-2-1 Product Countdown

Stem Cell Skin Care Reviews presents expert & user reviews and analysis of the best (& worst) products in leading edge anti-aging skin care science. Here are the 5 top ranked products as rated by expert reviewers, who are dermatologists, biologists, estheticians, physicians, and product formulators. Click on a stem cell skin care product name or image to view detailed information, or visitthe all reviewssection to examine a larger selection of stem cell skin care products and to search by name, category, or key word.

Rank

Product / Company

Source of Cells for Cytokines & Growth Factors

Expert Rating

User Rating

Cost (oz)

Online store

The editors and reviewers here are all science nerds and our passionate pursuit of the best stem cell skin creams on the planet separates us fromneurotypicals andputs us somewhere on the spectrum. That being said, we think this whole subject is critically important to survival of the home sapiens species. Especially to skin care aficionados (many of whom also qualify for nerddom). So our desire here is to find a way to communicate all this arcane knowledge into human-usable information. We might not get it right the first time around, so feel free to ask questions or just say say what??? whenever we obfuscate. We have gathered together a knowledge base which we hope will be helpful.

Link:
Best Stem Cell Skin Care Beauty Creams and Serums

Archives