Page 14«..10..13141516..2030..»

Archive for the ‘Skin Stem Cells’ Category

4 Neck Creams Dermatologists Swear By To Get Rid Of Wrinkles And Sagging Skin – SheFinds

Taking care of your neck is important for any anti-aging skincare routine. If neglected, wrinkles and sagging skin on your neck can be a major giveaway for your age. With that in mind, its important to not only focus on your face (especially forehead lines, eyes bags and crows feet, and smile lines), but also to pay attention to your neck when it comes to skincare.

Keep reading for neck creams dermatologists swear by!

Key Ingredients And Benefits

-Hyaluronic Acid, Aloe Vera, Citric Acid, Vitamin C, and Collagen: supports cell renewal, boosts elasticity and skin radiance, and reduces signs of aging

Promising Review

"I've only used this collagen cream from Maryann Organics for 3 days now which is only a short amount of times but noticed some amazing changes in my skin. I love that it is not greasy, nor shiny, and absorbs very quickly. The best part was the push pump to dispense the cream without getting your fingers into the cream. I'm 43 years old and started seeing lines from all areas of my face. Since I used the cream, my lines are less noticeable and my complexion is clearer and more brighter. I LOVE this cream! I cant wait to see more amazing results!!! I highly recommend trying this product." -Amazon Reviewer

Key Ingredients And Benefits

-Retinol: helps diminish the look of fine lines and wrinkles

-Vitamins A and E, Hyaluronic Acid, Green Tea, and Aloe Vera: helps improve texture and tone and increases skin's firmness and elasticity

Promising Review "Ive never used a moisturizer that works this well this fast! I was worried that it would be drying or sensitive because of the retinol but it feels so good on the skin and makes your skin look and feel amazing. It actually works so well that I only use it at night and for my skin it doesnt need to be used in the morning. My mother introduced me to it after I noticed how good her skin was looking lately. Cant believe how inexpensive this is for how high quality the ingredients and results are! The design of the container and pump is really cool too. It helps control the amount you want to use and insures that no product gets wasted. All in all I highly recommend this for any one at any age! My mom is in her 50s and Im in my 20s and it works well for the both of us!" -Amazon Reviewer

Key Ingredients And Benefits

-Peptides, Stem Cells, Collagen, and Elastin: helps tighten and rejuvenate the skin of the neck and dcolletage

-Vitamins C, Vitamin E, and Hyaluronic Acid: help restore firmness and elasticity and minimize the appearance of fine lines and wrinkles

Promising Review

"I really didnt have high expectations for this product. I panicked when I noticed my neck was making these crepe-y folds when I turned my head. I am so not happy about aging. Anyway, I shopped around and came upon this fabulous product and saw all the high ratings and great testimonials. This cream works! Im not even halfway through the bottle and I see a noticeable reductions in the folds and crepey-ness! My dcollet was fine before but now I notice the skin is more supple. Full disclosure: at night, I add a few drops of Vitamin E oil for extra oomph. In the morning, I apply sunscreen over it. Will definitely buy again." -Amazon Reviewer

Key Ingredients And Benefits

-Retinol SA: helps renew the surface of your skin

-Hyaluronic Acid: helps plump and hydrate the skin

-Glucose Complex: strengthens skin barrier

Promising Review

"This cream is amazing and best of all its easily absorbed and you can see the positive effects in a few days! I have been using it on my neck specifically at the back and sides where I have gotten these extremely deep and rough wrinkles. Its always embarrassed me that my skin is like this and Ive never been able to wear ponytails because its just too embarrassing to have my skin seen like this by others. Well, not anymore thanks to this cream! Now my neck area has made an incredible improvement, my deep wrinkles/creases have diminished incredibly and my double chin seems tighter/firmer and for a cream that I just recently started using- its improved my confidence so much already!" -Amazon Reviewer

SheFinds receives commissions for purchases made through the links in this post.

See original here:
4 Neck Creams Dermatologists Swear By To Get Rid Of Wrinkles And Sagging Skin - SheFinds

Poison hemlock and wild parsnip, A couple of bad actors – Commonwealth Journal’s History

This was reprinted with permission from Joe Boggs, OSU

Poison hemlock (Conium maculatum) and wild parsnip (Pastinaca sativa) are two of our nastiest non-native weeds found in Ohio. Poison hemlock is one of the deadliest plants in North America. Wild parsnip can produce severe, painful blistering. Both are commonly found growing together.

Poison hemlock and wild parsnip are members of the carrot family, Apiaceae. The old name for the family was Umbelliferae which refers to the umbel flowers. They are a key family feature with short flower stalks rising from a common point like the ribs on an umbrella.

Poison hemlock produces white flowers on stalks that create a more rounded look; perhaps a bit more like an umbrella. Wild parsnip has intense yellow flowers with the stalks producing a more flat-topped appearance.

Both are biennial weeds meaning that it takes two years for plants to produce seed. The seeds currently being produced will give rise to plants that spend their first year as low-growing basal rosettes. The plants produce a long, thick taproot while in this stage.

During their second year, plants "bolt" by producing erect, towering stalks and multi-branched stems topped with umbel flowers. Mature wild parsnip plants may top 6' tall while poison hemlock plants can tower to as much as 8 - 10' tall. Both are prolific seed producers

Wild parsnip plants have leaves that look vaguely like celery, another member of the carrot family. Mature plants have a single, thick, deeply grooved, greenish-yellow stem that sprouts lateral branches topped with flowers.

All stages of poison hemlock plants have bluish-green leaves that are 3-4 times pinnately compound. The deeply cut parsley-like leaflets have sharp points. Flowering plants have hairless, light-green to bluish-green stems that are covered with obvious reddish-purple blotches. However, the blotches may occasionally coalesce to cause stems to appear an almost solid color.

What are the Risks?

Poison hemlock plants contain highly toxic piperidine alkaloid compounds which cause respiratory failure and death in mammals. The roots are more toxic than the leaves and stems; however, all parts of the plant including the seeds should be considered dangerous.

The toxins must be ingested or enter through the eyes or nasal passages to induce poisoning; they do not cause skin rashes or blistering. Regardless, this plant should not be handled because sap on the skin can be rubbed into the eyes or accidentally ingested while handling food.

Wild parsnip sap contains psoralen which presents a completely different mode of action compared to the piperidine alkaloids in poison hemlock sap. Psoralen acts as a photosensitizing compound by inhibiting DNA synthesis in epidermal cells which kills these light-shielding cells responsible for protecting us from long-wave ultraviolet radiation (LWUVR) bombarding us in sunlight.

Severe blistering occurs when affected skin is exposed to LWUVR. The synergistic effect is called phytophotodermatitis (a.k.a. Berloque dermatitis) and the burn-like symptoms, as well as skin discoloration, may last for several months.

However, connecting skin blistering to exposure to wild parsnip sap can be a challenge. It takes around 24 hours for symptoms to first appear after exposure to LWURV and severe blistering typically doesn't peak until 48 -72 hours. The time required for symptoms to appear after exposure to the sap means the effect may be disconnected from the cause.

Another challenge with connecting the dots is that wild parsnip commonly grows in and around other weeds, particularly poison hemlock (Conium maculatum). Gardeners who are exposed to wild parsnip sap while weeding a mixed-patch may mistakenly blame the poison hemlock for their ultimate misery.

To Mow, or Not to Mow

The potential for poisonings from poison hemlock sap and the extreme skin reaction to the wild parsnip sap means these non-native invasive weeds should not be allowed to grow where they can be easily contacted by people. However, mechanical control through mowing, weed trimming, or hand-pulling is problematic. Certainly, wild parsnip presents a much higher risk with reports of sap spattered by mowers and string trimmers producing phytophotodermatitis on exposed arms and legs of equipment operators.

Still, mowing provides one option for managing poison hemlock and to a lesser degree wild parsnip. However, timing is everything: plants should be mowed in the spring once they've bolted but prior to the appearance of flowers. Waiting until after flowering presents a risk the cut flowers will still mature to seed.

Chemical Control: Case Study

A strong case can be made for herbicides providing the most effective and safest approach to managing both poison hemlock and wild parsnip.

Wild parsnip and poison hemlock are both susceptible to non-selective post-emergent herbicides such as glyphosate (e.g. Roundup). However, "non-selective" means all plants - both good and bad - may be killed and there is a considerable downside to killing the competition as well as the targeted weeds.

Post-emergent herbicides do not affect seeds. Thus, "herbicidal openings" that occur when all plants are killed provide the perfect opportunity for more wild parsnip and/or poison hemlock to spring forth from previously deposited seed. Thus, it's important to have a plan for establishing competitive plants after the wild parsnip dies off such as over-seeding with grasses.

Selective post-emergent herbicides that will preserve competitive plants, particularly grasses, while removing poison hemlock and wild parsnip include 2, 4-D, clorpyralid (e.g. Transline), metsulfuron (e.g. Escort XP), and some 2 and 3-way products such as Triamine (2,4-D + MCPA). However, timing is equally important. Apply after the spring emergence of the targeted weeds but before flowering.

For more information, contact the Pulaski County Extension Service at 606-679-6361. Learn about timely events or things to do in your home gardens by becoming a fan of Pulaski County Horticulture on Facebook, or following @hortagentbeth on Twitter, kyplants on Instagram, and Pulaski County Horticulture YouTube channel.

The Pulaski Co Extension office is open to the public by appointment only through the month of June. Extension employees are still on the job and can be reached via office phone. Read the entire directive on the Pulaski County Cooperative Extension website at pulaski.ca.uky.edu.

The Lake Cumberland Master Gardeners are temporarily out of pine straw. Another load will be coming soon.

Educational programs of Kentucky Cooperative Extension serve all people regardless of economic or social status and will not discriminate on the basis of race, color, ethnic origin, national origin, creed, religion, political belief, sex, sexual orientation, gender identity, gender expression, pregnancy, marital status, genetic information, age, veteran status, or physical or mental disability.

We are making critical coverage of the coronavirus available for free. Please consider subscribing so we can continue to bring you the latest news and information on this developing story.

More here:
Poison hemlock and wild parsnip, A couple of bad actors - Commonwealth Journal's History

Irish dad with rare form of cancer would have died within a week if he didn’t go to hospital – Irish Mirror

When Eoin OBrien found out he had a rare form of cancer, he was told he would have died within a week had he not gone to hospital.

Now, five years on, the dad of four is remarkably free of the disease.

Eoins life changed in May 2015 when he went to A&E with chest pains.

He was diagnosed with Hodgkins lymphoma, which causes abnormal growth of cells in the lymphatic system.

Due to fluid build-up, Eoins heart would have suffocated within days had he not been treated.

And after half a decade of pain and suffering, he is finally in remission.

The news came on his wife Karens birthday, making it all the more special for the pair and their daughters Sophie, 13, Abbie, 11, Maddie, eight, and three-year-old Emelie.

Karen said: To say that that was the best news ever would be an understatement, I would rather be told that 10 times over than even win the lotto.

Eoin was only 31 when he was diagnosed following a hospital visit after he started getting pains in his chest.

Doctors drained three-and-a-half litres of fluid from his chest and found a tumour between his lungs and heart.

After his first round of chemo didnt work, Eoin found a lump on his neck.

He was started on a higher dose of chemo which was, in his wife Karens opinion, the hardest one on him.

She explained: Eoin got the moon-face, he got the cancer look. Darkness under the skin of his eyes and that.

The pair hoped this treatment was working but were disappointed again when doctors told them it hadnt.

Two years later in 2017, when Karen was pregnant with Emelie, Eoin still wasnt responding to treatments.

He was due to go into hospital after his daughter was born for a planned stem cell transplant which was later cancelled.

The pair fought to get Eoin immunotherapy, which slowed down but didnt cure his cancer.

In 2019, he was told he could get an allogeneic stem cell transplant from a donor. Karen explained: So on the 6th of November, which we now class as Eoins re-birthday, he was given the transplant and he became so, so bad.

Eoin was at the stage where he wanted to give up. He didnt want to live anymore, he didnt want to go through it anymore.

Results of a scan in February had alarming results which left the two terrified the cancer had spread.

Thankfully, it was only an infection.

Eoin was hospitalised for six weeks and due to the coronavirus, wasnt allowed outside or to have visitors.

In May, the pair were given the news his transplant worked.

Karen said: Theres been a lot of ups and a lot of downs but were finally out the other side, so hopefully we can look forward to many, many years cancer-free.

Originally posted here:
Irish dad with rare form of cancer would have died within a week if he didn't go to hospital - Irish Mirror

Vitamin C Face Serum – Triple Strength Anti Aging, Wrinkle & Brightening Facial Care – Antioxidant Rich, Hydrating, Softening & Even Skin Tone…

Vitamin C Face Serum Triple Strength Anti Aging, Wrinkle & Brightening Facial Care Antioxidant Rich, Hydrating, Softening & Even Skin Tone Treatment Made With Niacinamide & Glycolic Acid 1 oz Price, Features and Real Customers Reviews.

Check The Updated Price of Vitamin C Face Serum Triple Strength Anti Aging, Wrinkle & Brightening Facial Care Antioxidant Rich, Hydrating, Softening & Even Skin Tone Treatment Made With Niacinamide & Glycolic Acid 1 oz:

Price: Check Price on Amazon

Top Features of Vitamin C Face Serum Triple Strength Anti Aging, Wrinkle & Brightening Facial Care Antioxidant Rich, Hydrating, Softening & Even Skin Tone Treatment Made With Niacinamide & Glycolic Acid 1 oz Are:

3 POWERFUL FORMS OF VITAMIN C Our Triple Strength facial serum incorporates three powerful forms of Vitamin C to effectively address aging concerns within multiple layers of the skin, while reducing signs of aging. Our anti aging serum also leverages the power of Glycolic Acid to boost cell turnover and exfoliating dulling skin cells, allowing for enhanced absorption of the active ingredients into the skin.ANTI AGING FACE SERUM The 3 advanced forms of Vitamin C in our anti wrinkle serum effectively addresses aging concerns. Enhanced with Glycolic Acid, Niacinamide and NASA Stem Cell Technology, our hydrating serum targets the appearance of deep wrinkles while renewing, brightening and softening the skin for a smooth and even complexion.HEALTHIER SKIN COMPLEXION Our brightening serum defends against external aggressors, supporting overall skin barrier strength. This anti aging skin care treatment leverages Niacinamide and Glucosamine to support skin strength and improve moisture. These benefits further support a reduction in the look of wrinkles and improves radiance, while brightening the look of skin.PERFECT FOR ALL SKIN TYPES Our vitamin c facial serum works great for men, women and all skin types including normal, dry, oily or combination. Its great for naturally treating anti aging skin concerns, deep wrinkles and mature skin! On cleansed & toned skin, apply a dime sized amount of the serum in light, upward motions. For best results, we recommend applying at night & following with your favorite moisturizer. Daily application of an SPF is strongly encouraged while using this product.100% SATISFACTION GUARANTEE InstaNatural products are developed and manufactured in registered, GMP compliant facilities in the USA. Every product is free from parabens, sls/sles, mineral oil, formaldehyde releasers, synthetic dyes, petroleum, polyethylene glycol (PEGs), DEA/MEA/TEA, and unsafe preservatives. We are Leaping Bunny Certified meaning we do not test our products on animals at any stage in product development. Feel free to contact us with any questions or concerns.

Verified Customers Reviews of Vitamin C Face Serum Triple Strength Anti Aging, Wrinkle & Brightening Facial Care Antioxidant Rich, Hydrating, Softening & Even Skin Tone Treatment Made With Niacinamide & Glycolic Acid 1 oz:

Read the original:
Vitamin C Face Serum - Triple Strength Anti Aging, Wrinkle & Brightening Facial Care - Antioxidant Rich, Hydrating, Softening & Even Skin Tone...

Skin Toner Market 2020: Analysis Of Sales, Overview, Segmentation And Growth Rate To 2027 – 3rd Watch News

Trusted Business Insights answers what are the scenarios for growth and recovery and whether there will be any lasting structural impact from the unfolding crisis for the Skin Toner market.

Trusted Business Insights presents an updated and Latest Study on Skin Toner Market 2019-2026. The report contains market predictions related to market size, revenue, production, CAGR, Consumption, gross margin, price, and other substantial factors. While emphasizing the key driving and restraining forces for this market, the report also offers a complete study of the future trends and developments of the market.The report further elaborates on the micro and macroeconomic aspects including the socio-political landscape that is anticipated to shape the demand of the Skin Toner market during the forecast period (2019-2029).It also examines the role of the leading market players involved in the industry including their corporate overview, financial summary, and SWOT analysis.

Get Sample Copy of this Report @ Global Skin Toner Market 2020 (Includes Business Impact of COVID-19)

Global Skin Toner Market, Trends, Industry Analysis, Market Size, and Growth Forecast till 2029 is an upcoming report in the process of preparation by the research and analysis team at Trusted Business Insights. The global skin toner market report is structured to deliver insightful details on the various market trends, industry analysis, growth and potential revenue opportunities, and micro and macro indicators. The report has been generated using exhaustive primary and secondary research efforts. The global skin toner market report includes a market competitive landscape, market revenue (US$) and value share (%), market growth, characteristics, regional breakdowns, trends, and market strategies. The report also delivers a full analysis of competitors and key players. The report offers information on market restraints, limitations, and market scenarios, which can impact market growth. In the report, the market is segmented on the basis of product type, product form, distribution channel, and regions and countries.

Request Covid 19 Impact

Skin Toner: Introduction

Among cosmetics products, skin toner or toner, is usually used on the face, and is considered a product that is designed to cleanse the skin and reduce pore size and appearance. Skin toner can be applied using a piece of cotton (a commonly used method), spraying onto the face, or by applying a toner gauze facial mask. A toner, enhances skin surface and imparts a fresh and smoother texture, and keeps the skin hydrated. Toner is a popular beauty product among women consumers, and use of the product has been increasing in the recent past.

Global Skin Toner Market: Dynamics

Drivers: Revenue growth of the global skin toner market is driven by growing awareness about skincare products and ability of some products to rejuvenate and repair damaged or aging cells and tissue. Taking care of skin and hair health and shifting to a healthy lifestyle is a popularly growing trend among millennial consumers in countries across the globe. Toner can alleviate incidence of acne and oily skin-related issues. A variety of skin toners are available in the market, and some have unique properties. Products have been developed and formulated to suit specific skin-related issues, while others are for general use and application. Consumers with dry skin are recommended to use rosewater-based skin toner for improved skin hydration. A current preference among consumers is rising towards plant stem cell-based skin toners, which contain antioxidants and offer anti-aging benefits.

Many cosmetic product manufacturers are increasing investments on the R&D to develop and formulate more ranges of natural or organic beauty products. Awareness among end-users is increasing regarding benefits associated with use of natural ingredients such as coconut oil, almonds, aloe vera, and others. Consumer preference for natural skin toners is based on ingredients source, and is currently among some of the factors that could influence purchase decision.

By Product Type:

The inorganic segment currently accounts for higher revenue share in the market as market penetration, awareness, and product visibility of organic-based products is relatively low, especially in some developing economies. However, rising concerns regarding potential adverse effects associated with skin creams and lotions that contain chemical or synthetic ingredients and formulations is expected to negatively impact revenue growth of the inorganic segment over the next few years.

By Product Form:

Among the product form segments, the fluid segment is projected to continue to lead in terms of revenue share, as this type of product is easier to apply and more convenient to use.

By Distribution Channel:

Among the distribution channel segments in the global skin toner market report, the conventional stores segment as well as the supermarket segment are expected to account for significantly high revenue growth over the forecast period.

However, revenue from the online channel segment is projected to expand at the fastest CAGR over the forecast period. This high projected growth can be attributed to convenience of shopping anytime-anywhere, and high visibility on e-Commerce platforms, as well as free and prompt home delivery and discount offers available online. Social media platforms and influencers using such channels to influence purchase potential, and a number of beauty bloggers or vloggers providing tips on usage and a broader understanding of toners and features, which makes purchasing beauty products is driving market growth. Online channels provide access to a larger product range, some of which might not be accessible in a conventional store.

By Region

The market in North America is expected to account for significant revenue growth. Followed by markets in Asia Pacific, Europe, and Middle East & Africa respectively.

Analysis by Region: Global Skin Toner Market

North America Market Analysis:

North American market growth is driven significantly by revenue contribution from sales in countries such as the US and Canada.

Europe Market Analysis:

Europe is a major market for skin toner owing to significantly high awareness regarding benefits of skincare. Many premium skin toner brands are France-based, and countries in Europe experience long and cold winters, which are major factors driving demand for cold creams, lotions, and other products designed for cleansing and nourishing the skin.

Asia Pacific Market Analysis

Asia Pacific market is projected to register fastest CAGR over the forecast period, which can be attributed to growing population in countries in the region, and expanding younger potential consumer base in counties in the region. These factors, clubbed with increasing consumer base that is focused on health and wellness, as well as personal appearance resulting in a more streamlined beauty regimen, along with increasing need for beauty and skincare products among consumers is projected to continue to drive market growth. Furthermore, launch of new products in countries in the region is expected to open up new avenues for existing players in the skin toner market in Asia Pacific.

Global Skin Toner Market Segmentation:

Segmentation by Product Type:

OrganicInorganic

Segmentation by Product Form:

FluidsMists

Segmentation by Distribution Channel:

SupermarketsConventional StoresOnline

Quick Read Table of Contents of this Report @ Global Skin Toner Market 2020 (Includes Business Impact of COVID-19)

Trusted Business InsightsShelly ArnoldMedia & Marketing ExecutiveEmail Me For Any ClarificationsConnect on LinkedInClick to follow Trusted Business Insights LinkedIn for Market Data and Updates.US: +1 646 568 9797UK: +44 330 808 0580

Continued here:
Skin Toner Market 2020: Analysis Of Sales, Overview, Segmentation And Growth Rate To 2027 - 3rd Watch News

Microneedling therapeutic stem cells into damaged tissues – PRNewswire

LOS ANGELES, June 10, 2020 /PRNewswire/ --Mesenchymal stem cells (MSCs) are multipotent in that they naturally replenish the cell types that build our bone, cartilage and adipose tissues. Their regenerative potential makes them exquisite candidates for cell-based therapies. However, clinicians often need to administer massive numbers of MSCs to reach sufficient numbers of cells that successfully engraft and remain functional over time.

Now, a new study in Advanced Functional Materials by a team at the Terasaki Institute for Biomedical Innovation in Los Angeles and the University of California, Los Angeles (UCLA) has reported a minimally-invasive approach, which deploys an array of "microneedles" that provide a bioactive depot of MSCs. By embedding MSCs in a gel-like material that prolongs their viability and functionality, and targeting damaged tissues with high spatial precision, the researchers showed their approach to accelerate wound healing in a mouse model with excised skin segments.

"Microneedles have been successfully used in the past to painlessly deliver drugs to target tissues such as skin, blood vessels and eyes. We demonstrate here with 'Detachable Microneedle Depots' that an analogous approach can deploy therapeutic cells at target sites," said co-corresponding author Ali Khademhosseini, the Director and CEO of the Terasaki Institute.

The team set out to investigate their microneedle concept in a mouse skin wound model in which a defined excision is made in the epidermal tissue layers. To be able to strategically place individual microneedles within the wound bed, a simple and effective deployment mechanism was devised by attaching an array of microneedles on a small strip of scotch tape with their pointed ends facing away from the tape. Precisely positioning the tape with its patterned microneedle surface on the wound, allowed the individual microneedles to penetrate into the wound bed. Then, the tape was peeled off, causing the microneedles to detach and remain embedded in the wound tissue.

"The concept would even be compatible with using patient-derived cells in more personalized device approaches," said Khademhosseini who is exploring further uses of this technology as part of the Terasaki Institute's research program.

The Terasaki Institute for Biomedical Innovation (terasaki.org) is a non-profit research organization that invents and fosters practical solutions that restore or enhance the health of individuals.

SOURCE Terasaki Institute

https://terasaki.org/institute/

Link:
Microneedling therapeutic stem cells into damaged tissues - PRNewswire

Major skin cancer research study to begin at The Hormel Institute – Austin Daily Herald – Austin Herald

Dr. Rebecca Morris, leader of the Stem Cells and Cancer lab at The Hormel Institute, received a multi-year grant to study stem cells originating in adult bone marrow and their possible effects on skin diseases, including cancer. The grant, from the Nation Institute of Healths National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), awarded Morris with $373,688 over two years for her research project Identification of Novel Epidermal Progenitors.

Morris said this research is significant because it will contribute new understanding of epithelial biology, and blood and bone marrow in general, provide possible new targets for epithelial cancer prevention and control, validate liquid biopsy of blood as a diagnostic tool, and help her and her team to achieve their goal of preventing and alleviating chronic skin diseases including cancer, psoriasis, and epidermolysis bullosa.

Many years ago, I contributed basic research on identification and isolation of adult tissue stem cells from skin epidermis, and demonstrated their role in skin cancer initiation and promotion, Morris said. Now, I am again thrilled to be on the edge of discovery of a new population of epithelial stem cells and have the opportunity to determine their roles in regeneration and cancer.

Cells in the body that cover surfaces (like the epidermis, or top layer of skin) or line spaces (like ducts in mammary gland or lining of the colon) are called epithelial cells. In adults, most cancers originate from these epithelial cells. However, new research has identified certain bone marrow derived epithelial cells (BMDECs) in normal, healthy human subjects.

Morris and her team do not believe anyone has yet described the features and nature of these cells, or analyzed their function.

The research team has hypothesized that the epithelial cells from the bone marrow are epithelial stem cells. They therefore hope to demonstrate that BMDECs include a novel population of adult tissue stem cells that can be gathered to chronically compromised epithelium, such as skin cancer or psoriasis, and regenerate it.

Skin cancer is by far the most common type of cancer in the United States, with millions of people diagnosed each year. As we enter summer, it is important to remember simple steps like staying out of the sun during the middle of the day, staying in the shade, and wearing sunscreen can help reduce your skin cancer risk.

Next steps for Morriss research include determining how these blood borne epithelial cells are recruited to the skin, the recruiting molecules, how the recruitment can be good or bad, and how to modulate their recruitment to alleviate disease.

See more here:
Major skin cancer research study to begin at The Hormel Institute - Austin Daily Herald - Austin Herald

Exploring the Therapeutic Potential of ST266 Against Numerous Diseases Including COVID-19 – Technology Networks

Noveome Biotherapeutics is a clinical-stage company focused on developing therapies for the regenerative repair of tissues. Their product ST266, a first-of-its-kind, multi-targeted, non-cellular platform biologic comprised of a complex mixture of biomolecules, is currently being evaluated as a potential treatment for the severe inflammatory response observed in the lungs of some COVID-19 patients.Technology Networks recently spoke with William J. Golden, Noveome Biotherapeutics Founder, Chairman and CEO, who explains the underlying basis for investigating ST266s potential against COVID-19. Golden also elaborates on many of the other indications for which ST266 is being developed to treat.Laura Lansdowne (LL): Could you provide our readers with a brief overview of Noveome Biotherapeutics?William J. Golden (WJG): Noveome is a clinical-stage biotherapeutics company located in Pittsburgh, PA. The company was founded in 2000 by Bill Golden and Lancet Capital. The group was interested in exploring non-embryonic stem cells and identified a technology at the University of Pittsburgh that was using cells derived from human amnion, a membrane that closely covers the fetus during development. The company, named Kytaron Technologies, Inc. at the time, licensed that amnion cell technology but, ultimately, Noveome scientists were able to discover, develop and patent their own unique population of cells, called Amnion-derived Multipotential Progenitor (AMP) cells, using a proprietary culture method that follows current Good Manufacturing Practice (cGMP) regulations. These novel cells were used to produce our product, ST266.LL: What is ST266? Could you elaborate on its mechanism of action in relation to the healing process?WJG: Noveomes product, ST266, is the secretome produced by the AMP cells. It is a completely cell-free solution and is comprised of hundreds of biologically active molecules, including cytokines and growth factors. Interestingly, these cytokines and growth factors exist at very low physiological levels ranging from pg/mL ng/mL concentrations.1 The fact that such low concentrations of these molecules are biologically active is quite remarkable when you consider that traditional protein-based therapies are usually administered at concentrations that are orders of magnitude greater than the concentrations found in ST266.Because the composition of ST266 is so complex, its multiple mechanisms of action have only been partially elucidated. Clinical and preclinical studies have shown ST266 to be anti-inflammatory,2,3 promote wound healing,4,5 reduce apoptosis, reduce vascular permeability (manuscript in preparation), and restore cellular homeostasis.3 Preclinical studies have also shown ST266 to be neuroprotective. In a traumatic brain injury model, ST266 significantly protected against reactive gliosis, suggesting potent anti-inflammatory activity, and resulted in significant recovery of rotarod motor function.6,7 In another study, ST266 was tested in the experimental autoimmune encephalopathy (EAE) mouse model of multiple sclerosis (MS). In this model, the mice develop optic neuritis, which is among the presenting symptoms of MS in humans. ST266 was administered to the nares of mice 15 or 22 days after disease induction. ST266 is absorbed via capillary action along the olfactory nerves which bypasses the blood-brain barrier. This unique route of administration allows for the delivery of high molecular weight biologics to the optic nerve of the eye and the central nervous system. ST266 attenuated visual dysfunction, prevented retinal ganglion cell (RGC) loss, reduced inflammation, and decreased the rate of demyelination of the optic nerve in EAE mice.3Mechanistically, ST266 simultaneously acts on multiple cell receptor-activated and intracellular signaling pathways. For example, in the EAE MS model, neuroprotective effects involved oxidative stress reduction, SIRT1-mediated mitochondrial function promotion, and pAKT signaling.3 In a Phase 2 UV light burn study, ST266 reduced erythema and DNA damage and increased the expression of XPA DNA repair proteins.2Importantly, ST266 has a proven clinical safety profile. It has been administered to 243 patients by various routes of administration (topical skin, topical ocular, topical oral, targeted intranasal), and no drug-related serious adverse events have been reported. Preclinical studies of systemically administered ST266 have also yielded no drug-related safety concerns.LL: For what indications is ST266 currently being evaluated as a treatment?WJG: We refer to ST266 as a platform biologic. By this, we mean that ST266 is one product that has the potential to treat numerous and varied diseases. In the clinic, we have shown anti-inflammatory activity when ST266 is applied topically to UV light-burned the skin2 and topical application to the gums of patients with gingivitis and periodontitis showed a reduction in proinflammatory cytokines in the patients crevicular fluid (manuscript in preparation). We are currently conducting a Phase 2 open label trial of ST266 to treat persistent corneal epithelial defects (PEDs) when applied topically to the eye. Results from this trial will be published soon. We are currently planning a Phase 2b multi-center, randomized, double-masked trial to further evaluate the safety and efficacy of ST266 in this indication. Finally, we are conducting a Phase 1 study in patients at risk for developing glaucoma. This study is using the intranasal route of delivery described above in combination with a novel delivery device. The goal is to deliver ST266 directly to the optic nerve, where it can protect the RGCs that are damaged in glaucoma. We envision this route of delivery will be applicable to central nervous system and other back-of-the eye indications.We also have several ongoing preclinical programs that are evaluating systemically administered ST266 for more generalized inflammatory conditions. These data are not yet published but combined with the data we have compiled in preclinical and clinical studies of topical skin, topical oral and topical ocular administration, we believe ST266 has the potential to be an effective therapy for numerous systemic inflammatory conditions.LL: Could you elaborate on the underlying basis for your evaluation of ST266 as a potential treatment for COVID-19?WJG: As you know, a major complication of COVID-19 is the severe inflammatory response seen in the lungs of some patients. This response is called cytokine storm or cytokine release syndrome. As the pandemic continues and more data have become available, it is now known that the cytokine storm does not just affect the lungs. Multi-organ damage occurs in many of these patients. We believe that systemic delivery of ST266 and its anti-inflammatory activity has the potential to calm the storm. Our as-yet-unpublished preclinical studies with intravenous ST266 support this hypothesis and we are moving rapidly to initiate intravenous ST266 in a Phase 1 study. Once safety in humans is established by this route of administration, we will commence Phase 2 studies in COVID-19 patients.William J. Golden was speaking to Laura Elizabeth Lansdowne, Senior Science Writer for Technology Networks.References

1. Steed, DL, C Trumpower, D Duffy, C Smith, V Marshall, R Rupp, and M Robson. (2008). Amnion-Derived Cellular Cytokine Solution: A Physiological Combination of Cytokines for Wound Healing. Eplasty 8: 15765.

2. Guan, Linna, Amanda Suggs, Emily Galan, Minh Lam, and Elma D. Baron. (2017). Topical Application of ST266 Reduces UV-Induced Skin Damage. Clinical, Cosmetic and Investigational Dermatology. DOI: https://doi.org/10.2147/CCID.S147112.

3. Khan, Reas S, Kimberly Dine, Bailey Bauman, Michael Lorentsen, Lisa Lin, Helayna Brown, Leah R Hanson, et al. (2017). Intranasal Delivery of A Novel Amnion Cell Secretome Prevents Neuronal Damage and Preserves Function In A Mouse Multiple Sclerosis Model. Scientific Reports. DOI: https://doi.org/10.1038/srep41768.

4. Bergmann, Juri, Florian Hackl, Taro Koyama, Pejman Aflaki, Charlotte a Smith, Martin C Robson, and Elof Eriksson. (2009). The Effect of Amnion-Derived Cellular Cytokine Solution on the Epithelialization of Partial-Thickness Donor Site Wounds in Normal and Streptozotocin-Induced Diabetic Swine. Eplasty 9: e49.

5. Franz, Michael G, Wyatt G Payne, Liyu Xing, D K Naidu, R E Salas, Vivienne S Marshall, C J Trumpower, Charlotte A Smith, David L Steed, and M C Robson. (2008). The Use of Amnion-Derived Cellular Cytokine Solution to Improve Healing in Acute and Chronic Wound Models. Eplasty 8: e21.

6. Deng-Bryant, Ying, Zhiyong Chen, Christopher van der Merwe, Zhilin Liao, Jitendra R Dave, Randall Rupp, Deborah a Shear, and Frank C Tortella. (2012). Long-Term Administration of Amnion-Derived Cellular Cytokine Suspension Promotes Functional Recovery in a Model of Penetrating Ballistic-like Brain Injury. The Journal of Trauma and Acute Care Surgery DOI: https://doi.org/10.1097/TA.0b013e3182625f5f.

7. Deng-Bryant, Ying, Ryan D. Readnower, Lai Yee Leung, Tracy L. Cunningham, Deborah A. Shear, and Frank C. Tortella. (2015). Treatment with Amnion-Derived Cellular Cytokine Solution (ACCS) Induces Persistent Motor Improvement and Ameliorates Neuroinflammation in a Rat Model of Penetrating Ballistic-like Brain Injury. Restorative Neurology and Neuroscience. DOI: https://doi.org/10.3233/RNN-140455.

Read the rest here:
Exploring the Therapeutic Potential of ST266 Against Numerous Diseases Including COVID-19 - Technology Networks

‘I found these 4 affordable winter skin saviours in the chemist beauty aisles.’ – Mamamia

The light, non-greasy, dermatologically tested formula contains Vitamin B3 to help hydrate and support the skin barrier, and SPF30 for added sun protection. It alsosits beautifully under your regular SPF you'll have on, and makeup.

It won't clog your pores and doesn't include common irritants like fragrance or colour. The no-fuss formula also won't mess with the rest of your skincare, making it the easy to slot into your existing routine. Done!

(*Side note:Always read the label. Follow the directions for use. Avoid prolonged sun exposure and wear protective clothing, hats and eyewear to further reduce risk. Frequent re-application is required.)

Anyone else get dry lips in winter? This lip balm is brilliant for a few reasons.

A) It's super affordable and accessible - you'll find it at most pharmacies.

B) It has SPF50+ broad spectrum protection.

C) The texture isn't too thick or too thin, it's just right.

And D) You can wear the clear formula over the top of alip stain or lip liner.

Just don't leave it in the car, OK?

(And yes, you know the drill:Always read the label. Follow the directions for use. Avoid prolonged sun exposure and wear protective clothing, hats and eyewear to further reduce risk. Frequent re-application is required.)

View post:
'I found these 4 affordable winter skin saviours in the chemist beauty aisles.' - Mamamia

Rapid Unit Sales of Oncology Drugs to Account for Incremental Revenues in the Global Market – Medic Insider

Oncology is a branch of study and treatment of cancer. Cancer is disease in which abnormal cells grow and divide without control. Oncology drugs help in diagnosis cancer. Some of the causes of cancer are tobacco and smoking, viral infections, genetic causes, carcinogens, bacterial infections, physical activities, eating habits and age. Various types of cancer that can be treated by oncology drugs are blood cancer, endocrine cancer, lung cancer, bone cancer, skin cancer, genitourinary cancer, gastrointestinal cancer, breast cancer, eye cancer, head and neck cancers and gynaecologic cancer. On the basis of treatment, oncology drugs market can be segmented into chemotherapy, immunotherapy, surgery, radiation therapy, stem cell transplant, hormone therapy and others.

Get Free Sample Copy With Impact Analysis Of COVID-19 Of Market Report @https://www.persistencemarketresearch.com/samples/3312

North America, followed by Europe, has the largest market for oncology drugs due to new drug development, government initiatives and developed healthcare infrastructure in this region. Asia is expected to show high growth rate in the oncology drugs market in next few years due to increasing incidence of cancer cases, rise in the use of tobacco products and growth in aging population in the region.

Technological advancement, increasing incidence of various type of cancers, rise in need for R&D activities in cancer and growing concerns over high death rates due to cancer are driving the market for oncology drugs. In addition, introduction of new drugs and therapies for cancer and government support to improve healthcare condition are expected to drive the market for oncology drugs. However, high cost of cancer treatments, strict government regulations, huge investment involvement in the development and clinical trials of the therapies and side effects of cancer treatments are some of the major factors restraining the growth for global oncology drugs market.

You Can Buy This PMR Healthcare Report From Here @https://www.persistencemarketresearch.com/checkout/3312

Growing demographics and economies in the developing countries such as China and India are expected to offer good opportunities in oncology drugs market in Asia. In addition, new innovations in cancer drugs and therapies and rise in awareness about the new drugs and therapies available in the market are expected to offer new opportunities for global oncology drugs market. Personalized medicines, increasing number of mergers and acquisitions, new product launches and rise in number of collaborations and partnerships are some of the trends that have been observed in global oncology drugs market.

Some of the major companies operating in the global oncology drugs market are Amgen, Bayer Healthcare AG, CELGENE CORPORATION, GlaxoSmithKline, ARIAD Pharmaceuticals, Inc., Eli Lilly and Company, Novartis, Hoffmann-La Roche Ltd., AstraZeneca, Boehringer Ingelheim GmbH, Pfizer and Teva Pharmaceuticals Industries.

See the original post:
Rapid Unit Sales of Oncology Drugs to Account for Incremental Revenues in the Global Market - Medic Insider

Ability to ‘Create’ Astrocytes Supports Their Damaging Role in MS… – Multiple Sclerosis News Today

An inflammatory environment can turn astrocytes, key supportive cells for neurons, into their killers, fostering the progression of neurodegenerative diseases like multiple sclerosis (MS), a new study shows.

This work, led by researchers at the New York Stem Cell Foundation(NYSCF), created for a first time astrocytes derived from human induced pluripotent stem cells (hIPSCs). The group then placed these cells in an inflammatory environment, and observed what happened.

Now that we can create this critical brain cell type from any individuals stem cells and capture its errant behaviors, we can better understand its role in diseases like multiple sclerosis, Parkinsons, and Alzheimers, Susan L. Solomon, the CEO of theNYSCF, said in a press release.

This will shed new light on the devastating process of neurodegeneration, pointing us towards effective treatments for this growing group of patients, Solomon added.

The study CD49f Is a Novel Marker of Functional and Reactive Human iPSC-Derived Astrocytes was published in the journal Neuron.

Astrocytes compose more than half of the cells of the central nervous system (brain and spinal cord), and work as support cells. They help to maintain brain homeostasis (stable equilibrium), provide neurons with metabolic support, enhance the connectivity of neural circuits, and control the brains blood flow.

Yet, these cells are also thought to be key players in the onset and progression of neurodegenerative diseases such as MS.

Knowledge on astrocyte biology has mostly come from animal models, namely rodents, since scientists struggle to obtain astrocytes from people.

NYSCF researchers developed a method to generate functional astrocytes that are derived from human IPSCs. (IPSCs themselves arederived from either skin or blood cells that have been reprogrammed back into a stem cell-like state, which allows for the development of an unlimited source of almost any type of human cell.)

They based their work on a previous protocol, which they developed to produce oligodendrocytes one type of cell capable of producing myelin, the protective layer covering nerve fibers and whose loss triggers MS.

Here, the researchers generated a mix of astrocytes and neurons.

They then conducted a screen to identify a surface protein that could be used to specifically purify astrocytes.

The marker CD49f was found to distinguish astrocytes from neuronal progenitors and neurons. At the genetic level, cells isolated using this marker showed activity of genes characteristic of both mature and immature astrocytes. However, when researchers looked at individual cells, they saw that CD49f was more enriched in mature astrocytes.

The hIPSCs-derived astrocytes expressing CD49f helped in neuronal growth, neural communication, provided metabolism support including glutamate uptake, and secreted molecules (called cytokines) in response to inflammation triggers.

We were excited to see that our stem-cell-derived astrocytes isolated with CD49f behaved the way typical astrocytes do: they take up glutamate, respond to inflammation, engage in phagocytosis which is like cell eating and encourage mature firing patterns and connections in neurons, said Valentina Fossati, PhD,the studys lead author.

CD49f expression was found to be specific for astrocytes in samples from both healthy and diseased human brains.

We looked at human brain tissue samples from both a healthy donor and a patient with Alzheimers disease and found that these astrocytes also expressed CD49f suggesting that this protein is a reliable indicator of astrocyte identity in both health and disease, Fossati added.

Researchers next focused on addressing the question of how astrocytes misbehave in disease.

They stimulated hIPSCs-derived cells with interleukin (IL)-1b and TNF-a, two molecules known to trigger the transition of astrocytes into a neurotoxic state (called A1 reactive astrocytes) in animal models. Cells reacted by secreting pro-inflammatory cytokines, including IL-6, IL-1 alpha, and ICAM-1.

Theseastrocytes lost their capacity to uptake (absorb) glutamate, a metabolite that is toxic to neurons. They also changed their morphology, becoming constricted instead of spreading out with long arms.

To assess whether reactive A1 astrocytes would damage neurons, the team grew neurons with stimulated and unstimulated astrocytes, or treated neurons with molecules produced by astrocytes.

Astrocytes in a reactive state were seen to decrease the electric activity of neurons and to increase their apoptosis a programmed process of cell death thats a form of suicide.

These findingsdemonstrate the specific neurotoxicity of A1 hiPSC-derivedastrocytes, the researchers wrote.

They also confirmprevious work in mice, where researchers observed that inflammation turns astrocytes neurotoxic. This work was led by Shane Liddelow, PhD, an assistant professor at the NYU Grossman School of Medicine and an author of the current study.

We observed in mice that astrocytes in inflammatory environments take on a reactive state, actually attacking neurons rather than supporting them, Liddelow said.

The latest work, the researchers concluded, showed that CD49f is a reactivity-independent,astrocyte-specific cell surface antigen that is present at allstages of astrocyte development in hiPSC-derived cultures.

Astrocytes isolated with this marker recapitulatein vitrocriticalphysiological functions, they continued, and following inflammatory stimulationbecome reactive, dysfunctional, and toxic, triggering neuronaldeath all of which opens a window for the study of their role in neurodegenerative disorders.

What we saw in the dish confirmed what Dr. Liddelow saw in mice: the neurons began to die, Fossati said. Observing this rogue astrocyte phenomenon in a human model of disease suggests that it could be happening in actual patients.

She and the others now look forward to using our new system to further explore the intricacies of astrocyte function in Alzheimers, multiple sclerosis, Parkinsons, and other diseases, in the hope it will point us toward new treatment opportunities that might slowor prevent neurodegeneration.

Patricia holds her Ph.D. in Cell Biology from University Nova de Lisboa, and has served as an author on several research projects and fellowships, as well as major grant applications for European Agencies. She also served as a PhD student research assistant in the Laboratory of Doctor David A. Fidock, Department of Microbiology & Immunology, Columbia University, New York.

Total Posts: 1,053

Patrcia holds her PhD in Medical Microbiology and Infectious Diseases from the Leiden University Medical Center in Leiden, The Netherlands. She has studied Applied Biology at Universidade do Minho and was a postdoctoral research fellow at Instituto de Medicina Molecular in Lisbon, Portugal. Her work has been focused on molecular genetic traits of infectious agents such as viruses and parasites.

Visit link:
Ability to 'Create' Astrocytes Supports Their Damaging Role in MS... - Multiple Sclerosis News Today

Photoaged Skin Therapy with Adipose-Derived Stem Cells – MedicalResearch.com

MedicalResearch.com Interview with:

Charles-de-SM.D., Ph.D.Rio de Janeiro, Brazil

MedicalResearch.com: What is the background for this study?

Response: Our clinical trial was based on our clinical skin observations in areas submitted to a lipotransfer previously, an ordinary practice in plastic surgery. These clinical observations lead us to investigate what will be the key element played in these findings. Our scientific support investigation addressed the Dardick1and Zuk, P2 studies, that demonstrated fibroblastic-like cells in adipose tissue with regenerative ability. Our clinical trial proposal is to investigate the adipose-derived stem cell (ADSC) role in the photoaged skin. The direct endpoint of the study was to assess the histological benefits provided by the subdermal ADSC injection. Mesenchymal stem cells were obtained from lipoaspirates, expanded in vitro, and introduced into the facial skin of 20 patients submitted after three to four months to a face-lifting surgery. In the retrieved skin, immunocytochemical and ultrastructural analysis quantified elastic matrix components, cathepsin-K, metalloprotease MMP-12, and the macrophage M2 markers: CD68, CD206 and heme-oxygenase-1.An overview of the trial steps is described in the infographic.

MedicalResearch.com: What are the main findings?

Response:A full de novo formation of oxytalan and elaunin fibers was observed in the subepidermal region, with a reconstitution of the papillary structure of the dermal-epidermal junction. Elastotic deposits in the deep dermis were substituted by a normal elastin fiber network. The coordinated removal of the pathologic deposits of old elastic fibers and their substitution by the normal ones was concomitant with activation of cathepsin-K and MPP12, and with expansion of the M2 macrophage infiltration.

MedicalResearch.com: What should readers take away from your report?

Response: This study has demonstrated ADSC to remodeling the skin extra cellular matrix, mainly in the elastic system.

MedicalResearch.com: What recommendations do you have for future research as a result of this study?

Response: Based on these findings, the future of thisresearch line aims to create new possibilities in regenerative cell therapy not only in skin diseases, but also in other clinical applicability in the case of organs and tissues with reduction and / or alteration in the elastic system (ex: aneurysms, cardiac valve disease and others), with a better understanding of the mechanisms involved and the control of these processes.

MedicalResearch.com: Is there anything else you would like to add? Any disclosures?

Response: It is interesting to be able, in future studies, to evaluate other mechanisms involved and the duration of effects regenerative effects on skin treated with ADSC. Another question could be considered: optimized ADSC (quantity) / area with the tissue effect found. We have not any to disclosure. This study was developed by federal university of Rio de Janeiro-Brasil and Verona University-Italy

Citation:

Charles-de-S, Luiz M.D., Ph.D.; Gontijo-de-Amorim, Natale Ferreira M.D., Ph.D.; Rigotti, Gino M.D., Ph.D.; Sbarbati, Andrea M.D., Ph.D.; Bernardi, Paolo Ph.D.; Benati, Donatella Ph.D.; Bizon Vieira Carias, Rosana Ph.D.; Maeda Takiya, Christina M.D., Ph.D.; Borojevic, Radovan Ph.D. Photoaged Skin Therapy with Adipose-Derived Stem Cells, Plastic and Reconstructive Surgery: June 2020 Volume 145 Issue 6 p 1037e-1049e doi: 10.1097/PRS.0000000000006867

References:

The information on MedicalResearch.com is provided for educational purposes only, and is in no way intended to diagnose, cure, or treat any medical or other condition. Always seek the advice of your physician or other qualified health and ask your doctor any questions you may have regarding a medical condition. In addition to all other limitations and disclaimers in this agreement, service provider and its third party providers disclaim any liability or loss in connection with the content provided on this website.

1

Originally posted here:
Photoaged Skin Therapy with Adipose-Derived Stem Cells - MedicalResearch.com

Eradicating balding a step closer with new procedure in the cross hairs – The New Daily

For Australias balding community, letting your hair down is just an idiom.

But soon, it may be a reality.

In a breakthrough in the battle against baldness, researchers from the University of Pennsylvania have managed to grow skin that develops distinct layers, including hair follicles,from stem cells.

Scientists were already able to grow skin cells, but recreating the complex, multi-layered skin structure has been a major challenge.

As the largest human organ, the skin has multiple functions including temperature regulation and bodily fluid retention to the sensing of touch and pain that increases the difficulty of synthesising it, researchers say.

But over a four-to-five month period, researchers succeeded in growing complex skin cells and hair follicles, which were grafted onto mice.

More than half of the mice sprouted hair from the process.

Its a development that may also affect those with genetic skin disorders and cancers, as well as those with burns or wounds.

But those who are a little thin on the top shouldnt get excited too fast.

There are several major questions that remain before this approach can become a reality, researchers Leo Wang and George Cotsarelis say.

Several other aspects of the authors approach will also need to be optimised before it can move to the clinic.

The hairs that grew in the current study were small; in future, furtheroptimisation of culture conditions will be needed to form large scalp hairs.

However, the authors conclude: The work holds great promise of clinical translation we are confident that research will eventually see this promise realised.

Follow this link:
Eradicating balding a step closer with new procedure in the cross hairs - The New Daily

Follica Announces Positive Feedback From End of Phase 2 Meeting With FDA for Its Lead Program to Treat Male Androgenetic Alopecia – BioSpace

BOSTON--(BUSINESS WIRE)-- Follica, Inc. (Follica), a biotechnology company developing a regenerative platform designed to treat androgenetic alopecia, epithelial aging and other related conditions, today announced positive feedback from a meeting with the U.S. Food and Drug Administration (FDA) as the company prepares to advance its lead program into Phase 3 development following a successful safety and efficacy optimization study for the treatment of hair loss in male androgenetic alopecia announced in December 2019.

This press release features multimedia. View the full release here: https://www.businesswire.com/news/home/20200603005934/en/

Follicas approach, which is designed to stimulate the growth of new follicles and new hair, is being developed as a potential new option for the millions of people seeking treatments to grow new hair. (Graphic: Business Wire)

Follica plans to launch its Phase 3 program this year. Overall, approximately 280 patients will be enrolled, with efficacy assessed against two co-primary endpoints: visible (non-vellus) hair count and patient-reported outcomes on a pre-established scale. The randomized, controlled, double-blinded studies will be conducted in multiple centers across the U.S. A maximal use study to further understand the pharmacokinetics of the treatment will be conducted in parallel. The trial design is consistent with feedback from the FDA during the End of Phase 2 meeting.

In the U.S. alone, 47 million men are affected by progressive hair loss caused by androgenetic alopecia, a condition that is largely unresolved today, leaving many dissatisfied with the current available treatments and looking for a new alternative. Our recent safety and optimization study points to a new level of effect, enabled by our proprietary approach, which stimulates the growth of new follicles and new hair, said Jason Bhardwaj, chief executive officer of Follica. Were grateful to the FDA for their guidance as we prepare for our pivotal program, and we look forward to advancing the development of our treatment regimen, which has demonstrated strong potential to address the current need for those who seek treatment for androgenetic alopecia.

Follicas approach is based on generating an embryonic window in adult scalp cells via a series of short office-based treatments with its proprietary Hair Follicle Neogenesis (HFN) device. The scalp treatments, which last just a few minutes, stimulate stem cells and enable the growth of new hair follicles. A topical drug is then applied to enhance efficacy by growing and thickening new hair follicles and hair on the scalp.

Follica reported topline results from its safety and optimization study in December 2019. That trial was designed to select the optimal treatment regimen using Follicas proprietary HFN device in combination with a topical drug and successfully met its primary endpoint. The selected treatment regimen demonstrated a statistically significant 44% improvement of visible (non-vellus) hair count after three months of treatment compared to baseline (p < 0.001, n = 19). Across all three treatment arms, the overall improvement of visible (non-vellus) hair count after three months of treatment was 29% compared to baseline (p < 0.001, n = 48), reflecting a clinical benefit across the entire trial population and a substantially improved outcome with the optimal treatment regimen. Additionally, a prespecified analysis comparing the 44% change in visible (non-vellus) hair count to a 12% historical benchmark set by approved pharmaceutical products established statistical significance (p = 0.005).

In addition to the safety and optimization study, Follica has validated its approach in prior clinical studies using prototype HFN devices with different treatment parameters and therapeutic compounds. Follicas translational work builds on research by George Cotsarelis, M.D., who isolated and characterized the expression pattern of stem cells from a critical region of the follicle. An expert in epithelial stem cell biology, Dr. Cotsarelis is chair of the department of dermatology at the University of Pennsylvania and a co-founder of Follica.

About Androgenetic Alopecia Androgenetic alopecia represents the most common form of hair loss in men and women, with an estimated 90 million people who are eligible for treatment in the United States alone. Only two drugs, both of which have demonstrated a 12% increase of non-vellus hair count over baseline for their primary endpoints, are currently approved for the treatment of androgenetic alopecia1. The most effective current approach for the treatment of hair loss is hair transplant surgery, comprising a range of invasive, expensive procedures for a subset of patients who have enough donor hair to be eligible. As a result, there remains a significant need for safe, effective, non-surgical treatments to grow new hair.

About Follica Follica is a biotechnology company developing a regenerative platform designed to treat androgenetic alopecia, epithelial aging and other related conditions. Founded by PureTech (LSE:PRTC), a co-inventor of the current platform, and a group of world-renowned experts in hair follicle biology and regenerative medicine, Follicas experimental treatment platform has been shown to stimulate the development of new hair follicles and hair in three previously conducted clinical studies. The companys proprietary treatment is designed to induce an embryonic window via a device with optimized parameters to initiate hair follicle neogenesis, the formation of new hair follicles from epithelial (skin) stem cells. This process is enhanced through the application of a topical compound. Follica completed a safety and efficacy optimization study in 2019, and its Phase 3 program in male androgenetic alopecia is expected to begin in 2020. Follicas technology is based on work originating from the University of Pennsylvania that has been further developed by Follicas internal program. Follicas extensive IP portfolio includes IP exclusively licensed from the University of Pennsylvania as well as Follica-owned IP.

1 Olsen EA et al, J Am Acad Dermatol. 2002 Sep;47(3):377-85Olsen EA et al, J Am Acad Dermatol. 2007 Nov;57(5):767-74. Epub 2007 Aug 29Price VH et al, J Am Acad Dermatol. 2002 Apr;46(4):517-23Kaufman et al, J Am Acad Dermatol. 1998 Oct; 39(4):578-589

View source version on businesswire.com: https://www.businesswire.com/news/home/20200603005934/en/

Read more:
Follica Announces Positive Feedback From End of Phase 2 Meeting With FDA for Its Lead Program to Treat Male Androgenetic Alopecia - BioSpace

Hope Realized – CU Anschutz Today

Not anymore.

Thanks to significant philanthropic support from The Sprout Foundation, a Denver-area foundation funded by Suzanne and Bob Fanch, and gifts from others including Wag and Annalee Schorr, the Ehlers-Danlos Syndrome Center of Excellence was launched in 2019. The goal of the center is to eventually develop a cure for EDS at the Gates Center for Regenerative Medicine, while better addressing the clinical needs of patients today through specialty care at Childrens Hospital Colorado. The clinical components of this new center address the critical need for patient-centered, coordinated EDS care where physicians come together to agree on the treatment plan, rather than leaving the patient and their family to determine the course of action.

Calla Winchell, left, with her grandfather, Dr. Wag Schorr, and her mother, Dr. Kate Schorr.

Simultaneously, the Gates Center for Regenerative Medicine scientists are conducting leading-edge research with the ultimate goal of finding a cure. Today, this research is aimed at discovering the genetic underpinnings of hypermobile EDS and leveraging this information to develop future therapies for patients like Calla.

The Fanches said, Sprout Foundation has funded research and the outstanding staff at the Gates Center for Regenerative Medicine to accelerate the cure for this life-changing disorder and also for clinical care to patients through the EDS Center of Excellence.

Joining in this effort are Callas own grandparents, Wag Schorr, an accomplished nephrologist and 1963 CU School of Medicine alumnus, and his wife, Annalee.

An essential component of the EDS Center of Excellence is a translational research program, which leverages existing campus resources and partnerships, including the Gates Center for Regenerative Medicine and the Colorado Center for Personalized Medicine.

The research program at the Gates Center is led by Dennis Roop, PhD, director of the Gates Center, in partnership with Ganna Bilousova, PhD, and Igor Kogut, PhD. The program brings EDS patients genetic information from clinical visits to the Gates Center where researchers are working on future treatments for the condition. In this virtuous cycle, patients inform future therapies in the lab that, in turn, could ultimately change lives back in the clinic.

Calla is one of those patients. She is motivated by the possibility of improving EDS research.

In preclinical models, scientists are collecting stem cells from Calla and other patients that indicate a possible mutation. These studies, using multiple patients, allow for a more accurate portrait of the errors in DNA. Early findings at the Gates Center suggest that a possible mutation for the hypermobile form of EDS may have been identified. The hope is that this research will lead to a potential treatment in the coming years.

Scientists are growing skin cells in the lab using Callas stem cells, with the EDS gene mutations removed. If successful, the modified stem cells will hook onto sites of inflammation and grow new cells restoring function to damaged tissues and organs. It sounds like science fiction, but it could be a reality at the CU Anschutz Medical Campus in the years to come.

The research advances taking place at the Gates Center will ultimately mean incredible hope and healing for people like Calla, and others with rare genetic disorders, who are eager to regain their health and their independence.

Im thrilled, she said. Im excited to receive coordinated care and treatments that will help me get back to my life. What if I could go to the grocery store and not have to use a wheelchair anymore?

By bringing research together with clinical care, the EDS Center of Excellence is helping turn such possibilities into realities.

Callas care plan is coordinated by a team of experts at the EDS multidisciplinary clinic at Childrens Hospital Colorado, led by the Medical Director of the Special Care Clinic Ellen Roy Elias, MD, in close collaboration with Kourtney Santucci, MD.

The clinic places the patient at the center of care, and brings forward all of the right health professionals required to determine a comprehensive care plan. In this model, the patient is seen by a team of specialists in a single day, with the goal of having a treatment plan at the end of the visit.

Callas grandfather, Dr. Schorr, says no more will Calla and others like her have to create a center of excellence for themselves as they traverse a complex and fragmented healthcare system to ensure their needs are met.

The pioneering work taking place at the EDS Center of Excellence began with Dr. Schorrs vision. In 2016, as a member of the Gates Center for Regenerative Medicine Advisory Board, Dr. Schorr approached director Dennis Roop and began laying the groundwork for research efforts in EDS, which he and Annalee funded later that year. Dr. Schorrs vision and commitment made it possible to develop the EDS Center of Excellence as a place to realize scientific advances in EDS research.

CU is poised for another breakthrough in medicine, said Dr. Schorr. I believe that EDS patients will soon have access to effective treatments, and possibly even a cure. If we are precise with our research and resources, we can resume our place at the forefront of the medical world. Thats our responsibility.

Another must, said Dr. Schorr, is to empower visionaries in their fields to pave the path toward new discoveries and major medical advances.

Leading this charge is CU School of Medicine Dean and Vice Chancellor for Health Affairs John Reilly, Jr., MD. Dean Reilly said, One of the great advantages of having our pediatric hospital partner, Childrens Hospital Colorado, and a research entity like the Gates Center on campus is the opportunity to collaborate. By bringing some of the best minds together to lead the next generation in EDS research, we get remarkable innovation, and leading-edge treatments and care. What our philanthropic partners have built here is inspiring, and together we are determined to bring hope to patients and their families. It has been exciting to see two families with a long friendship come together with a shared goal to create a center that will have a positive effect on so many patients and families.

With each new discovery at the EDS Center of Excellence, lives will improve through better care and better health. Each new discovery brings new opportunity for people with EDS to live lives they never knew they could have.

Guest Contributor: Courtney Keener, CU Anschutz Office of Advancement.

Read the original:
Hope Realized - CU Anschutz Today

Dermal Regeneration Matrix Device Market to Exhibit Increased Demand in the Coming Years – Lake Shore Gazette

Skin is the largest organ of the human body. It is composed of three layers: epidermis-the outermost layer; dermis-contains sweat glands, hair follicles and connective tissue and hypodermis-made up of fat and connective tissue. The main functions of the skin includes protection, sensation and regulation. The skin acts as a barrier and provides protection against harmful chemicals, radiation, microorganism and changing environmental conditions. It also helps regulate body temperature and maintain fluid balance. Skin is an extensive network of nerve cells and contains various receptors to detect changes in the environment such as touch, pain, heat and cold. Damage to skin due to burn or trauma can disrupt all the vital functions performed by the skin.

Currently, topical antibiotics, skin grafting, wound dressings and tissue-engineered substitutes are available in the market that are used to treat skin-related disorders. A skin graft can be done by natural substitute such as amniotic membrane, potato peel or artificial material that includes synthetic polymer sheet, polymer foam or spray. These substitute helps in the healing process. Skin regeneration refers to the regrowth of the damaged skin from the remaining tissue. Stem cell therapy has a vital application in skin regeneration.

Get Free Sample Copy With Impact Analysis Of COVID-19 Of Market Report @https://www.persistencemarketresearch.com/samples/12166

Dermal regeneration matrix device provides an appropriate environment that is necessary for the proliferation and differentiation of skin cells. It helps in triggering the bodys own repair mechanism by cell signaling, that drive the matrix environment in wound healing process. Dermal regeneration matrix device is used to treat skin burns and is also finds application in reconstructive surgery for contractures (scars). The dermal regeneration matrix device is placed over the damaged skin which provides an environment for regeneration of new skin and tissue. The matrix is made of cow collagen, silicone and shark cartilage.

In 1996, the U.S. Food and Drug Administration (FDA) first approved integra dermal regeneration matrix device for treatment of burn injuries. In 2002, dermal regeneration matrix device was approved for use in reconstructive surgery for burn scars. About 30 million people in the U.S. are suffering from diabetes, of which 15% experience a diabetic foot ulcer in their lifetime. In January 2016, FDA approved the use of dermal regeneration matrix for treatment of chronic diabetic foot ulcers (DFU). The usage of dermal regeneration matrix device is expected to expand the growth of dermal regeneration matrix device owing to increase usage in chronic foot ulcer.

Technological advancement and continued research in the development of artificial skin promises to bring more products to the marketplace. Increasing adoption of the device and long-term benefits associated with its application are some of the factors expected to fuel growth of the global dermal regeneration matrix device market over the forecast period. However, less awareness among the consumers and high cost of device are some of the key factors that could hamper growth of the market.

The global dermal regeneration matrix device is segmented on the basis of source, application, end user and geography.

You Can Buy This PMR Healthcare Report From Here @https://www.persistencemarketresearch.com/checkout/12166

On the basis of source, the global dermal regeneration matrix device market is segmented into cow collagen, silicone and shark cartilage. On the basis of end user, the global dermal regeneration matrix device market is segmented into hospitals and dermatology centers. The hospital segment is expected to contribute significantly to the total market in terms of market share. According to World Health Organization, over 265,000 deaths are caused due to burns each year. The majority of the burn cases occur in low and middle-income countries. Injuries such as traffic collisions, falls, burns, drowning, poisoning and others are expected to kills around five million people worldwide. Thus, the demand for dermal regeneration growth matrix is expected to be high in the low and middle-income countries over the forecast period.

On the basis of region, the global dermal regeneration matrix device market is segmented into five key regions: North America, Latin America, Europe, Asia Pacific and Middle East & Africa.

Some of the major players in the global dermal regeneration matrix device market include Integra LifeSciences Corporation, Platelet BioGenesis, Avita Medical, Stratatech, Organogenesis Inc., Smith & Nephew, Inc., ACell Inc., Symatese and others.

View post:
Dermal Regeneration Matrix Device Market to Exhibit Increased Demand in the Coming Years - Lake Shore Gazette

Future Growth of Cosmetic Skin Care Market by New Business Developments, Top Companies and Forecast to 2026 – Bulletin Line

Cosmetic Skin CareMarketBusiness Insights and Updates:

The latest Marketreport by a Data Bridge Market Researchwith the title[Global Cosmetic Skin CareMarket Industry Trends and Forecast to 2026].The new report on the worldwide Cosmetic Skin CareMarketis committed to fulfilling the necessities of the clients by giving them thorough insights into the Market. The various providers involved in the value chain of the product include manufacturers, suppliers, distributors, intermediaries, and customers.The reports provide Insightful information to the clients enhancing their basic leadership capacity identified.Exclusive information offered in this report is collected by analysis and trade consultants.

Global cosmetic skin care market is set to witness a substantial CAGR of 5.5% in the forecast period of 2019- 2026.

Cosmetic skin care is a variety of products which are used to improve the skins appearance and alleviate skin conditions. It consists different products such as anti- aging cosmetic products, sensitive skin care products, anti- scar solution products, warts removal products, infant skin care products and other. They contain various ingredients which are beneficial for the skin such as phytochemicals, vitamins, essential oils, and other. Their main function is to make the skin healthy and repair the skin damages.Get PDF Samplecopy(including TOC, Tables, and Figures) @https://www.databridgemarketresearch.com/request-a-sample/?dbmr=global-cosmetic-skin-care-market

Thestudy considers the Cosmetic Skin CareMarketvalue and volume generated from the sales of the following segments:Major Marketmanufacturerscovered in the Cosmetic Skin CareMarketare:LOral, Unilever, New Avon Company, Este Lauder Companies, Espa, Kao Corporation, Johnson & Johnson Services, Inc., Procter & Gamble, Beiersdorf, THE BODY SHOP INTERNATIONAL LIMITED, Shiseido Co.,Ltd., Coty Inc., Bo International, A One Cosmetics Products, Lancme, Clinique Laboratories, llc., Galderma Laboratories, L.P., AVON Beauty Products India Pvt Ltd, Nutriglow Cosmetics Pvt. Ltd, Shree Cosmetics Ltd

Segmentation:Global Cosmetic Skin Care Market

By Product

By Application

By Gender

By Distribution Channel

Get Table of Contents with Charts, Figures & Tables @https://www.databridgemarketresearch.com/toc/?dbmr=global-cosmetic-skin-care-market

Based on regions, the Cosmetic Skin CareMarketis classified into North America, Europe, Asia- Pacific, Middle East & Africa, and Latin AmericaMiddle East and Africa (GCC Countries and Egypt)North America (United States, Mexico, and Canada)South America(Brazil, Argentina etc.)Europe(Turkey, Germany, Russia UK, Italy, France, etc.)Asia-Pacific(Vietnam, China, Malaysia, Japan, Philippines, Korea, Thailand, India, Indonesia, and Australia)

Market Drivers:

Market Restraints:

Key Developments in the Market:

Key Benefits for Cosmetic Skin CareMarket:

Enquire Here For Discount Or Cosmetic Skin CareMarket Report Customization@https://www.databridgemarketresearch.com/inquire-before-buying/?dbmr=global-cosmetic-skin-care-market

About Us:Data Bridge Marketresearch endeavors to provide appropriate solutions to the complex business challenges and initiates an effortless decision-making process Data Bridge set forth itself as an unconventional and neoteric Marketresearch and consulting firm with unparalleled level of resilience and integrated approaches. We are determined to unearth the best Marketopportunities and foster efficient information for your business to thrive in the Market.We ponder into the heterogeneous Markets in accord with our clients needs and scoop out the best possible solutions and detailed information about the Markettrends. Data Bridge delves into the Markets across Asia, North America, South America, Africa to name few.

Contact Us:Data Bridge MarketResearchUS: +1-888-387-2818UK: +44 208 089 1725Hong Kong: +852 819+2 7475Email: [emailprotected]

Here is the original post:
Future Growth of Cosmetic Skin Care Market by New Business Developments, Top Companies and Forecast to 2026 - Bulletin Line

The 15 Best New Products to Try in Isolation This Month – InStyle

My summer countdown usually starts on the first day of fall. However, with social distancing still in place and travel completely off the cards for the foreseeable future, it's tough to get excited about what's arguably the best time of year.

Throughout quarantine, beauty products have given me a little bit of comfort and madethe stressand challenges of our current reality seem more manageable.

But even though everyone'sdaily routines have changed and the beauty industry has been greatly impacted by COVID-19, brands haven't stopped launching new products.

RELATED:Shopping for Makeup Post COVID-19 Lockdown Will Never Be the Same

This month's just-launched and soon-to-launch makeup, skincare, and haircare products include a number of treatments that are perfect for taking a time out and indulging in a little TLC in isolation. Briogeo's repairing hair mask, HoliFrog's glow-boosting cleanser, and Gucci Westman'svelvety eyeshadows are just a few examples.

Ahead, 15 new beauty products to give yourself some extra self-care while stuck at home.

Read the original here:
The 15 Best New Products to Try in Isolation This Month - InStyle

Genetic features pave way for targeted BPDCN therapies – Dermatology Times

Researchers are learning more about genetic aberrations common in the rare but clinically aggressive hematological cancer blastic plasmacytoid dendritic cell neoplasm. There is one targeted therapy approved by the U.S. Food and Drug Administration: Elzonris (tagraxofusp-erzs, Stemline). However, more treatment options are needed to improve the cancers clinical outcome, according to a review published May 2020 in Critical Reviews Oncology/Hematology.1

Dermatologists might be the first providers to encounter patients with blastic plasmacytoid dendritic cell neoplasm because more than 70% of these patients have cutaneous lesions. Those lesions often are asymptomatic and vary in size. The skin lesions tend to have nodules, plaques or bruise-like areas, a brown to violet color and might be solitary or multifocal, according to the authors.

Blastic plasmacytoid dendritic cell neoplasm often originates from type 2 myeloid-derived resting plasmacytoid dendritic cell precursors. Recent research suggests providers can diagnose the cancer when patients express at least four of five plasmacytoid dendritic cell specific markers, CD4, CD56, CD123, TCL1 and BDCA-2, without expressing myeloid, T-cell or B-cell lineage markers.

Commonly, [blastic plasmacytoid dendritic cell neoplasm] is characterized by high CD123 expression, aberrant NF-B [nuclear factor-B] activation, dependence on TCF4-/BRD4-network, and deregulated cholesterol metabolism, they wrote.

Despite advancing knowledge about the cancer type, patients median overall survival remains at 12 to 14 months, according to the paper. Conventional treatment approaches include chemotherapy, radiotherapy and ultimately hematopoietic stem cell transplantation. The challenges with conventional therapies are while blastic plasmacytoid dendritic cell neoplasm is sensitive to some chemotherapy regimens, patient relapse is high at more than 60%. And many patients with blastic plasmacytoid dendritic cell neoplasm are too old or frail to have intensive chemotherapy or hematopoietic stem cell transplantation, according to the authors.

Recently, the most attractive agent for [blastic plasmacytoid dendritic cell neoplasm] is tagraxofusp, which is composed of the catalytic and translocation domains of diphtheria toxin (DT) fused to interleukin-3 (IL-3), the authors wrote.

Blastic plasmacytoid dendritic cell neoplasm cells overexpress interleukin-3 receptor subunit alpha (IL3RA, also called CD123). Elzonris, or tagraxofusp-erzs, is a CD123-directed cytotoxin given intravenously, which is used to treat blastic plasmacytoid dendritic cell neoplasm in adults and in pediatric patients 2 years and older.

Researchers reported in a study of 47 blastic plasmacytoid dendritic cell neoplasm patients published in 2019 in the New England Journal of Medicine that tagraxofusp led to clinical responses in untreated and relapsed patients.2 The overall response rate with tagraxofusp was 90% and the primary outcome of complete response and clinical complete response was 72% among the previously untreated patients. Overall response was 67% in the previously treated patients. Serious adverse events including capillary leak syndrome, hepatic dysfunction and thrombocytopenia were common, according to the NEJM paper.

More targeted therapies are needed to treat blastic plasmacytoid dendritic cell neoplasm, but many potential therapeutic agents are not advancing to clinical trials, according to authors of the paper in Critical Reviews Oncology/Hematology.

Common blastic plasmacytoid dendritic cell neoplasm characteristics are genetically heterogeneous and provide valuable drug targets, according to the authors.

Apart from aberrant activation of NF-B signaling pathway, which is highly dependent on TCF4- and BRD4- transcriptional networks, cholesterol metabolism deregulation and CD123 expression, defects of DNA damage repair and mitosis are new, potential common features of the cancer. Corresponding therapies might be promising, the authors wrote.

Venetoclax, anti-CD123 CAR-T, XmAb14045 and IMGN632 are in clinical trials for blastic plasmacytoid dendritic cell neoplasm. But the authors noted that bortezomib, lenalidomide, 5-aza and pralatrexate could easily be pushed to the front line of the cancers treatment.

Disclosures:

The authors report no relevant disclosures.

References:

1. Zhang X, Sun J, Yang M, Wang L, Jin J. New perspectives in genetics and targeted therapy for blastic plasmacytoid dendritic cell neoplasm. Crit Rev Oncol Hematol. 2020 May;149:102928.2. Pemmaraju N, Lane AA, Sweet KL, et al. Tagraxofusp in Blastic Plasmacytoid Dendritic-Cell Neoplasm. N Engl J Med. 2019;380(17):1628-1637.

Here is the original post:
Genetic features pave way for targeted BPDCN therapies - Dermatology Times

The Science in Skin Care Introducing Stem Cell Technology

When it comes to our skin care goals, one key benefit seems to rule them all anti-aging. While aging skin may be inevitable, taking preventative action now can reduce the visible signs of aging long-term. Two key elements in this process are supporting collagen production and exfoliation. In other words, boosting the components of skin that keep it looking plump, while ridding the skins surface of older cells that can tend to build up over time.

In our efforts to support and maintain a youthful appearance, Stem Cellscan complement these two key elements and contribute to a significant improvement in overall skin tone and texture. Were taking it one step further by introducing you to NASA Stem Cell Technology.This revolutionary technology increases the overall efficacy of stem cells - allowing them to enhance our tried and trusted anti-aging skin care routines in order to seemingly defy gravity.

Stem Cells are naturally produced by our body and have the capacity to split and renew themselves over extended periods of time. This regeneration process is the key to skin rejuvenation as it supports the two key anti-aging elements - cell turnover and collagen production.

Like human stem cells, plant-derived stem cells have antioxidant properties and contain amino acids. Amino acids are the building blocks of proteins which promote cell renewal and maintain our skins overall hydration. However, when stem cells are created for topical application, they tend to flatten and lose efficacy under the influence of gravity.

If youd like to learn more about stem cells in skin care, check out this blog post!

Our Stem Cell Technology is a blend of cultured plant cells created without the influence of gravity, resulting in a powerful technology that is clinically proven to reduce the signs of aging. These cells more closely mimic those that we naturally produce, resulting in an overall increase in benefits.

NASA Stem Cell Technology was designed with a specific focus of targeting the signs of aging on the skin. Its key benefits include reducing the signs of lines and wrinkles, supporting the proliferation of skin cells and preventing/ delaying the visible signs of aging.

What could be more effective than incorporating a powerful technology such as NASAs Stem Cells in skincare? Coupling this key ingredient with clean and effective ingredients that are clinically proven to drive results. As our Director of Brand Development Heather Wilson says, Skin care products should be evaluated as a sum of their parts, not based on a single ingredient. That is why when creating products, we design them to include a combination of powerful actives, alongside a variety of botanicals, to create a formula that offers superior results versus a single ingredient.

Our Anti-Aging Collagen Serumpairs NASA Stem Cell Technology with Hyaluronic Acid, Collagen and a Peptide Complex to firm the appearance of lines and wrinkles to reveal a more vibrant, youthful complexion.

Hyaluronic Acid and Collagen act like a drink of water for the skin, hydrating and plumping the appearance of fine lines and wrinkles while Peptides support a healthy skin barrier and promote collagen production to reduce common signs of aging.

Suitable for all skin types, our Anti-Aging Collagen Serum:

This serum will provide a targeted treatment for deep lines and wrinkles due to its notable concentration of active ingredients in a safe and clean formula. It is lightweight, aroma-free and will appear milky white in color. It can be used both morning and night, after cleansing and before moisturizer.

If youre looking to deeply hydrate your skin and soften the look of fine lines and wrinkles:

Our Hyaluronic Acid Serum 85% pairs NASA Stem Cell Technology with a Multi-Molecular Hyaluronix and Niacinamide to deeply replenish the skin and reveal a more radiant, nourished and youthful complexion.

Multi-Molecular Hyaluronix is a form of Hyaluronic Acid comprised of various molecular weights to hydrate multiple layers of the skin while Niacinamide protects the skins natural barrier. Polyglutamic Acid adds another level of hydration while preventing water loss on the skin.

Suitable for all skin types, our Hyaluronic Acid Serum 85%:

This serum is lightweight, oil-free, aroma-free and will appear pale blue in color. For best results, we recommend shaking this product before use and apply twice a day, both morning and night. Follow up with your favorite moisturizer to seal in the benefits of this targeted treatment.

Our Dark Spot Corrector pairs NASA Stem Cell Technology with Glycolic Acid and Niacinamide to minimize sunspots and hyperpigmentation to reveal a more clear, bright complexion.

Glycolic Acidis an alpha hydroxy acid (AHA) that boosts cell turnover to exfoliate dulling skin cells and smooth the texture of skin. Niacinamide supports a healthy skin barrier while decreasing the appearance of discoloration and redness.

Suitable for all skin types, our Dark Spot Corrector:

This corrector is lightweight, oil-free, aroma-free and vegan. While AHAs provide incredible resurfacing abilities for the skin and are often preferred over physical exfoliants, they can increase our skins sensitivity to the sun. It is recommended that this product is used at night and an appropriate SPF is worn during the day.

For additional tips + tricks on how to fight the first signs of aging check out How to Fight Your First Wrinkles from our Natural Notes.

Stem cells provide the perfect boost to your favorite skin care ingredients to target and treat your anti-aging concerns. So much so that weve launched an entire collection because of it! So, whether youre looking to plump fine lines, reduce hyper pigmentation, or simply prevent future signs of aging - weve got the perfectsolutionfor you!

The rest is here:
The Science in Skin Care Introducing Stem Cell Technology

10 Best Stem Cell Beauty Products On The Market Today

Fight the signs of premature aging with these stem cell skin care beauty products. A lot of companies claim to incorporate the benefits of plant and human stem cells, as well as components secreted by them, into the best stem cell beauty products on the market. Below, we present what appears (based on company claims) to be ten of the best products available today.

As a publisher of stem cell news, we havent traditionally wandered into the world of claims made by stem cell beauty products suppliers. For obvious reasons, we cannot guarantee the accuracy of the claims made by these companies or the presence of specific active agents within them.

However, we get approached daily with questions about this topic and know that people are seeking information about it from a source that: 1) Doesnt inflate the claims, and 2) Understands the science.

For this reason, we have decided to share with you what appear to be interesting skin care options, coupled with a healthy dose of warnings reminding you that the stated claims may or may not be accurate.

Kimera Labs makes the top of this list for numerous reasons. First, the companys science it is solid. Instead of being a supplier of beauty products, the company is a specialty contract research organization (CRO) focusing on regenerative medicine applications, including exosome purification. Exosomes are small vesicles (~30-100nm) that are secreted by nearly all cell types and act as intracellular mail.

Exosomes transfer DNA, RNA, and proteins to other cells, thereby altering the function of the other cells.

Second, the company has an FDA registered tissue facility in Miami, FL, where it develops pharmaceutical grade, exosome-based regenerative therapies. The company has a 6,000 sq. ft. facility in Miramar, Florida, that includes impressive features such asISO:9001/13485 certification, cleanrooms, and a variety of high-end scientific equipment.

Third, the company is run by Dr. Duncan Ross, a highly regarded scientist with a Ph.D. in Immunology from the University of Miami. Dr. Ross is also a Principal at The Kimera Society, a non-profit organization dedicated to the advancementof stem cells, regenerative medicine,and cancer immunotherapies.

For those seeking stem cell beauty products, the companys core offering is XoGlo, a product which provides growth and healing signals to guide the re-deposition of tissue and avoid the scarring that often accompanies burns or other skin damage. You can see an incredible Case Study from the company in which XoGlo was used to heal second-degree burns in a patient in approximately seven days. The product can also be used for general skin health and enhancement.

More information on the XoGlois available here.

According to the company, this facial cleanser is formulated with stem cytokines that promote the skins ability to heal itself, leaving softer and smoother skin. It also has essential fatty acids, detoxifying actives, antioxidants, and anti-inflammatory botanicals that deeply cleanse your skin of excess oil, impurities, and surface debris. This makes the skin smoother, more balanced, and hydrated.

Lifeline says that it offers a moisture serum with a formula consisting of proteins and peptides from pluripotent stem cells. It works by reversing skin aging signs and actively moisturizing the skin with its cucumber melon extracts. The serum primarily targets the reduction of wrinkles and fine lines.

At $105 for a 1 oz bottle, it is notable that the company does not mention how it sources pluripotent stem cells, leaving key questions about its active ingredients unanswered.

Heres another skin care serum on this list of stem cellbeauty products. This serum is enriched with a tissue nutrient solution (TNS) technology that reduces wrinkles and fine lines and improves skin texture and tone. TNS is formulated with matrix proteins, cytokines, soluble collagen, antioxidants, and growth factors that are essential to keeping skin healthy.

This regenerative eye creamcontains autokine-CM obtained from adult stem cells through mini-liposuction. This unique ingredient is composed of extracted cytokines, matrix proteins, and growth factors from adult stem cells that help improve the skins ability to heal. It also aids in synthesizing elastin and collagen production, thus reducing fine lines and wrinkles, improving skin tone and texture, and increasing epidermal thickness in the eye area.

Venus Skin introduced a stem cell therapy serum packed with bio-signals from bone marrow mesenchymal stem cells for stimulation of skin tissue repair and healing. This reverses aging signs and rejuvenates the feel and look of the skin. It also contains essential vitamins A, C, and E to normalize skin functions, promote collagen synthesis in the skin, and reduce the appearance of scars, respectively.

This hydrating mask possesses a stem cell culture technology that penetrates deep into the skin for intense and long-lasting hydration. This leaves the skin well-moisturized and supple. It also fills fine lines and wrinkles and restores parched skin, bringing skin moisture and smoothness back.

This intensive facial mist restores the skins elasticity and moisture with its fine liquid particles that immediately penetrate the skin. It contains APL stem cell-conditioned medium extracts that help regenerate, whiten, and hydrate the skin and minimize pores and wrinkles. The facial mist also has chamomile extracts that bring a soothing effect to the skin.

Skin Drink Phytoceuticals highlights three potent anti-aging skin care ingredients in this serum.PhytoCellTec is an ingredient that safeguards the skin stem cells longevity, fights off skin aging, and delays biological aging of cells. Derm SRC works on reducing wrinkles and fine lines, while Ellagi-C promotes skin elasticity and suppleness.

This snail serum boasts an epidermal growth factor ingredient that stimulates the skins stem cell growth and cell survival. It also has a snail mucus extract that refreshes and brightens the skin. Aside from that, the serum contains other natural ingredients, such as macadamia seed oil and hydrolyzed placenta extract, for skin hydration and nourishment.

Which of these components actually enhance skin health and complexion? Hard to say, but the ingredient list certainly is exotic.

With this list of the best beauty products, it can be tricky to know which ones will enhance skin health. Stem cells are becoming a common ingredient in skin products, but regulation of this area is sparse, making it important to be vigilant in your selection.

A steep price tag doesnt guarantee results. Claims of active ingredients do not guarantee they are present. Even the confirmed presence of an ingredient by third-party testing does not substantiate its claimed effect.

However, there are hundreds of user reviews for some of these products, so the possibility for these skin care products to improve the appearance of your skin does exist. Importantly, many of these stem cell beauty products contain an impressive range of other ingredients, so you could benefit from them due to effects unrelated to the claimed stem cell components.

When judging the efficacy of these products, the only clear answer is that you need to be your own study of one.

Let this infographic be your guide. Download it now and use it as a reference later.

If you found this article valuable, subscribe to BioInformantsstem cell industry updates.We are the industry leaders in stem cell research, with research cited byThe Wall Street Journal, Xconomy, AABB, andVogue Magazine.Bringing you breaking news on an ongoing basis, join nearly aa million loyal readers, including physicians, scientists, executives, investors, and philanthropists.

Do you have questions about whether a stem cell treatment could address your medical condition?

As the worlds largest publisher of stem cell industry news, we understandably cannot provide clinical treatments or advice. However, GIOSTAR can provide you with medical guidance and advice. In alignment with what we believe at BioInformant, it offers cutting-edge, extensively researched stem cell therapy options.

Click here toSchedule a Consultationor ask GIOSTAR your questions.

Read the original post:
10 Best Stem Cell Beauty Products On The Market Today

On the Origins of Modern Biology and the Fantastic: Part 19 Nalo Hopkinson and Stem Cell Research – tor.com

She just wanted to be somewhere safe, somewhere familiar, where people looked and spoke like her and she could stand to eat the food. Midnight Robber by Nalo Hopkinson

Midnight Robber (2000) is about a woman, divided. Raised on the high-tech utopian planet of Touissant, Tan-Tan grows up on a planet populated by the descendants of a Caribbean diaspora, where all labor is performed by an all-seeing AI. But when she is exiled to Touissants parallel universe twin planet, the no-tech New Half-Way Tree, with her sexually abusive father, she becomes divided between good and evil Tan-Tans. To make herself and New Half-Way Tree whole, she adopts the persona of the legendary Robber Queen and becomes a legend herself. It is a wondrous blend of science fictional tropes and Caribbean mythology written in a Caribbean vernacular which vividly recalls the history of slavery and imperialism that shaped Touissant and its people, published at a time when diverse voices and perspectives within science fiction were blossoming.

Science fiction has long been dominated by white, Western perspectives. Vernes tech-forward adventures and Wells sociological allegories established two distinctive styles, but still centered on white imperialism and class struggle. Subsequent futures depicted in Verne-like pulp and Golden Age stories, where lone white heroes conquered evil powers or alien planets, mirrored colonialist history and the subjugation of non-white races. The civil rights era saw the incorporation of more Wellsian sociological concerns, and an increase in the number of non-white faces in the future, but they were often tokensparts of a dominant white monoculture. Important figures that presaged modern diversity included Star Treks Lieutenant Uhura, played by Nichelle Nichols. Nichols was the first black woman to play a non-servant character on TV; though her glorified secretary role frustrated Nichols, her presence was a political act, showing there was space for black people in the future.

Another key figure was the musician and poet Sun Ra, who laid the aesthetic foundation for what would become known as the Afrofuturist movement (the term coined by Mark Dery in a 1994 essay), which showed pride in black history and imagined the future through a black cultural lens. Within science fiction, the foundational work of Samuel Delany and Octavia Butler painted realistic futures in which the histories and cultural differences of people of color had a place. Finally, an important modern figure in the decentralization of the dominant Western perspective is Nalo Hopkinson.

A similarly long-standing paradigm lies at the heart of biology, extending back to Darwins theoretical and Mendels practical frameworks for the evolution of genetic traits via natural selection. Our natures werent determined by experience, as Lamarck posited, but by genes. Therefore, genes determine our reproductive fitness, and if we can understand genes, we might take our futures into our own hands to better treat disease and ease human suffering. This theory was tragically over-applied, even by Darwin, who in Descent of Man (1871) conflated culture with biology, assuming the Wests conquest of indigenous cultures meant white people were genetically superior. After the Nazis committed genocide in the name of an all-white future, ideas and practices based in eugenics declined, as biological understanding of genes matured. The Central Dogma of the 60s maintained the idea of a mechanistic meaning of life, as advances in genetic engineering and the age of genomics enabled our greatest understanding yet of how genes and disease work. The last major barrier between us and our transhumanist future therefore involved understanding how genes determine cellular identity, and as well see, key figures in answering that question are stem cells.

***

Hopkinson was born December 20, 1960 in Kingston, Jamaica. Her mother was a library technician and her father wrote, taught, and acted. Growing up, Hopkinson was immersed in the Caribbean literary scene, fed on a steady diet of theater, dance, readings, and visual arts exhibitions. She loved to readfrom folklore, to classical literature, to Kurt Vonnegutand loved science fiction, from Spock and Uhura on Star Trek, to Le Guin, James Tiptree Jr., and Delany. Despite being surrounded by a vibrant writing community, it didnt occur to her to become a writer herself. What they were writing was poetry and mimetic fiction, Hopkinson said, whereas I was reading science fiction and fantasy. It wasnt until I was 16 and stumbled upon an anthology of stories written at the Clarion Science Fiction Workshop that I realized there were places where you could be taught how to write fiction. Growing up, her family moved often, from Jamaica to Guyana to Trinidad and back, but in 1977, they moved to Toronto to get treatment for her fathers chronic kidney disease, and Hopkinson suddenly became a minority, thousands of miles from home.

Development can be described as an orderly alienation. In mammals, zygotes divide and subsets of cells become functionally specialized into, say, neurons or liver cells. Following the discovery of DNA as the genetic material in the 1950s, a question arose: did dividing cells retain all genes from the zygote, or were genes lost as it specialized? British embryologist John Gurdon addressed this question in a series of experiments in the 60s using frogs. Gurdon transplanted nuclei from varyingly differentiated cells into oocytes stripped of their genetic material to see if a new frog was made. He found the more differentiated a cell was, the lower the chance of success, but the successes confirmed that no genetic material was lost. Meanwhile, Canadian biologists Ernest McCulloch and James Till were transplanting bone marrow to treat irradiated mice when they noticed it caused lumps in the mices spleens, and the number of lumps correlated with the cellular dosage. Their lab subsequently demonstrated that each lump was a clonal colony from a single donor cell, and a subset of those cells was self-renewing and could form further colonies of any blood cell type. They had discovered hematopoietic stem cells. In 1981 the first embryonic stem cells (ESCs) from mice were successfully propagated in culture by British biologist Martin Evans, winning him the Nobel Prize in 2007. This breakthrough allowed biologists to alter genes in ESCs, then use Gurdons technique to create transgenic mice with that alteration in every cellcreating the first animal models of disease.

In 1982, one year after Evans discovery, Hopkinson graduated with honors from York University. She worked in the arts, as a library clerk, government culture research officer, and grants officer for the Toronto Arts Council, but wouldnt begin publishing her own fiction until she was 34. [I had been] politicized by feminist and Caribbean literature into valuing writing that spoke of particular cultural experiences of living under colonialism/patriarchy, and also of writing in ones own vernacular speech, Hopkinson said. In other words, I had models for strong fiction, and I knew intimately the body of work to which I would be responding. Then I discovered that Delany was a black man, which opened up a space for me in SF/F that I hadnt known I needed. She sought out more science fiction by black authors and found Butler, Charles Saunders, and Steven Barnes. Then the famous feminist science fiction author and editor Judy Merril offered an evening course in writing science fiction through a Toronto college, Hopkinson said. The course never ran, but it prompted me to write my first adult attempt at a science fiction story. Judy met once with the handful of us she would have accepted into the course and showed us how to run our own writing workshop without her. Hopkinsons dream of attending Clarion came true in 1995, with Delany as an instructor. Her early short stories channeled her love of myth and folklore, and her first book, written in Caribbean dialect, married Caribbean myth to the science fictional trappings of black market organ harvesting. Brown Girl in the Ring (1998) follows a young single mother as shes torn between her ancestral culture and modern life in a post-economic collapse Toronto. It won the Aspect and Locus Awards for Best First Novel, and Hopkinson was awarded the John W. Campbell Award for Best New Writer.

In 1996, Dolly the Sheep was created using Gurdons technique to determine if mammalian cells also could revert to more a more primitive, pluripotent state. Widespread animal cloning attempts soon followed, (something Hopkinson used as a science fictional element in Brown Girl) but it was inefficient, and often produced abnormal animals. Ideas of human cloning captured the public imagination as stem cell research exploded onto the scene. One ready source for human ESC (hESC) materials was from embryos which would otherwise be destroyed following in vitro fertilization (IVF) but the U.S. passed the Dickey-Wicker Amendment prohibited federal funding of research that destroyed such embryos. Nevertheless, in 1998 Wisconsin researcher James Thomson, using private funding, successfully isolated and cultured hESCs. Soon after, researchers around the world figured out how to nudge cells down different lineages, with ideas that transplant rejection and genetic disease would soon become things of the past, sliding neatly into the hole that the failure of genetic engineering techniques had left behind. But another blow to the stem cell research community came in 2001, when President Bushs stem cell ban limited research in the U.S. to nineteen existing cell lines.

In the late 1990s, another piece of technology capturing the public imagination was the internet, which promised to bring the world together in unprecedented ways. One such way was through private listservs, the kind used by writer and academic Alondra Nelson to create a space for students and artists to explore Afrofuturist ideas about technology, space, freedom, culture and art with science fiction at the center. It was wonderful, Hopkinson said. It gave me a place to talk and debate with like-minded people about the conjunction of blackness and science fiction without being shouted down by white men or having to teach Racism 101. Connections create communities, which in turn create movements, and in 1999, Delanys essay, Racism and Science Fiction, prompted a call for more meaningful discussions around race in the SF community. In response, Hopkinson became a co-founder of the Carl Brandon society, which works to increase awareness and representation of people of color in the community.

Hopkinsons second novel, Midnight Robber, was a breakthrough success and was nominated for Hugo, Nebula, and Tiptree Awards. She would also release Skin Folk (2001), a collection of stories in which mythical figures of West African and Afro-Caribbean culture walk among us, which would win the World Fantasy Award and was selected as one ofThe New York Times Best Books of the Year. Hopkinson also obtained masters degree in fiction writing (which helped alleviate U.S. border hassles when traveling for speaking engagements) during which she wrote The Salt Roads (2003). I knew it would take a level of research, focus and concentration I was struggling to maintain, Hopkinson said. I figured it would help to have a mentor to coach me through it. That turned out to be James Morrow, and he did so admirably. Roads is a masterful work of slipstream literary fantasy that follows the lives of women scattered through time, bound together by the salt uniting all black life. It was nominated for a Nebula and won the Gaylactic Spectrum Award. Hopkinson also edited anthologies centering around different cultures and perspectives, including Whispers from the Cotton Tree Root: Caribbean Fabulist Fiction (2000), Mojo: Conjure Stories (2003), and So Long, Been Dreaming: Postcolonial Science Fiction & Fantasy (2004). She also came out with the award-winning novelThe New Moons Arms in 2007, in which a peri-menopausal woman in a fictional Caribbean town is confronted by her past and the changes she must make to keep her family in her life.

While the stem cell ban hamstrung hESC work, Gurdons research facilitated yet another scientific breakthrough. Researchers began untangling how gene expression changed as stem cells differentiated, and in 2006, Shinya Yamanaka of Kyoto University reported the successful creation of mouse stem cells from differentiated cells. Using a list of 24 pluripotency-associated genes, Yamanaka systematically tested different gene combinations on terminally differentiated cells. He found four genesthereafter known as Yamanaka factorsthat could turn them into induced-pluripotent stem cells (iPSCs), and he and Gurdon would share a 2012 Nobel prize. In 2009, President Obama lifted restrictions on hESC research, and the first clinical trial involving products made using stem cells happened that year. The first human trials using hESCs to treat spinal injuries happened in 2014, and the first iPSC clinical trials for blindness began this past December.

Hopkinson, too, encountered complications and delays at points in her career. For years, Hopkinson suffered escalating symptoms from fibromyalgia, a chronic disease that runs in her family, which interfered with her writing, causing Hopkinson and her partner to struggle with poverty and homelessness. But in 2011, Hopkinson applied to become a professor of Creative Writing at the University of California, Riverside. It seemed in many ways tailor-made for me, Hopkinson said. They specifically wanted a science fiction writer (unheard of in North American Creative Writing departments); they wanted someone with expertise working with a diverse range of people; they were willing to hire someone without a PhD, if their publications were sufficient; they were offering the security of tenure. She got the job, and thanks to a steady paycheck and the benefits of the mild California climate, she got back to writing. Her YA novel, The Chaos (2012), coming-of-age novelSister Mine (2013), and another short story collection, Falling in Love with Hominids (2015) soon followed. Her recent work includes House of Whispers (2018-present), a series in DC Comics Sandman Universe, the final collected volume of which is due out this June. Hopkinson also received an honorary doctorate in 2016 from Anglia Ruskin University in the U.K., and was Guest of Honor at 2017 Worldcon, a year in which women and people of color dominated the historically white, male ballot.

While the Yamanaka factors meant that iPSCs became a standard lab technique, iPSCs are not identical to hESCs. Fascinatingly, two of these factors act together to maintain the silencing of large swaths of DNA. Back in the 1980s, researchers discovered that some regions of DNA are modified by small methyl groups, which can be passed down through cell division. Different cell types have different DNA methylation patterns, and their distribution is far from random; they accumulate in the promoter regions just upstream of genes where their on/off switches are, and the greater the number of methyl groups, the lesser the genes expression. Furthermore, epigenetic modifications, like methylation, can be laid down by our environments (via diet, or stress) which can also be passed down through generations. Even some diseases, like fibromyalgia, have recently been implicated as such an epigenetic disease. Turns out that the long-standing biological paradigm that rejected Lamarck also missed the bigger picture: Nature is, in fact, intimately informed by nurture and environment.

In the past 150 years, we have seen ideas of community grow and expand as the world became more connected, so that they now encompass the globe. The histories of science fiction and biology are full of stories of pioneers opening new doorsbe they doors of greater representation or greater understanding, or bothand others following. If evolution has taught us anything, its that nature abhors a monoculture, and the universe tends towards diversification; healthy communities are ones which understand that we are not apart from the world, but of it, and that diversity of types, be they cells or perspectives, is a strength.

Kelly Lagor is a scientist by day and a science fiction writer by night. Her work has appeared at Tor.com and other places, and you can find her tweeting about all kinds of nonsense @klagor

Visit link:
On the Origins of Modern Biology and the Fantastic: Part 19 Nalo Hopkinson and Stem Cell Research - tor.com

Generation of self-organized sensory ganglion organoids and retinal ganglion cells from fibroblasts – Science Advances

INTRODUCTION

A ganglion is a cluster or group of nerve cells found in the peripheral nervous system (PNS) or central nervous system (CNS). They often interconnect with each other and with other structures in the PNS and CNS to form a complex nervous network. There are three groups of ganglia in the PNS, which are the dorsal root ganglia (DRG), cranial nerve ganglia, and autonomic ganglia, and two types of ganglia in the CNS, which are the basal ganglia in the brain and retinal ganglion in the retina. Unlike other ganglia, which are essentially cell clusters, retinal ganglia consist of a layer/sheet of dispersive retinal ganglion cells (RGCs). Diverse types of neurons in the somatosensory ganglia such as DRG are specialized for different sensory modalities such as proprioception, mechanoreception, nociception (i.e., pain perception), thermoception, and pruriception (i.e., itch perception) (1, 2). Similarly, there are numerous subtypes of RGCs that are specialized for transmitting from the retina different visual information (e.g., color, contrast, and motion direction) to the central visual system in the brain (3). In the human, a variety of pain, itch, neurological, and degenerative disorders affect sensory ganglia (SGs) and RGCs. Mutations in the FXN (frataxin) and IKBKAP genes, for example, result in debilitating Friedreichs ataxia and familial dysautonomia, respectively (4, 5). Dominant gain-of-function mutations in the sodium channel Nav1.7 gene SCN9A, which is expressed in sensory neurons, are linked to two severe pain syndromesinherited erythromelalgia and paroxysmal extreme pain disorder, while its recessive loss-of-function mutations cause dangerous congenital insensitivity to pain (6). Recently, peripheral SG dysfunction has also been linked to tactile sensitivity and other behavioral deficits associated with the autism spectrum disorders (7). Both genetic and environmental risk factors contribute to glaucoma, which is a leading cause of blindness worldwide and characterized by progressive degeneration of RGCs and the optic nerve (8).

Despite the difference in morphology and embryonic origin, somatosensory and retinal ganglia share extensive overlap of gene expression and we proposed more than two decades back that both might also share genetic regulatory hierarchies (9, 10). This assumption has largely turned out to be the case. During embryogenesis, somatosensory ganglion neurons arise from the multipotent neural crest (NC) cells through a process of cell migration and coalescence (1). RGCs are also derived from multipotent retinal progenitor cells and destined to the ganglion cell layer by migration. It has been shown that the neurogenic bHLH transcription factors (TFs) Ngn1 and/or Ngn2 are involved in the determination of peripheral sensory neurons (11), and that the homeodomain TFs Isl1 and Brn3a or Brn3b are required for the specification and differentiation of different subtypes of neurons in the somatosensory and retinal ganglia (1217). Moreover, there is substantial functional redundancy between Ngn1 and Ngn2 as well as between Brn3a and Brn3b in the development of sensory neurons and RGCs (11, 18, 19).

Somatic cell reprogramming by defined TFs into sensory neurons provides a powerful strategy for studying mechanisms of SG development and sensory disease pathogenesis and for generating cells for patient-specific cell replacement therapy, drug screening, and in vitro disease modeling. It has been shown recently that nociceptor and other subtypes of sensory neurons can be directly induced from murine and human fibroblasts by Brn3a and Ngn1 or Ngn2 or by a combination of five TFs including Ascl1, Ngn1, Isl2, Myt1l, and Klf7 (20, 21). The induced sensory neurons express characteristic marker proteins and are electrically active and selectively responsive to various agonists known to activate pain- and itch-sensing neurons (20, 21). However, networked SG did not appear to be consistently generated in these cases, and it is unclear whether RGCs were induced by these combinations of TFs.

Given the advantages of organoids in studying developmental mechanisms and modeling and treating relevant diseases, we sought to generate ganglion organoids and RGCs from mouse and human fibroblasts using TFs controlling in vivo development of sensory and retinal ganglia. The extensive molecular homology between SG neurons and RGCs creates a dilemma as to how to distinguish these two types of neurons. In the past, several RGC markers including Brn3a, Brn3b, Isl1, Thy1.2, Sncg, Math5, Rbpms, and RPF-1 were used to identify RGCs induced from embryonic stem cells (ESCs), induced pluripotent stem cells (iPSCs), and somatic cells (2225). However, this is a questionable practice because although these markers are sufficient to identify RGCs within the retina, they are inadequate as specific markers for identifying induced RGCs (iRGCs), given their expression in SG and other CNS regions as well (10, 15). We thus carefully screened for RGC-specific markers by comparing expression patterns of numerous known markers in the retina and DRG. This analysis revealed Pax6 expression in RGCs but not in DRG and that RGCs can be identified as Pax6+Brn3a+ or Pax6+Brn3b+ double-positive cells. Equipped with this knowledge, we set to generate induced SG (iSG) organoids and iRGCs from fibroblasts by testing the combination of Ascl1, the pioneer neurogenic TF for somatic cell reprogramming of neurons (26), with a variety of SG and retinal TFs. This screen identified a triple-factor combination ABI (Ascl1-Brn3a/3b-Isl1) as the most efficient way to induce self-organized and networked iSG and iRGCs from fibroblasts.

Previous studies by our group and others have demonstrated that SGs and RGCs share similar transcriptional regulatory mechanism for their development, for instance, both Brn3 TFs (Brn3a and Brn3b) and Isl1 are involved in the specification and differentiation of DRG neurons and RGCs (1214, 16). More recently, Ascl1 has been shown to play a pioneering role in induced neuron (iN) reprogramming from somatic cells (26). As a first step to generate iSG and iRGCs directly from somatic cells, we sought to induce SG neurons and RGCs from mouse embryonic fibroblasts (MEFs) by testing the combination of Ascl1 with each of 22 SGs and retinal TFs (Brn3b, Isl1, Math5, Ebf1, Pax6, Tfap2a, Nr4a2, Nrl, Crx, Ptf1a, Neurod1, Lhx2, Ngn1, Ngn2, Chx10, Sox2, Rx, Meis1, Foxn4, Otx2, Sox9, and Six3). When MEFs were infected with doxycycline (Dox)inducible Ascl1 and Brn3b (AB) or Isl1 (AI) lentiviruses and cultured in the neural differentiation medium containing Dox, they started to change morphology by day 7 and form visible neuronal clusters by day 14 (Fig. 1, C and D). This phenomenon did not occur when Ascl1 acted alone or was combined with each of the rest of 20 TFs (Fig. 1B). Neither did this happen when MEFs were infected with both Brn3b and Isl1 viruses or with only control green fluorescent protein (GFP) viruses (Fig. 1, A and E). When we combined Ascl1 with both Brn3b and Isl1 (ABI), they again induced morphological changes of MEFs but more importantly induced conspicuously more neuronal clusters than either the AB or AI double-factor combinations (Fig. 1, C, D, F, and N, and fig. S1, A and B), suggesting a synergistic effect between Brn3b and Isl1 in reprogramming MEFs into neuronal clusters.

(A to I) Morphological changes of MEFs infected with the indicated lentiviruses (A, Ascl1; B, Brn3b; I, Isl1) and cultured for 14 days. Networked iSGs were induced by combinations of Ascl1 with Brn3b (AB), Isl1 (AI), or both Brn3b and Isl1 (ABI), with the ABI triple-factor combination as the most efficient. Arrows point to the thick fasciculated nerve fibers interconnecting iSG. Scale bars, 160 m (A to F) and 80 m (G to I). (J to M) Scattered iNs and clustered iSG induced by AI, ABI, A, or BAM (Brn2 + Ascl1 + Myt1l) were immunolabeled for Tuj1 and counterstained with nuclear 4,6-diamidino-2-phenylindole (DAPI). Note the morphological differences of Tuj1-immunoreactive neurons between conditions. Scale bars, 40 m. (N) Quantification of iSG induced by single and combinations of TFs. MEFs (6 104) were seeded into each well of 12-well plates and infected with lentiviruses expressing the indicated TFs or GFP, and iSGs in each well were then counted at day 14 following virus infection. Data are means SD (n = 3). Asterisks indicate significance in one-way analysis of variance test: *P < 0.0001. (O) Snapshots of a time-lapse video showing how individual neurons induced by ABI self-organized into an iSG. The arrow, arrowhead, and asterisk indicate the positions of three individual iNs at different time points. Scale bar, 62.5 m. (P) Schematic indicating the outcome (iNs or iSG) of MEFs induced by BAM, AI, AB, or ABI.

The neuronal clusters induced by either double- or triple-factor combinations (AB, AI, and ABI) appeared to be interconnected by thick fasciculated nerve fibers and resemble SG plexus in morphology (Fig. 1, G to I) and thus were designated as iSG organoids. The iSG neurons and associated nerve fibers were highly immunoreactive for the neuronal marker Tuj1 (Fig. 1, J and K, and fig. S2, D to I). Tuj1 immunolabeling also showed that AI- and ABI-induced neurons mostly formed iSG, and only a small number of them were scattered outside the iSG (Fig. 1, J, K, and P). By contrast, Tuj1 immunoreactivity showed that Ascl1 alone induced neurons mostly with an immature morphology and that the BAM (Brn2, Ascl1, and Mytl1) combination induced mature neurons that were scattered instead of clustered (Fig. 1, L, M, and P, and fig. S2, A to C), consistent with previous reports (27). Therefore, we identified the AB, AI, and ABI combinations of TFs capable of inducing MEFs into iSG, with the ABI triple-factor combination as the most efficient.

To investigate how ABI-reprogrammed neurons are organized into iSG, we used long-term time-lapse microscopy to track them over time in culture. For this purpose, MEFs were prepared from the CAG-GFP transgenic mouse embryos (28) and induced by ABI for 10 days before time-lapse recording. Compared to MEFs, reprogrammed individual neurons appeared to be rounder and neurite-bearing and displayed much higher contrast and brighter GFP fluorescence (Fig. 1O and movies S1 and S2). Over a period of tens of hours, they first formed smaller cellular clusters via migration, which then coalesced into bigger and bigger clusters that resembled SG. We did not observe this self-organization phenomenon for neurons induced by Ascl1 (movies S3 and S4).

The induction of iSG by TFs from MEFs could be through direct cell conversion or might be mediated through an intermediate proliferative progenitor. To distinguish these possibilities, we pulse-labeled cells with 5-ethynyl-2-deoxyuridine (EdU) for 24 hours at day 14 of reprogramming with AI or ABI and found that almost no Tuj1-positive cells were labeled by EdU, whereas approximately 15% of Tuj1-negative cells (e.g., MEFs) were labeled (fig. S2, G to J and N to P). We then reprogrammed MEFs with ABI in the presence of EdU for 13 days starting from day 1 of reprogramming. In this case, only 6.1% of Tuj1-positive cells were labeled by EdU, whereas 73.1% of Tuj1-negative cells were labeled (fig. S2, J to M), suggesting that iSGs are most likely induced by direct cell transdifferentiation without undergoing a proliferative intermediate state. In agreement with these results, as determined by quantitative reverse transcription polymerase chain reaction (qRT-PCR) assays, we detected no increase of expression levels of the neural progenitor marker genes Nestin and Olig2 over the entire time course (from day 1 to day 12) of ABI reprogramming (fig. S2Q). Similarly, the expression of pluripotent factor genes Oct4, Klf4, and Nanog was not induced during the time course of ABI reprogramming (fig. S2R). Furthermore, immunostaining showed that from day 1 to day 12 of ABI reprogramming, no protein expression was seen for the neural progenitor marker Nestin, pluripotent progenitor markers Nanog and Oct4, or Sox2, a marker for both neural and pluripotent progenitor cells (fig. S2, S and T). Thus, iSGs are most likely induced by direct cell transdifferentiation without undergoing an intermediate state of neural or pluripotent progenitors.

Given the demonstrated functional redundancy and similar DNA binding and transcriptional properties between Brn3a and Brn3b (10, 18, 19), we investigated whether these two factors are interchangeable in somatic cell reprogramming. We tested whether Brn3a was able to replace Brn3b in reprogramming MEFs into iSG and found that this indeed was the case (Fig. 1N and fig. S3, A to I).

By immunofluorescent staining and qRT-PCR assays, we examined a variety of molecular neuronal markers, both general and cell type specific, to characterize the iSG reprogrammed from MEFs by ABI (Ascl1 + Brn3b + Isl1 or Ascl1 + Brn3a + Isl1). We found that they were highly immunoreactive for Tuj1 and Map2 (Fig. 2, A and O), two general neuronal hallmarks. They also expressed synapsin and Vamp (synaptobrevin) (Fig. 2, B and C), suggesting that the networked iSG neurons were capable of forming synapses and releasing synaptic vesicles. In the normal SG, the heavy neurofilament NF200 and intermediate neurofilament peripherin are expressed in the A-fiber and C-fiber neurons, respectively, and both were seen to be expressed in the iSG (Fig. 2, D, E, and P). Many neurons in the iSG were also immunoreactive for the vesicular glutamate transporters 1 and 2 (vGLUT1 and vGLUT2) (Fig. 2, F and G), consistent with the fact that peripheral sensory neurons are mostly excitatory glutamatergic neurons. As determined by qRT-PCR, these immunolabeling results were confirmed by the marked up-regulation of expression of Tuj1, Map2, NF200, vGlut1, vGlut2, and vGlut3 genes in the ABI-induced iSG compared to MEFs infected by GFP lentiviruses (Fig. 2W).

(A to P) iSGs induced by Ascl1, Brn3b, and Isl1 (A to N) or Ascl1, Brn3a, and Isl1 (O and P) were double-immunostained with the indicated antibodies and counterstained with nuclear DAPI. They were immunoreactive for Tuj1, Map2, synapsin, Vamp, NF200, peripherin, vGLUT1, vGLUT2, TrkA, TrkB, TrkC, c-Ret, TH, p75NTR, and Brn3a. Scale bars, 80 m (A) and 40 m (B to P). (Q to V) Sections from iSG induced by Ascl1, Brn3b, and Isl1 were immunostained with the indicated antibodies and counterstained with nuclear DAPI. Scale bars, 12.7 m. (W) qRT-PCR analysis showing that in MEFs infected with ABI (Ascl1 + Brn3b + Isl1) viruses, compared to those infected with GFP viruses, there was a significant increase in expression of the indicated genes, which represent general and subtype-specific sensory neuron markers. Data are means SD (n = 3 or 4). Asterisks indicate significance in unpaired two-tailed Students t test: *P < 0.05, **P < 0.001, ***P < 0.0001. (X) qRT-PCR analysis showing that in MEFs infected with ABI viruses, compared to those infected with GFP viruses, there was a significant increase in expression of the indicated genes, which represent nociception pathway genes of sensory neurons. Data are means SD (n = 3 or 4). Asterisks indicate significance in unpaired two-tailed Students t test: *P < 0.05, **P < 0.005, ***P < 0.0005. (Y) Quantification of Tuj1-positive neurons that express each of the three Trk receptors (TrkA, TrkB, or TrkC) individually or combined (TrkABC) in MEFs infected with the ABI viruses. Data are means SD (n = 3).

In the DRG, neurotrophin receptor expression marks subtypes of sensory neurons. For instance, TrkA is expressed by cutaneous nociceptive and thermoceptive neurons, TrkB by a subset of cutaneous mechanoreceptive neurons, and TrkC by proprioceptive neurons (1). In the iSG reprogrammed by ABI, qRT-PCR assays revealed that there was a significant up-regulation of TrkA, TrkB, and TrkC gene expression (Fig. 2W). Moreover, immunolabeling confirmed the presence of TrkA, TrkB, and TrkC proteins in both somas and nerve fibers of the induced ganglion neurons (Fig. 2, H to J). Each of the Trk receptors was found in approximately 30% of the iNs, and 87% of the iNs were labeled by costaining for all three Trk receptors (Fig. 2Y), suggesting that each Trk receptor was expressed in a distinct subpopulation of induced ganglion neurons. c-Ret and TH are expressed in subpopulations of nonpeptidergic nociceptors and C-low threshold mechanoreceptors, respectively (1, 2). Correspondingly, we observed expression of both proteins in the iSG and associated nerve fibers (Fig. 2, K and L). In addition, pan-sensory neuron markers Brn3a (for iSG induced by Ascl1 + Brn3b + Isl1) and the nerve growth factor (NGF) receptor p75NTR were also found in iSG neurons (Fig. 2, M and N). qRT-PCR validated the up-regulation of Brn3a and p75NTR expression in the iSG and additionally revealed up-regulation of CGRP, a marker for a subpopulation of peptidergic nociceptive neurons (Fig. 2W).

The TrkA-positive nociceptive neurons in the iSG were further characterized by qRT-PCR analysis. In the iSG, we found significantly up-regulated expression of receptor ion channel genes Trpv1/2/3 and Trpa1, which detect heat and cold, respectively (Fig. 2X) (29). There was also expression of P2X3, Bdkrb1, and Accn1/2, which are receptor genes responsible for damage sensing (Fig. 2X) (29). In addition, induced expression was observed for other pain perception pathway genes including sodium channel gene Scn11a, potassium channel gene Kcnq2, calcium channel genes Cacna1a and Cacna2d1, and neurotransmitter receptor genes Gria1 and Nk1r (Fig. 2X) (29). To further investigate whether distinct types of sensory neurons were aggregated together within the same iSG, we carried out immunostaining analyses of cryosections of ABI-induced iSG. Besides colabeling between Tuj1 and Brn3a in the same iSG, we found that peripherin+ and HuC/D+ neurons, P2X3+ and vGLUT2+ neurons, and TrkA+ and TrkB+ neurons coexisted in the same iSG (Fig. 2, Q to T). Moreover, we detected coexpression of three markers, such as TrkA, P2X3 and NF200, and TrkA, peripherin, and HuC/D, in the same iSG (Fig. 2, U and V), suggesting that individual iSGs are likely aggregated from distinct sensory neuron types.

Over the time course of ABI reprogramming, qRT-PCR assays showed that the expression of general neuronal marker genes Tuj1 and Map2 was progressively induced starting from day 3, whereas other sensory neuronal marker genes including Trpv2, TrkC, and Brn3a were not induced until day 6 or 9 (fig. S1, C to E). Consistent with this, Brn3a-immunoreactive cells did not emerge until day 6 with a mostly scattered pattern, but by day 9 or 12, they mostly coalesced into iSG (fig. S1G). Therefore, as expected, sensory neuronal markers were induced slightly later than general neuronal markers during ABI reprogramming. Concomitant with neuronal induction, the fibroblast marker genes Col1a1 and Twist2 were gradually down-regulated starting from day 3 (fig. S1F).

Immunostaining of iSG induced by AI or AB suggested that they also contained neurons that expressed typical sensory neuronal markers Tuj1, Map2, Dcx (doublecortin), synapsin, NF200, peripherin, vGLUT1, TrkA, TH, HuC/D, and Brn3a (fig. S3, J to S). Together, these data indicate that certain combinations of TFs (ABI, AI, and AB) are capable of reprogramming MEFs into iSG that contain proprioceptive, mechanoreceptive, nociceptive, and thermoceptive sensory neurons.

To assess the electrophysiological properties of neurons within and outside the iSG reprogrammed from MEFs by ABI or AI, we performed whole-cell patch-clamp recordings of cells with neuronal morphology (Fig. 3A). Following 9 days of induction, the recorded neurons (two of two) generated potassium currents and small sodium currents but no action potentials, suggesting that they were functionally immature. At 2 weeks, the great majority of neurons (34 of 37) had typical sodium and potassium currents and exhibited action potential responses (Fig. 3, B to F). Among them, most (70.3%) are multispiking neurons, and the rest (21.6%) are single-spiking (Fig. 3, B, C, E, and K), similar to those reprogrammed from human fibroblasts by Brn3a and Ngn1 or Ngn2 (21). The inward sodium current could be specifically blocked by tetrodotoxin (TTX) and recovered by its removal (Fig. 3, H to J). Moreover, consistent with the synapsin immunoreactivity (Fig. 2B and fig. S3L), some neurons (2 of 37) exhibited spontaneous postsynaptic currents (Fig. 3G), suggesting the formation of functional synapses between iNs. Therefore, the iSG neurons induced by ABI or AI display membrane and physiological properties of mature neurons.

(A) Micrograph showing a typical iSG neuron chosen for patch-clamp recording. (B to D) Current-clamp recordings revealed multiple action potential responses (multiple-spiking) of a differentiated iSG neuron under current injection (B and C). Voltage-clamp recordings of the same neuron indicated fast activated and inactivated inward sodium currents as well as outward potassium currents (D). (E and F) Current injection revealed a single action potential response (single-spiking) of an iSG neuron (E). Voltage-clamp recordings of the same neuron indicated fast activated and inactivated inward sodium currents as well as outward potassium currents (F). (G) Spontaneous postsynaptic currents recorded from a differentiated iSG neuron. (H to J) The sodium currents of an iSG neuron were completely blocked by TTX and were partially restored by its washout. (K) Observed ratios of iSG neurons that are multiple-spiking and single-spiking, or display no action potential (AP). (L to N) iSG induced by ABI and corresponding fluorescent signals after incubation with Fluo-8 AM. Scale bars, 20 m. (O to Q) Calcium changes indicated by fluorescent intensity in normal Ringers solution (O), 10 M capsaicin (P), and 100 mM KCl (Q). Scale bars, 20 m. (R) Representative calcium responses to 100 M menthol and 100 mM KCl. Calcium responses were calculated as the change in fluorescence (F) over the initial baseline fluorescence (F0). (S) Representative calcium responses to 10 M capsaicin and 100 mM KCl. (T and U) Scatter dot plots showing the positive responses of individual cells to menthol, capsaicin, or KCl. Data are means SEM (n = 19 to 44).

The nociceptive sensory neurons express ion channels Trpv1, Trpm8, and Trpa1, which respond to heat, cold, and noxious chemicals, respectively (29). By calcium imaging, we used specific agonists capsaicin (10 M) and menthol (100 M) for Trpv1 and Trpm8 to confirm the functional expression of these two channels in iSG neurons (20, 21). KCl (100 mM) was transiently perfused to monitor the functional viability of the cells at the beginning and end of recording. Only cells that showed responses to KCl were chosen for analysis. Nearly all the iSG clusters induced by ABI showed green fluorescence following incubation with the calcium indicator Fluo-8 AM (Fig. 3, L to N). We found that among all the recorded cells, 56.8% of them (25 of 44) responded to capsaicin and 70.4% (19 of 27) to menthol (Fig. 3, O to U), suggesting that a large number of iSG neurons express ion channels characteristic of nociceptive sensory neurons.

We investigated the ability of iSG neurons to survive and integrate in the DRG by microinjecting dissociated iSG neurons reprogrammed from CAG-GFP mouse embryos (28), into adult rat DRG explants (fig. S4A). Following 2 weeks of culture of the transplanted explants, we found that the GFP+ iSG neurons survived, spread, and integrated in the DRG and were immunoreactive for the pan-sensory neuron marker HuC/D (fig. S4B). Moreover, a large fraction of them were immunoreactive for TrkA, while a small portion expressed TrkB or TrkC (fig. S4, C to E), indicating that iSG neurons maintain subtype specificity in the DRG.

Consistent with their sensory neuron identity, after a week in culture, iSG neurons reprogrammed from Tau-GFP mouse embryos (30) spontaneously aggregated with rhodamine-labeled sensory neurons dissociated from E13.5 mouse DRGs to form DRG-like organoids interconnected by nerve fibers (fig. S4, N to Q). In contrast, when GFP+ iSG neurons were cocultured with P0 mouse skin cells, they did not co-aggregate with skin cells; instead, they projected to and innervated vimentin-immunoreactive epidermal cells with multiple terminal nerve endings (fig. S4, J to M), in agreement with the fact that DRG neurons normally innervate their peripheral targets in the epidermis.

Previous studies have demonstrated that peripheral SG neurons and RGCs share many common molecular hallmarks, making it difficult to distinguish these two types of sensory neurons in cell culture. During the past decade in stem cell research, a number of supposedly specific molecular markers have been used to identify differentiated or induced SG neurons and RGCs (2225); unfortunately, however, no efforts have been made to confirm the specificity of these markers, casting doubt on some of the previous conclusions. Because Brn3a, Brn3b, and Isl1 are TFs crucial for retinal cell development, in particular, RGC development (13, 14, 16), there is a possibility that they may also be able to reprogram MEFs into RGCs. We thus set out to identify molecular markers that can definitively distinguish RGCs from peripheral sensory neurons. We postulated that such unique identifiers could be single-molecule markers or a combination of multiple-molecule markers that must be present only in RGCs within the retina but not in peripheral sensory neurons or any other tissues.

In the mammalian retina, our early studies have identified Brn3a and Brn3b as the gold standard markers for RGCs, but meanwhile revealed their expression in peripheral SG and other CNS areas (10, 15). In the mouse, immunolabeling of retinal and DRG sections confirmed the specificity of Brn3a and Brn3b in RGCs within the retina as well as their widespread expression in DRG neurons (fig. S5A), indicating that Brn3a and Brn3b alone cannot distinguish RGCs from DRG neurons outside the retina. Similarly, many other commonly used RGC markers including Thy1.2, RPF-1, Rbpms, HuC/D, Six6, Ebf, Isl1, Zeb2, Lmo4, Ldb1, and Sncg all displayed expression in the DRG (fig. S5A). Expressed in both RGCs and DRGs were also a number of sensory neuron markers including CGRP, peripherin, vGLUT2, vGLUT3, GABA, TrkA, TrkB, TrkC, and P2X3 (fig. S5A). Pax6 appeared to be the only exception among all the tested markers, which is expressed in RGCs and inner nuclear layer within the retina but absent from DRG (fig. S5A). Given the expression of Brn3a, Brn3b, Thy1.2, RPF-1, and Rbpms only in RGCs within the retina, a combination of Pax6 with any of these proteins could serve as a potential unique identifier for RGCs.

The uniqueness of double-positive markers was tested by immunolabeling sections of other CNS areas. Double-immunostaining showed that neuronal cells immunoreactive for both Pax6 and RPF-1, Thy1.2, Rbpms, HuC/D, or Tuj1, albeit absent from the DRG, were present not only in the retina but also in the spinal cord (Fig. 4A), precluding their use as specific RGC markers. The Isl1+Pax6+ double-positive cells were absent from the DRG and spinal cord but present within both the ganglion cell layer and inner nuclear layer in the retina (Fig. 4A), precluding also this combination as a specific RGC identifier. By contrast, Brn3a+Pax6+ and Brn3b+Pax6+ double-positive cells were exclusively RGCs in the retina and were not found in the DRG or spinal cord (Fig. 4A). Given the detection of Brn3a/Brn3b expression in the midbrain and cerebellum (10, 15), we investigated whether there were Brn3a+Pax6+ and Brn3b+Pax6+ double-positive cells in these two brain regions and found none at stages E13.5, P4, and P21 (Fig. 4A). Thus, these results together demonstrate that a combination of Pax6 and Brn3a or Brn3b double markers can serve as specific identifiers for RGCs.

(A) Cryosections from the indicated regions and stages of mice were stained by double immunofluorescence with the indicated antibodies and counterstained with nuclear DAPI. Arrows point to representative double-positive cells. GCL, ganglion cell layer; INBL, inner neuroblastic layer; INL, inner nuclear layer; IPL, inner plexiform layer; ONBL, outer neuroblastic layer; ONL, outer nuclear layer; OPL, outer plexiform layer. Scale bars, 40 m. (B to E) MEFs were infected with the ABI lentiviruses, cultured for 14 days, and double-immunostained with the indicated antibodies and counterstained with nuclear DAPI. Arrowheads in (D) indicate colocalized cells in the outlined region located outside the iSG. Scale bars, 40 m (B and C) and 20 m (D and E). (F to I) MEFs infected with the ABI lentiviruses and cultured for 14 days were dissociated and double-immunostained with anti-Brn3a and anti-Pax6 antibodies and counterstained with nuclear DAPI. Arrows indicate colocalized cells. Scale bars, 20 m. (J) qRT-PCR analysis of expression levels of the indicated genes (ex, exogenous; en, endogenous) in MEFs infected with ABI or GFP viruses (means SD, n = 3 or 4). *P < 0.05, **P < 0.001, ***P < 0.0001. (K and L) Quantification of DAPI- or Tuj1-positive cells that express Brn3a or Pax6 in MEFs infected with the ABI viruses (means SD, n = 4). (M) Quantification of Brn3a+Pax6+ iRGCs induced by ABI (means SD, n = 4).

We used the single-cell RNA sequencing (scRNA-seq) technology to further confirm the specificity of Brn3a+Pax6+ and Brn3b+Pax6+ double-positive markers. A Brn3b-GFP knockin mouse line was generated, and RGCs were enriched and sequenced by scRNA-seq. In addition, we isolated adult mouse DRG cells, which were then similarly sequenced. Clustering and expression analyses of the sequenced RGCs revealed that most of them expressed Pax6, Brn3a, or Brn3b and both Pax6 and Brn3a or Brn3b; in particular, the great majority of RGCs were positive for both Pax6 and Brn3a (fig. S5B). By contrast, there was a complete absence of DRG cells expressing both Pax6 and Brn3a or Brn3b, although Brn3a and Brn3b were present in most DRG cells (fig. S5B), consistent with the idea that a combination of Pax6 and Brn3a or Brn3b double markers can be used to distinguish RGCs from DRG cells.

To assess whether ABI and AI are able to induce iRGCs in addition to iSG, we immunostained ABI-reprogrammed MEF cells with antibodies against Tuj1, Brn3a, and/or Pax6. Double-labeling between Tuj1 and Brn3a or Pax6 showed that Brn3a-expressing cells were concentrated in the iSG, whereas the great majority of Pax6-expressing cells were distributed outside of the iSG and only few of them were seen in the iSG (Fig. 4, B and C). Moreover, all Pax6-positive cells coexpressed Brn3a and most of them displayed relatively weak Brn3a expression (Fig. 4, D and F to I), indicating that iRGCs were reprogrammed from MEFs by ABI. Similar to the distribution of endogenous RGCs that are spread throughout the RGC layer, the Brn3a+Pax6+ iRGCs were scattered and did not organize into clustered mini-ganglia (Fig. 4, C and D), unlike the induced peripheral SG neurons. Quantification of immunoreactive cells indicated that approximately 21.1% of all cells were induced by ABI into Brn3a+ neurons, whereas only about 2.6% of them were reprogrammed into Pax6+ cells (Fig. 4K). Furthermore, there were 93.1% of Tuj1+ cells coexpressing Brn3a, 10.5% of Tuj1+ cells coexpressing Pax6, and 12.6% Brn3a+ cells coexpressing Pax6 (Fig. 4, L and M), suggesting that only a small fraction of the ABI-reprogrammed neurons are Brn3a+Pax6+ double-positive iRGCs and that most of them are Brn3a+ iSG neurons. Similarly, a small number of Brn3a+Pax6+ iRGCs were induced by AI (fig. S3, T and U).

In agreement with the induction of a small proportion of iRGCs by ABI, immunostaining showed that some cells outside the iSG were positive for Thy1.2 (Fig. 4E). qRT-PCR assays revealed a significant up-regulation of several commonly used RGC marker genes including the endogenous Brn3b, Brn3a, RPF-1, Pax6, Sncg, HuC, and HuD in MEFs infected with ABI lentiviruses compared to those infected with GFP viruses (Fig. 4J), consistent with the induction of iRGCs by ABI from MEFs. Moreover, during the time course of ABI reprogramming, we were able to show by qRT-PCR assay that Pax6 expression was progressively induced starting from day 9 (fig. S1E).

We further characterized the iSG neurons and iRGCs by bulk and single-cell transcriptome profiling. First, we carried out bulk RNA-seq analysis of ABI- and GFP-transduced MEFs after 2 weeks of induction (Fig. 5A). Scatter plot and hierarchical cluster analyses showed that there were numerous genes whose expression was down-regulated or up-regulated in ABI-transduced compared to GFP-transduced MEFs (fig. S6, A to C, and table S1). We performed gene set enrichment analysis (GSEA) of the altered genes followed by network visualization (31), and one major group of clustered networks emerged (fig. S6D). This group encompasses only up-regulated genes that are enriched for GO (gene ontology) terms relevant to neural function and development such as synaptic signaling, synaptic vesicle, synapse organization, neurotransmitter transport, regulation of neurotransmitter levels, exocytosis, calcium ion binding, ligand-gated channel activity, neuron projection, axon, and nervous system development. These results are consistent with the induction of functional SG and retinal ganglion neurons by ABI from MEFs. In agreement with this and qRT-PCR assays (Figs. 2, W and X, and 4J), bulk RNA-seq confirmed up-regulation of many SG and retinal ganglion genes in ABI-transduced MEFs, including NF200, Brn3a, TrkB, vGlut3, Trpv1, P2X3, Gria1, Pax6, Sox11, Sncg, and Thy1 (fig. S6, E and F).

(A) Schematic illustration of the processes for bulk RNA-seq and scRNA-seq analyses. (B) t-distributed Stochastic Neighbor Embedding (t-SNE) plot of the 15 cell clusters generated from the sequenced single iSG neurons. (C to J) t-SNE plots colored by expression of the indicated conventional SG marker genes. (K) Violin plots showing expression patterns of the indicated conventional SG marker genes in single-cell clusters.

We separated ABI-induced iSG from MEFs by mild dissociation and filtering and then carried out scRNA-seq analysis of single iSG cells using the 10 Genomics Chromium platform (Fig. 5A) (32). After processing the sequencing data by the Cell Ranger software pipeline, we clustered the 3231 sequenced single cells into 15 clusters using the Seurat software package (Fig. 5B), which is an R toolkit for single-cell genomics (33). Investigation of gene expression patterns showed high levels of expression of general neuronal marker genes such as Tuj1, Tau, and Map2 in clusters 1, 3, 5, 8, and 11, whereas they are expressed much more weakly in the rest of the clusters (fig. S7, A and C to E). By contrast, many of the previously identified MEF marker genes (34) including Klf4, Mmp2, and Postn have, in general, an opposite expression pattern, displaying little expression in clusters 1, 3, 5, 8, and 11 but obvious expression in the rest of the clusters (fig. S7, A and F to H). Pseudotime trajectory of the sequenced cells constructed using Monocle (35) yielded three presumptive states along which Klf4 expression progressively decreases, while the expression of Tuj1, Tau, and Map2 progressively increases (fig. S7, I and J). Thus, in iSG induced from MEFs by ABI for 2 weeks, there are still some cells that express both neuronal and MEF markers, suggesting that MEFs undergo a transitional intermediate stage that exhibits both MEF and neuronal characteristics before completely reprogrammed into mature iSG neurons (fig. S7B). Consistent with this idea, there were many cells coexpressing both Tuj1 and the fibroblast marker gene vimentin in a number of the clusters (fig. S7K). By days 6 to 12 of ABI reprogramming, we also detected by immunolabeling some cells and nerve bundles that were immunoreactive for both Tuj1 and vimentin proteins (fig. S7L).

Consistent with the induction of iSG neurons, there is expression of NF200, peripherin, p75NTR, TrkB, TrkC, Trpv1, Trpv2, P2X3, Accn2, Kcnq2, Cacna1a, and CGRP in various clusters of sequenced iSG cells (Fig. 5, C to K). In particular, NF200, P2X3, Accn2, Kcnq2, and Cacna1a are primarily expressed in clusters 1, 3, 5, 8, and 11, and peripherin, Trpv1, and Trpv2 are mainly present in a small number of cells in clusters 1, 3, and 5 (Fig. 5, C, D, F to I, and K), indicating their expression in mature iSG neurons and their expression specificity. Many genes that are markers for both SG neurons and RGCs, such as Thy1, Sncg, Rbpms, Gap43, HuC, Sox11, Sox12, Zeb2, Brn3a, Brn3c, and RPF-1, are also expressed in various clusters of sequenced iSG cells (Fig. 6). However, the RGC marker Pax6 is only enriched in small cell clusters 12 and 13 and expressed in few cells in other clusters, consistent with the observation that only a very small number of iSG cells were immunoreactive for Pax6 (Figs. 4C and 6I). The Pax6+ cells in clusters 12 and 13 do not appear to be iRGCs because they lack expression of RGC markers Brn3a, Brn3c, and RPF-1 (Fig. 6I). In agreement with the observation that iRGCs were scattered and rarely present in iSG, there are only a small number of cells coexpressing both Pax6 and Tuj1, Thy1, Gap43, HuC, Sox11, Brn3a, or RPF-1, primarily in clusters 5 and 6 (Fig. 4C and fig. S8, A to H).

(A to H) t-SNE plots colored by expression of the indicated conventional RGC marker genes. (I) Violin plots showing expression patterns of the indicated conventional RGC marker genes in single-cell clusters.

We reprogrammed human skin fibroblasts (HSFs) into iSG with a mixture of the three individual ABI lentiviruses only at a low efficiency. To increase the reprogramming efficiency, we created a Dox-inducible lentiviral construct containing Ascl1, Isl1, and Brn3b in a single open reading frame (ORF) tethered by the P2A and T2A self-cleaving peptide sequences (Fig. 7A). HSFs infected by these single ABI-expressing viruses readily formed well-networked iSG in approximately 45 days in the neural differentiation medium (Fig. 7B). Immunostaining of these iSG showed that they contained typical sensory neurons expressing TUJ1, MAP2, NF200, PERIPHERIN, SYNAPSIN, VGLUT1, TRKA, TRKB, TH, and BRN3A (Fig. 7, C to K). Moreover, similar to MEFs, a small number of iRGCs were induced from HSFs by ABI that were immunoreactive for both PAX6 and BRN3A (Fig. 7, L and M). qRT-PCR assays showed that TUJ1 expression was gradually induced by ABI starting from day 10 but the more mature neuron marker gene MAP2 was not induced until day 20 (Fig. 7N). In contrast, the fibroblast marker genes COL1A1 and TWIST2 were progressively down-regulated starting from day 10 (Fig. 7O), concurrent with TUJ1 induction.

(A) Schematic of the lentiviral construct. (B to M) Networked iSG induced by ABI from HSFs (B) and iSG and iRGCs double-immunostained with the indicated antibodies and counterstained with DAPI (C to M). (N to Q) qRT-PCR assay showing the time course [days 1 (D1) to 20 (D20)] of expression changes of the indicated marker genes in HSFs infected with ABI or GFP viruses (means SD, n = 4). *P < 0.0001 for (N), (P), and (Q) and *P < 0.01, **P < 0.001, ***P < 0.0001 for (O). hES, human embryonic stem cell; hiNSC, human neural stem cell. (R) Schematic of EdU labeling schedule. (S to U) ABI-transduced HSFs were labeled by EdU for 29 days and colabeled for both TUJ1 and EdU before (S) and after dissociation (T). (U) Corresponding quantification (means SD, n = 4). *P < 0.0001. (V to X) ABI-transduced HSFs were labeled by EdU for 24 hours and colabeled for both TUJ1 and EdU before (V) and after dissociation (W). (X) Corresponding quantification (means SD, n = 4). *P < 0.0001. (Y, Z, and A) Current-clamp recordings revealed single action potential responses (single-spiking) of a differentiated iSG neuron (Y). Voltage-clamp recordings of the same neuron indicated fast activated and inactivated inward sodium currents as well as outward potassium currents (Z and A). The sodium currents of the iSG neuron were effectively blocked by TTX and were partially restored by its washout (A). (B and C) Current-clamp recordings revealed an iSG neuron with multiple action potential responses (multiple-spiking). (D) Spontaneous postsynaptic currents recorded from a differentiated iSG neuron. Scale bars, 80 m (B) and 20 m (C to M, S, T, V, and W).

To determine whether iSG induction was mediated by a pluripotent or neural progenitor intermediate, we investigated by qRT-PCR assay expression of pluripotent factor genes and neural progenitor marker genes during HSF reprogramming by ABI. We found no significant change in expression levels of pluripotent factor genes OCT4, KLF4, and NANOG during the reprogramming process (from day 1 to day 20) (Fig. 7P). Similarly, there was no induction of NESTIN and OLIG2 expression in the reprogramming process (Fig. 7Q), suggesting that iSGs were reprogrammed from HSFs by ABI without an intermediate state of pluripotent or neural progenitors. Consistent with this, by day 30 of reprogramming, almost no reprogrammed TUJ1+ neurons were labeled by EdU when EdU was added to the reprogramming cell culture for 29 days or 24 hours (Fig. 7, R to X), confirming that iSG reprogramming occurred in the absence of an intermediate state of proliferative progenitors.

The electrophysiological properties of reprogrammed human iSG neurons were evaluated by whole-cell patch-clamp recording. At day 60, most neurons (15 of 17) exhibited typical sodium and potassium currents and showed action potential responses (Fig. 7, Y, Z, and A). In addition, the inward sodium current could be specifically and completely blocked by TTX and partially recovered by its removal (Fig. 7A). Similar to mouse iSG neurons, some (4 of 17) were multi-spiking, while the others (11 of 17) were single-spiking (Fig. 7, Y, B, and C), although in human iSG single-spiking neurons appeared to be more abundant than those in mouse iSG (Fig. 3K). Among all neurons recorded from day 25 to day 39, a small fraction (4 of 44) displayed spontaneous postsynaptic activities (Fig. 7D), indicating the ability for human iSG neurons to form functional synapses, in agreement with their synapsin labeling (Fig. 7F). Thus, the human iSG neurons induced by ABI from HSFs have the physiological properties characteristic of mature neurons.

We further investigated the ability of human iSG neurons to survive and integrate in the DRG by microinjecting GFP-tagged human iSG neurons into adult rat DRG explants (fig. S4A). Two weeks after transplantation, we found that the GFP+ neurons survived and integrated in the DRG, and were all (99 of 99) immunolabeled by an anti-human nuclei antibody (fig. S4F), indicating that material transfer did not occur between the transplanted and host cells. The transplanted GFP+ cells were immunoreactive for pan-sensory neuron markers, and some of them were immunoreactive for TrkA, TrkB, or TrkC (fig. S4, G to I), suggesting that similar to mouse iSG neurons, transplanted human iSG neurons can also survive in the DRG and maintain sensory neuron subtypes.

Although scattered sensory neurons (iSNs) were previously induced from fibroblasts by TFs (20, 21), to our knowledge, this is the first time to demonstrate that self-organized iSG organoids can be consistently induced directly from somatic cells by defined TFs. The bHLH TF Ascl1 has been shown to be a pioneer neurogenic TF in converting fibroblasts into neurons in in vitro somatic cell reprogramming (26). However, the neurons reprogrammed by Ascl1 alone are mostly slow-maturing and excitatory (36). Addition of Brn2 and Myt1l (BAM) improved the reprogramming efficiency, maturing speed, and varieties of the iNs (27, 36, 37). The iNs induced by BAM were rather generic but motor neurons could be specifically induced when BAM were combined with four other TFs (Lhx3, Hb9, Isl1, and Ngn2) (38). Similarly, when trying BAM with other combinations, Wainger et al. (20) found that the combination of five factors (Ascl1, Myt1l, Ngn1, Isl2, and Klf7) could successfully convert fibroblasts into nociceptor neurons. Notably, all of these reprogramming formulas include Ascl1 as a key component. Alternatively, the bHLH TFs Ngn1 and Ngn2 were combined with Brn3a to reprogram fibroblasts into mature iSNs (21).

Our experiments in this study have demonstrated that the ABI TF combination is most effective in inducing MEFs into self-organized mini-SG, while the AI and AB combinations have a weaker activity (fig. S8J). Thus, Brn3a/3b appears to act synergistically with Isl1 to improve the induction efficiency of iSG organoids. As revealed by time-lapse microscopy, the larger iSG organoids are formed by cell migration and coalescing smaller cell aggregates. The mini-SG induced from both murine and human fibroblasts contain mature and functional sensory neurons. They exhibit typical inward sodium currents, which can be blocked by TTX and recover after TTX removal, and are a mixture of neurons displaying multiple-spiking action potentials or single-spiking action potential. They also show calcium responses to potassium chloride, capsaicin, and menthol. All these features closely resemble their endogenous counterparts.

The iSG neurons reprogrammed by ABI display extensive cell diversities in their expression of characteristic receptors, ligands, ion channels, neuropeptides, neurotransmitters, and so on, similar to the endogenous sensory neurons. In agreement with iSNs induced by Ngn1/2 and Brn3a (21), the iSGs contain roughly equivalent percentages (~30%) of TrkA+, TrkB+, and TrkC+ neurons, supporting the notion that Trk receptors may arise in a stochastic manner such that each donor cell has an approximately equivalent chance to express one of the Trk receptor genes. By bulk RNA-seq, scRNA-seq, and/or qRT-PCR analyses, we investigated the characteristic markers involved in sensory signaling pathways including transduction, conduction, and synaptic transmission of sensory signals. At the transduction level, we found up-regulated expression of genes responsible for perceptions to stimuli such as heat (Trpv1, Trpv2, Trpv3), cold (Trpa1), damage (P2X3, Bdkrb1), and touch (Trpc1, Trpc4, Asic2/Accn1, Accn2). Trpv1, also known as capsaicin receptor that is expressed mainly in the nociceptive neurons (29), has been shown to be present and functional in iSG neurons by capsaicin stimulation. The signaling conduction of sensory neurons is primarily mediated by sodium channels, which propagate the signals, and potassium channels, which usually act to reduce excitability. We found that the expression of many Na+ channels (Scn1a, 2a1, 2b, 3a, 3b, 7a, 11a) and K+ channels (Kcnq2, 4; Kcna2, 3, 4, 5, 6; Kcnb2, c1, d2, e4, f1, h2, j2, k3, s3, t1, etc.) were up-regulated in iSG neurons. For synaptic transmission, neurotransmitter receptors and presynaptic voltage-gated Ca2+ channels are two groups of important regulatory molecules. Correspondingly, the expression of a variety of neurotransmitter receptors (Nk1r, Nr3c2; Gria1, 2, 4; Grid1, k1, k2, k4, k5; Grin1, 2a, etc.) and Ca2+ channels (Cacna1a, 1b, 1d, 2d1, 2d2, 2d3; Cacnb1, g4, etc.) were significantly up-regulated in iSG neurons.

Apart from the molecular and electrophysiological properties, ABI-reprogrammed iSG neurons also have salient cellular and innervation characteristics of sensory neurons. For instance, when transplanted, they can survive, integrate, and maintain the nociceptive, mechanoreceptive, and proprioceptive subtypes in the DRG. Moreover, the iSG neurons exhibit strong affinity for endogenous DRG neurons and spontaneously aggregate with them to form interconnected DRG-like organoids in culture. In addition, we have demonstrated by coculture that the iSG neurons have the capacity to innervate the peripheral targets of sensory neurons, i.e., epidermal cells, indicating that the iSGs contain bona fide sensory neurons reprogrammed from fibroblasts by ABI.

Therefore, the combination of ABI TFs is able to reprogram murine and human fibroblasts into self-organized iSG organoids composed of heterogeneous sensory neurons, closely resembling the endogenous SG. Previously, Ascl1 in combination with Brn3a, Brn3b, or Brn3c was shown to induce iNs from MEFs (39). Although the sensory neuron identity of the iNs was not investigated, some of the data suggest the formation of iSG organoids by the Ascl1 and Brn3a combination (39). This is consistent with our work that showed that the AB combination enabled induction of iSG organoids, albeit fewer than those induced by the ABI combination (Fig. 1). Similarly, the data reported in a previous study also suggest the formation of iSG organoids by the nociceptive neurons reprogrammed from MEFs using a 5-TF combination (20). However, unlike the ABI combination, the 5-TF combination did not appear to induce iSG organoids from human fibroblasts (20), suggesting a difference in reprogramming capacity and/or efficiency by different combinations of TFs.

The peripheral ganglia, including cranial ganglia, DRG, trigeminal ganglia, enteric system ganglia, autonomic ganglia, and others, are derived from migrating NC cells. The NC is thought to be a unique cell population found in vertebrates and is initially induced at the neural plate border as a result of neural plate folding and fusion (40). After undergoing an epithelial-to-mesenchymal transition, the NC cells delaminate from the neuroepithelium and become highly migratory. Most NC cells migrate as a chain or group in a so-called collective cell migration, in which cell contact and cooperation allow them to migrate directionally. Guided by local cues and long-range chemoattractants, NC cells reach their destination and differentiate into ganglia and other tissue types.

Mutations in crucial genes controlling the migration and differentiation of NC cells may cause aganglionosis such as Hirschsprungs disease, which may occur by itself or in association with other genetic disorders such as Down syndrome, Waardenburg-Shah syndrome, Mowat-Wilson syndrome, or Bardet-Biedl syndrome (41). This group of genes includes RET, ZEB2, EDNRB, SOX10, and PHOX2B, and mutations of them or their regulatory sequences may increase the risk of Hirschsprungs disease more than 1000-fold (41). In our RNA-seq data, the expression of Ret, Zeb2, Ednrb, and Sox10 was significantly up-regulated in the iSG neurons, in agreement with their importance in the differentiation and formation of SG. Other known risk genesBbs4, Bbs10, Edn3, Gfra1, and Arvcf (41)were also significantly elevated in iSG. The hereditary sensory and autonomic neuropathies (HSANs) consist of several clinically heterogeneous disorders characterized by defective development and maintenance, and progressive degeneration of sensory and autonomic nervous systems. Mutations in the SPTLC1, WNK1, IKBKAP, and TRKA genes have been shown to cause HSAN types I to IV, respectively (42). In addition, loss-of-function mutations in SCN9A and PRDM12 result in congenital insensitivity to pain (6, 43). Indifference to pain appears to be desirable but risks the loss of a vital protective mechanism with dangerous consequences such as unknowingly chewing tongues and lips and damaging digits and joints. On the other hand, pain hypersensitivity reduces the quality of life and may increase susceptibility to chronic pain.

The ability to reprogram somatic cells into iSG organoids by ABI presents new possibilities for modeling sensorineural diseases, studying their pathogenesis, screening for counteractive drugs, and developing cell replacement therapies. For example, patient-derived iSG organoids may be used as an in vitro model for pain to screen and evaluate potential drug treatments. In the future, iSG organoids and neurons may also be used in transplantation as a cell replacement therapy for damaged or degenerated SG. In this respect, we found that transplanted iSG neurons were able to integrate and maintain the nociceptive, mechanoreceptive, and proprioceptive subtypes in the DRG. It has long been recognized that genetic factors are a major contributor to personalized pain perception and the efficacy of analgesic drugs (29). Generation of iSG organoids from autologous somatic cells may thus provide an exciting novel approach to model personalized pain and sensory pathology and help to achieve precision medicine for pain.

In this study, we made efforts to define specific molecular markers to identify RGCs both in vitro and in vivo. This is important because it is impossible to apply commonly used RGC markers to distinguish RGCs from SG neurons in vitro given the high molecular similarity between these two cell types. Since the 1990s, we have established the Brn3 family of TFs, Brn3a, Brn3b, and Brn3c, as the gold standards to identify RGCs in the retina (10, 15). However, Brn3 proteins are not unique to the retina but expressed in other sensory and CNS tissues as well, e.g., trigeminal ganglia, DRG, spiral ganglia, and midbrain (10, 15, 44). Apart from Brn3 proteins, Thy1.2, Sncg, and Rbpms are also commonly used as specific RGC markers. But here again, we show their abundant expression in DRG neurons. Therefore, although because of the spatial separation of the retina from SG in the organism, these so called RGC-specific markers are able to distinguish RGCs from SG neurons in vivo, they are unable to do so in vitro. Unfortunately, however, a number of previous studies used these supposedly RGC-specific markers to identify RGCs induced from ESCs, iPSCs, and somatic cells in vitro (2225), casting doubt on some of the arrived conclusions.

To avoid misidentifying iRGCs and iSG neurons in vitro, we screened for molecular markers that can definitively distinguish RGCs from SG neurons. A rigorous criterion was set that these unique identifiers should be single-molecule markers or a combination of multiple-molecule markers that must be present only in RGCs within the retina but not in SGs or any other tissues. Following a careful examination of a large number of known RGC and SG neuron markers, it became apparent that none of them alone were specific to RGCs. Further double-immunolabeling analysis indicated that a combination of Pax6 and Brn3a or Brn3b double markers satisfied the criterion of specific RGC identifiers. Brn3a+Pax6+ and Brn3b+Pax6+ double-positive cells were found exclusively in RGCs of the retina but not in the DRG, spinal cord, midbrain, or cerebellum, where Brn3a, Brn3b, or Pax6 is normally expressed. Moreover, scRNA-seq analysis confirmed Brn3a+Pax6+ and Brn3b+Pax6+ cells as RGCs and their complete absence in the DRG. Thus, we are able to define the combination of Pax6 with either Brn3a or Brn3b double protein markers as specific identifiers for RGCs. Armed with this knowledge, we found that ABI TFs had the capacity to reprogram MEFs into a small number of Brn3a+Pax6+ iRGCs, representing about 13% of all Brn3a+ neurons. Unlike iSG organoids resembling endogenous SG, iRGCs did not coalesce into clusters but remained scattered, similar to the dispersive distribution pattern of endogenous RGCs in the retina (fig. S8, I and J). Therefore, ABI-induced iSG and iRGCs maintain the morphology characteristic of their endogenous equivalents.

In summary, in a screen of multiple SG and RGC TFs, we have identified a triple-factor combination ABI as the most efficient combination to reprogram self-organized and networked iSG organoids from mouse and human fibroblasts. By immunostaining, qRT-PCR, whole-cell patch-clamp recording, calcium imaging, and bulk and scRNA-seq approaches, we are able to demonstrate that the iSG organoids display molecular and cellular features, subtype diversity, electrophysiological properties, and peripheral innervation patterns characteristic of peripheral SGs. Furthermore, using immunolabeling and scRNA-seq analyses, we have identified bona fide RGC-specific molecular markers to demonstrate that the ABI combination has the additional capacity to induce from fibroblasts a small number of iRGCs. Unlike the ABI-reprogrammed iSG organoids characteristic of endogenous SG, iRGCs maintain a dispersive distribution pattern resembling that of endogenous RGCs in the retina. The iSG organoids and iRGCs may be used to model sensorineural/retinal diseases, to screen for effective drugs and potentially, as cell-based replacement therapy.

All experiments on rodents were performed according to the IACUC (Institutional Animal Care and Use Committee) standards and approved by Sun Yat-sen University and Zhongshan Ophthalmic Center. The C57BL/6 mice were purchased from the Vital River Laboratories (Beijing, China).

The full-length ORFs of Brn3a, Brn3b, Isl1, Math5, Ebf1, Pax6, Tfap2a, Nr4a2, Nrl, Crx, Ptf1a, Neurod1, Lhx2, Ngn1, Ngn2, Chx10, Sox2, Rx, Meis1, Foxn4, Otx2, Sox9, or Six3 were subcloned into the Eco RI site of the FUW-TetO vector (45). In addition, by overlapping PCR subcloning, Ascl1, Isl1, and Brn3b were tethered by P2A and T2A self-cleaving peptide sequences into a single ORF, which was inserted into the same FUW-TetO backbone. Lentiviruses were prepared as previously described (34).

The MEFs were prepared as previously described (34). For isolation of mouse epidermal cells, P0 C57BL/6 mice were anesthetized with ice for 5 min and the brain was removed using a sterilizing razor in a 10-cm culture dish containing Hanks balanced salt solution (HBSS) (Gibco). The epidermis was isolated from the remaining tissue using a pair of fine-tip forceps under a dissection microscope, transferred into a fresh 6-cm culture plate containing 1 ml of 0.25% trypsin, thoroughly minced using a pair of surgical scissors and forceps, and incubated for 15 min at 37C in a CO2 incubator. Six-milliliter MEF medium containing Dulbeccos modified Eagles medium (DMEM)/High Glucose (HyClone) supplemented with 10% fetal bovine serum (Gibco), 1 penicillin/streptomycin (Gibco), 1 MEM nonessential amino acids (NEAA) (Gibco), and 0.008% (v/v) 2-mercaptoethanol (Sigma-Aldrich) was added into the plate to terminate the reaction. After being mixed using a 10-ml pipette, the digested tissue was transferred to a 15-ml fresh tube, centrifuged at 1000 rpm for 5 min, and resuspended in 5-ml fresh MEF medium. The isolated epidermal cells were expanded by culture in the MEF medium at 37C in a CO2 incubator. The HSFs were purchased from the American Type Culture Collection (CRL1502, 12-week gestation). MEFs, mouse epidermal cells, and HSFs were all maintained and expanded in the MEF medium.

To induce iSG and iRGCs from MEFs, 3 104 MEF cells (at passage 3) were cultured in 500-l MEF medium in a well of a 24-well plate containing a glass coverslip precoated with Matrigel (Corning). They were infected the next day with 500-l mixture of lentiviruses and fresh MEF medium in the presence of polybrene (10 g/ml). After 16-hour infection, the virus and medium mixture was removed. The cells were induced for 4 days in the neuron basic medium [(DMEM/F12 (1:1) (Life Technologies) supplemented with 1 B27 (Gibco) and basic fibroblast growth factor (bFGF) (10 ng/ml) (R&D Systems)] in the presence of Dox (2 ng/ml) (Sigma-Aldrich) and then for another 4 days in the neuron maintenance medium containing the neuron basic medium supplemented with insulin-like growth factor 1 (IGF-1) (100 ng/ml), brain-derived neurotrophic factor (BDNF) (10 ng/ml), and glial cell linederived neurotrophic factor (GDNF) (10 ng/ml) in the presence of Dox (2 g/ml). The medium was replaced with the neuron maintenance medium without Dox following the 8-day induction period. By 14 days after infection with Ascl1, Brn3b/3a, and Isl1 (ABI), Ascl1 and Brn3b/3a (AB), or Ascl1 and Isl1 (AI) lentiviruses, many visible neuronal clusters were formed.

With modifications, the HSFs were similarly induced. In brief, after virus infection, the human cells were cultured in the neuron basic medium with Dox for 10 days and then in the neuron maintenance medium without Dox for another 10 days. On day 21, the medium was replaced with the neuron mature medium, which is the maintenance medium supplemented with NGF (20 ng/ml), NT-3 (20 ng/ml), and 10 M forskolin. Thirty days after viral infection, many neuronal clusters were visible, which were usually smaller than those induced from MEFs. To improve the induction efficiency of the HSFs, we created a Dox-inducible lentiviral construct containing Ascl1, Isl1, and Brn3b in a single ORF as described above.

RNA extraction and qRT-PCR analysis were carried out as previously described (34). The qRT-PCR primers used are shown in table S2.

Immunostaining of tissue sections and cells was carried out as previously described (34, 46). The following antibodies (with dilution information) were used: mouse anti-Brn3a (Santa Cruz Biotechnology, sc-390780; 1:1000), mouse anti-Brn3a (Santa Cruz Biotechnology, sc-8429; 1:100), goat anti-Brn3b (Santa Cruz Biotechnology, sc-6026; 1:1000), rat anti-Thy1.2 (BD Biosciences, 550543), goat antiRPF-1 (Santa Cruz Biotechnology, sc-104627; 1:100), rabbit anti-Rbpms (PhosphoSolutions, 1830-RBPMS; 1:500), mouse anti-HuC&D (Life Technologies, A-21271; 1:500), rabbit anti-Pax6 (BioLegend, 901301; 1:2000), mouse anti-Pax6 (Developmental Studies Hybridoma Bank, Pax6; 1:1000), rabbit anti-Six6 (Sigma-Aldrich, HPA001403; 1:500), rabbit anti-Ebf (Santa Cruz Biotechnology, sc-33552; 1:1000), mouse anti-Isl1 (Abcam, ab20670; 1:2000), rabbit anti-Zeb2 (Santa Cruz Biotechnology, sc-48789; 1:1000), rat anti-Lmo4 (1:1000; (47), rabbit anti-Ldb1 (Abcam, ab96799; 1:1000), rabbit anti-Sncg (GeneTex, GTX110483; 1:200), rabbit anti-CGRP (Neuromics, RA24112; 1:200), rabbit anti-peripherin (Millipore, ab1530; 1:1000), rabbit anti-vGLUT1 (Synaptic System,135303; 1:500), mouse anti-vGLUT2 (Abcam, ab79157; 1:500), mouse anti-vGLUT3 (Sigma-Aldrich, SAB5200312; 1:500), rabbit anti-GABA (Sigma-Aldrich, A-2052; 1:1000), goat anti-TrkA (Abcam, ab76291; 1:500), rabbit anti-TrkA (Abcam, ab76291; 1:500), goat anti-TrkB (R&D Systems, AF1494; 1:500), goat anti-TrkC (R&D Systems, AF1404; 1:500), rabbit anti-P2X3 (Millipore, AB5895; 1:100), mouse anti-Tuj1 (Millipore, MAB5564; 1:500), rabbit anti-Tuj1 (Abcam, ab18207; 1:2000), mouse anti-Map2 (Sigma-Aldrich, M1406; 1:2000), rabbit anti-synapsin (Calbiochem, 574778; 1:500), goat anti-Dcx (Santa Cruz Biotechnology, sc-8066; 1:500), mouse anti-NF200 (Millipore, MAB5266; 1:500), rabbit anti-TH (Protos Biotech, CA-101bTHrab; 1:1000), rabbit anti-Vamp (Synaptic System, 104203; 1:500), rabbit anti-p75NTR (Abcam, ab8874; 1:500), mouse anti-c-Ret (Sigma-Aldrich, o4886; 1:1000), goat anti-GFP (Abcam, ab6673; 1:2000), rabbit anti-GFP (MBL, 598; 1:2000), chicken anti-GFP (Abcam, ab13970; 1:2000), rabbit anti-vimentin (Abcam, ab92547; 1:2000), and mouse anti-human nuclei (Millipore, MAB1281; 1:200). The secondary antibodies used included donkey anti-rabbit, donkey anti-goat, and donkey anti-mouse Alexa 488 immunoglobulin G (IgG), Alexa 594 IgG, Alexa 546 IgG, Alexa 647 IgG, or Alexa 594 IgM (1:1000; Invitrogen). 4,6-Diamidino-2-phenylindole (DAPI) (Invitrogen) was used for nuclear counterstaining. Images were captured with a laser scanning confocal microscope (Carl Zeiss, LSM700).

One day following infection with ABI lentiviruses, the MEFs were cultured in the presence of 10 M EdU (Life Technologies) for 13 days, or 13 days after infection with AI or ABI viruses, the MEFs were cultured for 24 hours in the presence of 10 M EdU. The cells were then fixed, and EdU staining was carried out according to the manufacturers instruction (Life Technologies). For HSF reprogramming by ABI, EdU was added to the reprogramming cell culture for 29 days starting from day 1 of reprogramming or for 24 hours starting at day 29. Images were captured with a confocal microscope.

For time-lapse recording, we used the JuLI Stage (NanoEntek) with a motorized stage, computer-controlled lens change, and a built-in incubator that supplied humidified 5% CO2 at 37C for live cell recording. MEFs (5 104) derived from the CAG-GFP transgenic mice (28) were induced for 10 days by infection with the ABI lentiviruses or Ascl1 lentiviruses in a well of a 12-well plate precoated with Matrigel. The plate was then placed into the incubator of the JuLI Stage for time-lapse recording for 50 hours. A series of pictures were taken from each well of the 12-well plate in a period of 50 hours under the control of the JuLI EDIT software, which can also edit and replay these pictures in a continuous mode like a movie.

To prepare single iSG cells, MEFs were infected with the ABI (Ascl1, Brn3b, and Isl1) lentiviruses and induced for 2 weeks. Following addition of 500-l Accutase (Millipore) into a well of a 12-well plate, neuronal clusters were suspended by gently pipetting up and down several times using a 1-ml pipette and transferred into a 70-m cell strainer (Falcon) to collect neuronal clusters. Most neuronal clusters attached to the Nylon membrane of the 70-m cell strainer, which was cut from the cell strainer using a pair of scissors and placed into a low-adhesion 6-cm plate containing 4-ml neuron basic medium. To separate the neuronal clusters from the Nylon membrane, the plate was shaken left and right 10 times. The neuronal clusters were then transferred into a 15-ml tube, centrifuged at 1000 rpm for 5 min, resuspended with 1-ml Accutase, and incubated for 5 min at 37C in a CO2 incubator. The neuronal clusters were dissociated into many single cells, which were subsequently used for injection of DRG explants, qRT-PCR, and scRNA-seq analysis.

After euthanization of the rat by the asphyxiation method (CO2 inhalation), the vertebral columns were isolated from the rest of the tissue using a pair of sharp scissors and washed three times with HBSS in a 10-cm culture dish. Both sides of the vertebral columns were mounted onto a surgical mat using needles, and a double cut was made using a pair of surgical scissors to expose the ventral side of the spinal cord. After removal of the spinal cord, DRGs were exposed in the contralateral dorsal spinal roots and pulled out using a pair of fine tweezers. They were collected into a 6-cm culture dish containing HBSS after removal of the attached excessive fibers and connective tissues under a dissection microscope. Four DRGs were transferred onto a Millipore Millicell-CM Low Height Culture Plate Insert using a 3-ml Pasteur pipette, and the rest of HBSS was removed using a 200-l pipette. Then, the insert was placed into a well of a six-well plate containing 1-ml DRG culture medium [BEM (Gibco) supplemented with 20 mM glucose, 1 KIT (Gibco), putrescine (16 ng/ml) (Sigma-Aldrich), 10 mM vitamin C (Sigma-Aldrich), NGF (20 ng/ml) (PeproTech), and 10 mM 5-fluoro-2-deoxyuridine (FDU) (Sigma-Aldrich)]. After being cultured for 1 day, each DRG was injected with 4 103 GFP-labeled single iSG cells. Two weeks following iSG cell injection, the explants were processed and immunostained as described above.

Whole-cell patch-clamp recordings of the iNs were performed with the EPC 10 USB amplifier (HEKA Electronik, Lambrecht, Germany) as previously described (34). Neurons induced from MEFs for 9 or 14 days or from HSFs for 25 to 60 days were used for patch-clamp recordings. In brief, coverslips with adhered cells were transferred into a recording chamber and bathed with Ringers containing 125 mM NaCl, 2.5 mM KCl, 1 mM MgSO4, 2 mM CaCl2, 1.25 mM NaH2PO4, 26 mM NaHCO3, and 20 mM glucose, bubbled with 95% O2 and 5% CO2. Cell responses were recorded with 6- to 9-megohm resistance pipettes that were filled with an internal solution containing 105 mM K-gluconate, 5 mM KCl, 5 mM NaOH, 15 mM KOH, 0.5 mM CaCl2, 2 mM MgCl2, 5 mM EGTA, 2 mM adenosine 5-triphosphate, 0.5 mM guanosine 5-triphosphate, 10 mM Hepes, and 2 mM ascorbate (pH 7.2). The cells and recording pipettes were viewed on a monitor that was coupled to a charge-coupled device camera (Evolve, Photometrics, Tucson, USA) mounted on an upright microscope. Oxygenated external solution was continuously perfused into the recording chamber at a flow rate of 1.5 to 2 ml/min by a peristaltic pump (LEAD-2, Longer Pump, Hebei, China). Capacitive transients were compensated via the Patch Master software (PatchMaster, HEKA), and the series resistance was compensated by ~50%. For current-clamp recording, a small, constant holding current was injected to maintain resting membrane potential (Vrest) at 70 mV and current pulses with a step size of 10 pA were applied to induce action potentials. Voltage-clamp recordings were performed on the same cells directly following current clamp recordings. A simple step protocol from 90 to +30 mV for 200 ms was applied to assess the voltage-gated sodium channels and voltage-gated potassium channels. TTX (Tocris, USA) was added to the bath solution to a final concentration of 0.5 M and perfused into the recording chamber for 5 min. After recording of the currents again, TTX was washed out, followed by the third time of recording.

The fluorescent probe Fluo-8 AM (AAT Bioquest, Sunnyvale, Canada) was used to detect the changes of intracellular calcium. As described above, MEFs were induced for 2 to 3 weeks to form neuronal clusters by infection with the ABI lentiviruses. Under dark environment, glass coverslips with adhered neuronal clusters were loaded with Fluo-8 AM (10 m) for 25 to 30 min at room temperature. After three rinses with Ringers solution, the coverslip was placed into a recording chamber. An upright microscope (Olympus, BX51W1) equipped with a mercury lamp with a 488-nm filter was used to excite Fluo-8. A digital camera (Hamamatsu Photonics, Japan) that was also equipped on the microscope was used to record the fluorescent signal. The software HCImage Live (Hamamatsu Corporation, USA) was used to control the camera and ImageJ for data analysis. Following a 30-s recording of the baseline (F0), 100 mM KCl was puffed to detect the activity of the cells. After a 2-min wash with Ringers, fluorescent signals were decreased to the baseline. Then, 100 M menthol or 10 M capsaicin was puffed to stimulate the iNs. KCl (100 mM KCl) was applied again after menthol/capsaicin to confirm the viability of the tested cells. Only the cells that responded to KCl two times successively were chosen for analysis.

Bulk RNA-seq analysis was performed with modification as previously described (48). Two weeks after infection of MEFs with lentiviruses, total RNA was extracted from GFP-transduced and ABI (Ascl1, Brn3b, and Isl1)transduced MEFs using the TRIzol reagent according to the manufacturers instruction. Ribosomal RNA was depleted before preparation of RNA-seq libraries, which were subsequently sequenced using an Illumina HiSeq 4000 sequencer (Biomarker Technologies, China). The obtained sequence reads were trimmed and mapped to the mouse reference genome (mm10) using HISAT2 (https://daehwankimlab.github.io/hisat2/), and gene expression and changes were analyzed using Cufflinks and Cuffdiff. Hierarchical cluster and scatter plot analyses of gene expression levels were performed using the R software (http://cran.r-project.org). GSEA was carried out as described (31), which was followed by network visualization in Cytoscape using the EnrichmentMap plugin (https://enrichmentmap.readthedocs.io/en/latest/).

Single iSG cells were prepared as described above. Single adult mouse DRG cells were prepared as described previously (49). In brief, DRGs were collected, transferred into a low-adhesion 6-cm pate with 2 ml of DMEM/F12 medium containing collagenase IV (1.25 mg/ml), and incubated at 37C in a 5% CO2 incubator for 50 min. Then, the medium was replaced with 2-ml DMEM/F12 medium containing 0.025% trypsin and incubated for 30 min. Following the addition of 2-ml DMEM/F12 medium containing 33% fetal bovine serum, all the medium was removed using a 10-ml pipette. After being washed three times with 2-ml HBSS, the DRGs were transferred into a 1.5-ml tube containing 1.2-ml DMEM/F12 and triturated by pipetting up and down several times using a 1-ml pipette to obtain single DRG cells. A Brn3b-GFP reporter mouse line was created using the CRISPR-Cas9 gene editing system to label adult RGCs by GFP, which were enriched by fluorescence-activated cell sorting. A more detailed description of this mouse line and RGC enrichment procedure will be published elsewhere.

The number and viability of prepared single cells were quantified using Countess II (Thermo Fisher Scientific, AMQAX1000). Next, single-cell libraries were generated with the Chromium Single Cell 3 V2 Chemistry Library Kit, Gel Bead & Multiplex Kit, and Chip Kit from 10x Genomics. In brief, cell suspension at concentration of 1.2 million/ml was loaded in a Single Cell 3 Chip along with the RT Single Cell 3 Gel Beads and the Partitioning oil, and Single Cell Gel Bead-In-Emulsions were generated in the Chromium Controller. Reverse transcription reaction was run to obtain complementary DNA (cDNA), which was amplified by PCR. To generate the libraries, Enzymatic Fragmentation, End Repair, and A-tailing Double Sided Size Selection were used to incorporate the barcodes and index read sequences. The libraries were qualified by bioanalyzer (Agilent Technologies) and quantified by a Qubit dsDNA High Sensitivity Assay kit (Invitrogen) and then sequenced on Illumina X Ten platform in 150 paired-end configuration.

Raw reads were processed using the 10x Genomics Cell Ranger pipeline (https://support.10xgenomics.com/single-cell-gene-expression/software/downloads/latest) with the mm10 as the reference. Cell Ranger can cluster the single cells, identify the marker genes of each cluster, and export a matrix with unique molecular identifier (UMI) values of each gene in a single cell. The R software package Seurat (https://satijalab.org/seurat, version 2.2) (33) was used for further analysis. Default parameters were used for most of the Seurat analyses. For the FeaturePlot function, max.cutoff was 0.5. The pseudotime trajectory analysis of iSG cells was performed using Monocle 2 (http://cole-trapnell-lab.github.io/monocle-release/) (35).

Statistical analysis was performed using the GraphPad Prism 7 and Microsoft Excel computer programs. The results are expressed as means SD for experiments conducted at least in triplicates. Unpaired two-tailed Students t test or one-way analysis of variance test were used to assess differences between two groups, and a value of P < 0.05 was considered statistically significant.

Acknowledgments: We thank E. Shiang for help with the artwork. Funding: This work was supported, in part, by the National Natural Science Foundation of China (81670862, 81721003, 31871497, 81870682, and 31700900), National Key R&D Program of China (2017YFA0104100, 2018YFA0108300, and 2017YFC1001300), National Basic Research Program (973 Program) of China (2015CB964600), Local Innovative and Research Teams Project of Guangdong Pearl River Talents Program, Science and Technology Planning Projects of Guangzhou City (201904020036 and 201904010358), China Postdoctoral Science Foundation (2019 M650223), and the Fundamental Research Funds of the State Key Laboratory of Ophthalmology, Sun Yat-sen University. Author contributions: D.X., K.J., Y.S., and M.X. conceived and designed the research. D.X., Q.D., Y.G., X.H., M.Z., J.Z., P.R., Z.X., Y.L., and Y.H. performed the experiments and analyzed the data. D.X., K.J., and M.X. interpreted the data and wrote the manuscript. All authors contributed to critical reading of the manuscript. Competing interests: The authors declare that they have no competing interests. Data and materials availability: All data needed to evaluate the conclusions in the paper are present in the paper and/or the Supplementary Materials. Additional data related to this paper may be requested from the authors. The RNA-seq and scRNA-seq data have been deposited in the NCBI Gene Expression Omnibus database under accession codes PRJNA595403 and PRJNA597624, respectively.

Read more:
Generation of self-organized sensory ganglion organoids and retinal ganglion cells from fibroblasts - Science Advances

Research at MDI Biological Laboratory explores novel pathways of regeneration and tumorigenesis – Bangor Daily News

BAR HARBOR Research by scientists at the MDI Biological Laboratoryis opening up new approaches to promoting tissue regeneration in organs damaged by disease or injury.

In recent years, research in regenerative biology has focused on stem cell therapies that reprogram the bodys own cells to replace damaged tissue, which is a complicated process because it involves turning genes in the cells nucleus on and off.

A recent paper in the journal Genetics by MDI Biological Laboratory scientist Elisabeth Marnik, Ph.D., a postdoctoral fellow in the laboratory of Dustin Updike, Ph.D., offers insight into an alternate pathway to regeneration: by recreating the properties of germ cells.

Germ cells, which are the precursors to the sperm and egg, are considered immortal because they are the only cells in the body with the potential to create an entirely new organism. The stem cell-like ability of germ cells to turn into any type of cell is called totipotency.

By getting a handle on what makes germ cells totipotent, we can promote regeneration by unlocking the stem cell-like properties of other cell types, said Updike. Our research shows that such cells can be reprogrammed by manipulating their cytoplasmic composition and chemistry, which would seem to be safer and easier than changing the DNA within a cells nucleus.

Using the tiny, soil-dwelling nematode worm, C. elegans, as a model, the Updike lab studies organelles called germ granules that reside in the cytoplasm (the contents of the cell outside of the nucleus) of germ cells. These organelles, which are conserved from nematodes to humans, are one of the keys to the remarkable attributes of germ cells, including the ability to differentiate into other types of cells.

In their recent paper entitled Germline Maintenance Through the Multifaceted Activities of GLH/Vasa in Caenorhabditis elegans P Granules, Updike and his team describe the intriguing and elusive role of Vasa proteins within germ granules in determining whether a cell is destined to become a germ cell with totipotent capabilities or a specific type of cell, like those that comprise muscle, nerves or skin.

Because of the role of Vasa proteins in preserving totipotency, an increased understanding of how such proteins work could lead to unprecedented approaches to de-differentiating cell types to promote regeneration; or alternatively, to new methods to turn off totipotency when it is no longer desirable, as in the case of cancer.

The increase in chronic and degenerative diseases caused by the aging of the population is driving demand for new therapies, said MDI Biological Laboratory President Hermann Haller, M.D. Dustins research on germ granules offers another route to repairing damaged tissues and organs in cases where therapeutic options are limited or non-existent, as well as an increased understanding of cancer.

Because of the complexity of the cellular chemistry, research on Vasa and other proteins found in germ granules is often overlooked, but that is rapidly changing especially among pharmaceutical companies as more scientists realize the impact and potential of such research, not only for regenerative medicine but also for an understanding of tumorigenesis, or cancer development, Updike said.

Recent research has found that some cancers are accompanied by the mis-expression of germ granule proteins, which are normally found only in germ cells. The mis-expression of these germ-granule proteins seems to promote the immortal properties of germ cells, and consequently tumorigenesis, with some germ-granule proteins now serving as prognosis markers for different types of cancer, Updike said.

Updike is a former postdoctoral researcher in the laboratory of Susan Strome, Ph.D., at University of California, Santa Cruz. Strome, who was inducted into the National Academy of Sciences last year, first discovered P granules more than 30 years ago. She credits Updike, who has published several seminal papers on the subject, with great imagination, determination and excellent technical skill in the pursuit of his goal of elucidating the function and biochemistry of these tiny organelles.

The lead author of the new study from the Updike laboratory, Elisabeth A. Marnik, Ph.D., will be launching her own laboratory at Husson University in Bangor, Maine, this fall. Other contributors include J. Heath Fuqua, Catherine S. Sharp, Jesse D. Rochester, Emily L. Xu and Sarah E. Holbrook. Their research was conducted at the Kathryn W. Davis Center for Regenerative Biology and Medicine at the MDI Biological Laboratory.

Updikes research is supported by a grant (R01 GM-113933) from the National Institute of General Medical Sciences (NIGMS), an institute of the National Institutes of Health (NIH). The equipment and cores used for part of the study were supported by NIGMS-NIH Centers of Biomedical Research Excellence and IDeA Networks of Biomedical Research Excellence grants P20 GM-104318 and P20 GM-203423, respectively.

We aim to improve human health and healthspan by uncovering basic mechanisms of tissue repair, aging and regeneration, translating our discoveries for the benefit of society and developing the next generation of scientific leaders. For more information, please visitmdibl.org.

Go here to read the rest:
Research at MDI Biological Laboratory explores novel pathways of regeneration and tumorigenesis - Bangor Daily News

Ageing: An expos on what really causes us to show our age – The South African

Most people are seeking the secret to anti-ageing, but did you ever wonder how the skin actually ages or how you could slow the process down?

Ageing is a natural process accompanied by a continuous alteration of the body. Your body produces visible changes in its structure, function and vulnerability to environmental stress and disease. Genetics, as well as the lifestyle we lead, play a big role in the ageing process.

Your skin is an organ, and its function is to regulate the excretion of metabolic waste products, regulate the bodies temperature as well as containing receptors for pain, tactile sensation, and pressure. Therefore, the health and appearance of your skin, like the health of your other organs correspond with your lifestyle and dietary habits, as well as with age-related factors such as the imbalance of hormones.

Ageing of the skin can be influenced by many factors including ultraviolet radiation, excess alcohol consumption, tobacco abuse, and environmental pollution.

What a lot of people dont realise is that as their body weight increases and their blood sugar levels rise, biochemical reactions interrupt the structural framework of their skin. With all these factors combined they lead to cumulative deterioration in the appearance of the skin as well as the function of the skin.

Within the skin ageing is associated with a loss of fibrous tissue, a slower rate of cellular renewal, and a reduced vascular and glandular network. The barrier function that maintains cellular hydration also becomes impaired. The subcutaneous tissue (known as the hypodermis or the third layer of the skin) flattens.

The rate at which these functions decline can be more than 50% by middle age depending upon ones genetic makeup, lifestyle and normal physiological functions within the skin. If we dont take action to support our skins intrinsic defence systems, the youthful qualities of our skin will deteriorate rapidly. Luckily for us, we can harness insights gathered through the latest scientific innovations and slow or potentially reverse the signs and symptoms of accelerated skin ageing.

Intrinsic skin ageing is primarily determined by genetic factors, hormonal imbalances and metabolic reactions like oxidative stress. Signs of intrinsic ageing include skin sagging, thinning and cracking, and the appearance of fine lines and wrinkles.

There are numerous external factors that affect the skin and cause signs and symptoms of premature ageing. Generally, most premature ageing is caused by over-exposure to the suns UV rays. However, there are other contributing factors, for example, atmospheric factors such as air pollution, visible light and infrared radiation. Lifestyle choices such as smoking, chronic stress and excessive alcohol consumption can lead to older-looking skin.

The most common signs of extrinsic ageing are thinning of the skin, laxity, fragility and the increased appearance of wrinkles.

As the skin is a visual organ, the beauty industrys main objective is to improve the appearance of skin with extensive topical treatments and products. However, often overlooked is the need to support the health and beauty of the skin from within.

Ideally one should centre their diet upon fruits, vegetables, whole grains, legumes, monounsaturated fats (like those found in olive oil), and a healthy ratio of omega-3 to omega-6 polyunsaturated fatty acids. Generally, consumption of shellfish, fish rich in omega 3 fatty acids, regular tea drinking, and greater consumption of fruits and vegetables have been known to be associated with improved skin health.

Gut health is crucial to healthy skin. The human skin hosts a variety of microorganisms, collectively known as the skin microbiota. Within the skin, there is a complex network of interactions between the microbes and cells. Friendly bacteria, such as Lactobacillus and Bifidobacteria are well researched for effectively treating infections, promoting healthy immunity, and reducing inflammation in the skin. Oral pre- and probiotics help to rebalance the skin microbiota and optimise the skin barrier function.

In addition, oral probiotics boost cellular antioxidant capacity and combat inflammation in general. Probiotics also help to neutralise toxic byproducts, defend the lining of the intestine, increase the bioavailability of some nutrients and reinforce the intestinal barrier against infectious microbes that may harm healthy skin.

Cosmeceuticals are topical products that exert both cosmetic and therapeutic benefits which have continued to evolve in order to ward off the signs of skin ageing. Some of the most popular topicals include exfoliating and depigmenting agents, antioxidants and regenerating products, such as peptides and stem cells.

Sunscreens (with dual protection against UVA and UVB in a photostable complex) are the most important topical as they protect us from the UV damage caused by the sun. Sun exposure is definitely one of the biggest contributing factors to premature ageing and is actually known as photo-ageing.

Another phenomenal topical is retinoids which have proven their safety and efficacy in reducing photo-damaged skin and are a popular treatment for anti-ageing. Retinoids help combat and reverse the visual effects of ageing, such as wrinkles, laxity, and discolouration. Retinoids accelerate cell turnover and can also improve blemishes and the appearance of pores.

The use of alpha-hydroxy acids (AHAs) has also been known to improve skin texture and reduce the signs of ageing by promoting the shedding of our superficial dead skin cells which in turn helps to restore hydration and a smoother texture. Whats nice about alpha-hydroxy acids is that they can pretty much treat any skin condition or concern because there are so many different types of acids. Theres literally something for everything. The most common ingredients used in product formulations and peels include citric acid, glycolic acid, lactic acid, malic acid, pyruvic acid as well as tartaric acid.

Antioxidants are being increasingly used in anti-ageing skincare. Topical antioxidants are effective in fending off damaging free radicals and reducing inflammation within the skin. A few popular ones used are ascorbic acid (vitamin C,) tocopherols (vitamin E,) alpha-lipoic acid and coenzyme Q10. Emerging natural antioxidants proving effective include EGCG (from green tea), resveratrol, Centella Asiatica (Gotu Kola,) proanthocyanidins (grapeseed,) curcumin, pomegranate, silymarin/silibinin (milk thistle), coffeeberry, melatonin, and marine-based ingredients.

Within the skin, the deterioration of collagen results in the formation of protein fragments, called peptides. These peptides are then recognised by collagen-producing cells, which respond by increasing collagen production in order to repair the damaged skin. However, as we age this positive feedback between skin breakdown and the initiation of new collagen formation becomes inefficient. Therefore by applying specialised peptides to your skin topically you can effectively trick collagen-producing cells into boosting collagen production. There are many other active ingredients used in topical products that are focused on anti-ageing among other things.

So basically all we need to do is protect the skin from the inside by consuming nutrient-packed foods as well as reducing our exposure to extrinsic factors that cause premature ageing along with using topical skincare products. Not as difficult as we may have thought, hey?

This content has been created as part of our freelancer relief programme. We are supporting journalists and freelance writers impacted by the economic slowdown caused by #lockdownlife.

If you are a freelancer looking to contribute to The South African,read more here.

Read this article:
Ageing: An expos on what really causes us to show our age - The South African

Archives