Category Archives: Somatic Stem Cells

A human accumulates as many mutations in 80 years as a mouse in its short life – EL PAS in English

Giraffes that weigh up to 1,200 kilograms accumulate as many mutations in their cells throughout their lives as naked mole-rats, which rarely weigh more than 35 grams. Similarly, living beings as long-lived as humans and as short-lived as rats reach the end of their days with a similar number of changes in the DNA of their cells. Recent research comparing different mammals shows that there is a kind of natural law that links the life expectancy of each species to mutagenesis. This connection could shed light on processes as complex as cancer or aging.

Since the 1950s, many scientists in the field have linked aging with various processes of cell deterioration. One such process is somatic mutation. These are genetic changes, between 20 and 50 a year in humans, that occur in the life cycle of the cell. They occur in healthy cells, although some of them can trigger tumors or other alterations. Occurring at the cellular level, somatic mutations should be related in some way to body mass and life expectancy. The reason why a correlation with the first would be expected is because in theory, the greater the mass, the greater the probability of cancer developing in some cell. On the other hand, if mutations play an important role in aging, they would theoretically have a connection to life expectancy. So researchers at the Wellcome Sanger Institute (United Kingdom) specialized in genomic research decided to study these genetic alterations in 16 species of mammals, looking for patterns that pointed to a common basis in the decline of life over time.

The research, published a few days ago in the journal Nature, is new for many reasons. One is because of its comparative analysis of species that differ significantly in size, life expectancy or diet. It is also new in its analysis of somatic mutations within cells, something impossible to do until very recently. It is also novel because of the object of its study: cells from a part of the colon (called intestinal crypts) that have the particularity that they all come from the same stem cell, making it possible to accurately calculate the number of mutations they contain. But, above all, it is new because of its findings, which may have implications for some of the fundamental questions about human health and other living beings.

If it depended only on the number of cells, a mouse would never develop a tumor... and you would expect all whales to die of colon cancer before the age of 30, simply because of their size

The main finding is that the number of mutations per year or mutation rate is adjusted to the life expectancy of each species. Although very diverse, this rate is inversely proportional to longevity. Thus, while humans, with their life expectancy of around 80 years, have an annual rate of 47 mutations in the cells of each intestinal crypt, mice, with a survival rate of only two years, suffer 796. At the end of their lifetime, all the animals under study accumulated a similar number of mutations. In addition, researchers found that three of the main mutagenesis processes identified, such as the substitution of one base for another in the sequence, are shared between the different species. So there is a connection between mutation and aging. Another very different thing is to know how that connection works.

Adrin Bez-Ortega is a researcher at the Wellcome Sanger Institute and, together with Alex Cagan, is the main author of this research. He cautions about connecting somatic mutations in the cell with the aging process of organisms. First, because we do not have data that would allow the results to be extended to other tissues beyond the colon. Second, because we have not studied it in other living beings that are not mammals. Even more importantly, they do not know the meaning of this relationship. We discovered a correlation between life expectancy and mutation rate, but we do not know if the mutation rate has evolved to adapt to the life expectancy of the animal or if the mutations affect this life expectancy, highlights the Spanish researcher. It is certain, he says, that there are other factors at play as well. For example, colon cells accumulate about 50 mutations a year, but suffer thousands of instances of cellular damage. These damages appear to also play a role in senescence.

The work confirms one of the most intriguing paradoxes in molecular biology. It was raised almost 50 years ago by the British epidemiologist Richard Peto. He observed that species were beyond logic in terms of the risk of suffering cancer. If tumors are the result of mutations or harmful changes at the cellular level, the more complex the cell, the more tumors there should be. But reality shows that the incidence of cancer in animals does not seem related to body mass. This new study confirms the so-called Peto paradox.

The incidence of cancer is similar between the species, even though they have differences in mass, notes Bez-Ortega. In general, larger animals tend to live longer. Since all cells have the same chance of developing cancer, the larger the cells, the greater the risk, one might think. But no. There is an incidence curve for cancer in humans that is known. If it depended only on the number of cells, a mouse would never develop a tumor, because it has far fewer cells and would not live long enough. Actually, they have an incidence similar to ours. And you would expect all whales to die of colon cancer before the age of 30, simply because of their size. But we see that evolutionary pressure has adjusted the rate of cancer, so that humans, elephants and mice have an almost identical curve once you control for their life expectancy. And we dont know why this happens, concludes the Spanish scientist.

The evolution of multicellular life requires carefully coordinating all cells so that each one plays exactly the right role in the organism. Somatic mutations can upset this balance

igo Martincorena heads research on somatic mutation in healthy and cancerous cells at the Wellcome Sanger Institute in the UK. He is also a senior author on the research published in Nature. Regarding the main findings of the work, he recognizes how intriguing it is. The evolution of multicellular life requires carefully coordinating all cells so that each one plays exactly the right role in the organism. Somatic mutations can upset this balance, as evidenced in the case of cancer, so the evolution of life has required controlling the accumulation of somatic mutations, he says in an email exchange. So the finding that the somatic mutation rate per year is much lower in species that live longer suggests that [these rates] are under evolutionary control.

Regarding the relationship between mutations and aging, Martincorena insists that senescence does not have a single cause. The most compelling hypothesis right now is that it is caused by the accumulation of various types of molecular damage in our cells, including somatic mutations, telomere shortening, protein aggregation, and epigenetic changes. We believe that these changes at the molecular level cause changes in our cells and tissues that give rise to the typical manifestations of aging. But whats not clear is how much each type of damage contributes. Our results suggest that somatic mutations probably contribute to aging to some extent, but exactly how much and in what way remains to be demonstrated, adds Martincorena, who concludes with an acknowledgment of the limitations of science: If there is still much understand in cancer, in aging our knowledge is still much more primitive.

Edited by S.U.

View original post here:
A human accumulates as many mutations in 80 years as a mouse in its short life - EL PAS in English

Century Therapeutics Reports Fourth Quarter and Year-end 2021 Financial Results and Provides … – The Bakersfield Californian

IND submission for lead program CNTY-101 on track for mid 2022; Phase 1 ELiPSE-1 trial of CNTY-101 in relapsed/refractory lymphoma expected to commence after IND submission

Entered into a strategic collaboration with Bristol Myers Squibbto develop iPSC-derived allogeneic cell therapies

Ended 2021 with cash, cash equivalents, and marketable securities of $358.8M; Cash runway into 2025, including proceeds received from Bristol Myers Squibb in connection with the Collaboration Agreement

PHILADELPHIA, March 17, 2022 (GLOBE NEWSWIRE) -- Century Therapeutics, Inc., (NASDAQ: IPSC), an innovative biotechnology company developing induced pluripotent stem cell (iPSC)-derived cell therapies in immuno-oncology, today reported financial results and business highlights for the fourth quarter and year ended December 31, 2021.

Throughout 2021, we continued to make steady progress in developing our comprehensive, next-generation iPSC-based cell therapy platform, executed on our powerful discovery engine, and we believe we are positioned to transition to a clinical stage company in 2022. With this foundation in place, we are on track to advance multiple product candidates to the clinic over the next three years, said Lalo Flores, Chief Executive Officer, Century Therapeutics. Additionally, we look forward to continuing our partnership in the years ahead with Bristol Myers Squibb, a global leader in oncology and hematology, to further expand our pipeline of iPSC-derived cell therapy products for treating hematological and solid tumor malignancies. We are committed to maximizing the potential utility of our platform technology and look forward to what we expect to be a very productive year ahead.

Business Highlights

Entered into a collaboration and license agreement with Bristol Myers Squibb in January 2022 to develop and commercialize up to four iPSC-derived, engineered natural killer cell (iNK) and / or T cell (iT) programs for hematologic malignancies and solid tumors. Under the terms of the agreement, Century received a $100 million upfront payment and Bristol Myers Squibb made a $50 million equity investment in Century Therapeutics common stock. The agreement provides for future program initiation fees and development, regulatory, and commercial milestone payments totaling more than $3 billion plus royalties on product sales.Announced that, subject to U.S. Food and Drug Administration (FDA) acceptance of its Investigational New Drug (IND) application, the Company plans to initiate a Phase 1 trial, ELiPSE-1, to assess CNTY-101 in patients with relapsed/refractory aggressive lymphoma or indolent lymphoma after at least two prior lines of therapy, including patients who have received prior CAR T cell therapy. In vivo data

demonstrated strong antitumor activity against human lymphoma cell lines with CNTY-101.Announced plans to focus its initial T cell development program on cells. Data

suggest that CAR-iT cells provide an opportunity to deliver allogeneic T cell therapies without risk for graft-versus-host disease. CNTY-102 will be a CAR- iT candidate targeting CD19, and a second antigen for relapsed/refractory B cell lymphoma and other B cell malignancies. Added to the NASDAQ Biotechnology Index (NASDAQ: NBI) in December 2021.

Upcoming Milestones

Current Good Manufacturing Practice (cGMP) manufacturing facility expected to be operational in 2022.CNTY-101 IND filing remains on track for mid-2022. Subject to U.S. FDA acceptance of its IND application, the Company plans to initiate the Phase 1 ELiPSE-1 trial of CNTY-101 in relapsed/refractory lymphoma in 2022.Expect to submit an IND for CNTY-103 in 2023. CNTY-103 is Centurys first solid tumor candidate for glioblastoma.

Fourth Quarter and Year-end 2021 Financial Results

Cash Position:Cash, cash equivalents, and marketable securities were $358.8 million as of December 31, 2021, as compared to $76.8 million as of December 31, 2020. Net cash used in operations was $89.0 million for the twelve months ended December 31, 2021, compared to $41.3 million for the twelve months ended December 31, 2020.Research and Development (R&D) expenses: R&D expenses were $75.6 million for the year ended December 31, 2021, compared to $39.7 million for the year ended December 31, 2020. The increase in R&D expenses was primarily due to an increase in personnel expenses related to increased headcount to expand the Companys R&D capabilities, costs for preclinical studies, costs for laboratory supplies, and facility costs.General and Administrative (G&A) expenses: G&A expenses were $19.2 million for the year ended December 31, 2021, compared to $9.5 million for the year ended December 31, 2020. The increase was primarily due to an increase in personnel related expense due to an increase in employee headcount and an increase in the Companys professional fees as a result of expanded operations to support its infrastructure as well as additional costs to operate as a public company.Net loss: Net loss was $95.8 million for the year ended December 31, 2021, compared to $53.6 million for the year ended December 31, 2020.

Financial Guidance

The Company expects full year GAAP Operating Expenses to be between $155 million and $165 million including non-cash stock-based compensation expense of $10 million to $15 million. The Company expects its cash, cash equivalents, and marketable securities, including proceeds from the Bristol Myers Squibb collaboration agreement, will support operations into 2025.

About Century Therapeutics

Century Therapeutics, Inc. (NASDAQ: IPSC) is harnessing the power of adult stem cells to develop curative cell therapy products for cancer that we believe will allow us to overcome the limitations of first-generation cell therapies. Our genetically engineered, iPSC-derived iNK and iT cell product candidates are designed to specifically target hematologic and solid tumor cancers. We are leveraging our expertise in cellular reprogramming, genetic engineering, and manufacturing to develop therapies with the potential to overcome many of the challenges inherent to cell therapy and provide a significant advantage over existing cell therapy technologies.We believe our commitment to developing off-the-shelf cell therapies will expand patient access and provide an unparalleled opportunity to advance the course of cancer care. For more information on Century Therapeutics please visit https://www.centurytx.com/.

Forward-Looking Statements

This press release contains forward-looking statements within the meaning of, and made pursuant to the safe harbor provisions of, The Private Securities Litigation Reform Act of 1995. All statements contained in this press release, other than statements of historical facts or statements that relate to present facts or current conditions, including but not limited to, statements regarding our cash and financial resources, our clinical development plans, the development of our U.S. manufacturing facility, and our financial guidance are forward-looking statements. These statements involve known and unknown risks, uncertainties and other important factors that may cause our actual results, performance, or achievements to be materially different from any future results, performance or achievements expressed or implied by the forward-looking statements. In some cases, you can identify forward-looking statements by terms such as may, might, will, should, expect, plan, aim, seek, anticipate, could, intend, target, project, contemplate, believe, estimate, predict, forecast, potential or continue or the negative of these terms or other similar expressions. The forward-looking statements in this presentation are only predictions. We have based these forward-looking statements largely on our current expectations and projections about future events and financial trends that we believe may affect our business, financial condition, and results of operations. These forward-looking statements speak only as of the date of this press release and are subject to a number of risks, uncertainties and assumptions, some of which cannot be predicted or quantified and some of which are beyond our control, including, among others: our ability to successfully advance our current and future product candidates through development activities, preclinical studies, and clinical trials; our reliance on the maintenance of certain key collaborative relationships for the manufacturing and development of our product candidates; the timing, scope and likelihood of regulatory filings and approvals, including final regulatory approval of our product candidates; the impact of the COVID-19 pandemic on our business and operations; the performance of third parties in connection with the development of our product candidates, including third parties conducting our future clinical trials as well as third-party suppliers and manufacturers; our ability to successfully commercialize our product candidates and develop sales and marketing capabilities, if our product candidates are approved; and our ability to maintain and successfully enforce adequate intellectual property protection. These and other risks and uncertainties are described more fully in the Risk Factors section of our most recent filings with the Securities and Exchange Commission and available at http://www.sec.gov. You should not rely on these forward-looking statements as predictions of future events. The events and circumstances reflected in our forward-looking statements may not be achieved or occur, and actual results could differ materially from those projected in the forward-looking statements. Moreover, we operate in a dynamic industry and economy. New risk factors and uncertainties may emerge from time to time, and it is not possible for management to predict all risk factors and uncertainties that we may face. Except as required by applicable law, we do not plan to publicly update or revise any forward-looking statements contained herein, whether as a result of any new information, future events, changed circumstances or otherwise.

For More Information:

Company: Elizabeth Krutoholow investor.relations@centurytx.com

Investors: Melissa Forst/Maghan Meyers century@argotpartners.com

Media: Joshua R. Mansbach century@argotpartners.com

See the original post:
Century Therapeutics Reports Fourth Quarter and Year-end 2021 Financial Results and Provides ... - The Bakersfield Californian

The Incredible Story of Emily Whitehead & CAR T-Cell Therapy : Oncology Times – LWW Journals

Emily Whitehead:

Emily Whitehead

Warriors come in all shapes and sizes. Take for example Emily Whitehead, as fresh-faced a 16-year-old as has ever graced the planet. Her eyes nearly sparkle with intellectual curiosity and dreams for a fulfilling future. But Emily is not a typical teen. She is the first pediatric patient in the world to receive CAR T-cell therapy for relapsed/refractory acute lymphoblastic leukemia (ALL). She is a singular figure in the annals of medicine. She is a soldier on the front lines of the war on cancer. And like the shot heard round the world, her personal medical assault sparked a revolution in cancer care that continues to power forward.

It has been 10 years since the only child of Thomas and Kari Whitehead of Philipsburg, PA, received an infusion of CAR T cells at the hands of a collaborative medical team from the Children's Hospital of Philadelphia (CHOP) and the Hospital of the University of Pennsylvania. That team included, among others, luminary CAR T-cell therapy pioneer, Carl June, MD, the Richard W. Vague Professor in Immunotherapy in the Department of Pathology and Laboratory Medicine and Director of the Center for Cellular Immunotherapies at Penn's Perelman School of Medicine; as well as Stephan Grupp, MD, PhD, Professor of Pediatrics at the Perelman School of Medicine (at that time, Director of the Cancer Immunotherapy Program at CHOP) and now Section Chief for Cell Therapy and Transplant at the hospital. He had been working with June on cell therapies since 2000.

Tremendous progress has flowedgushedfrom the effort to save Emily Whitehead; many more lives have been saved around the globe since that fatefulyet nearly fatalundertaking. While all the progress that has come from this story must be our ultimate theme, it cannot be fully appreciated without knowing how it came to be.

In 2010, Emily, then 5 years old, went from a being a healthy youngster one day, to a child diagnosed with ALL. Chemotherapy typically works well in pediatric ALL patients; Emily was one of the exceptions. After 2 years of intermittent chemotherapy, she continued to relapse. And when a bone marrow transplant seemed the only hope left, her disease was out of control and the treatment just wasn't possible. The Whiteheads were told by her medical team in Hershey, PA, nothing more could be done. They were instructed to take Emily home where she could die peacefully, surrounded by family.

But peaceful surrender didn't interest the Whiteheads; they rejected any version of giving up. It ran contrary to Tom Whitehead's vision of her recovery, something he said was revealed to him in the whispers. He saw, in a prophetic whispering dream, that Emily would be treated in Philadelphia. More importantly, he saw she would survive. It is as if it happened yesterday, said Tom, remembering how unrelentingly he called doctors at CHOP and said, We're coming there, no matter what you can or cannot do. We're not letting it end like this.

Since we treated Emily, we have treated more than 420 patients with CAR T cells at CHOP. She launched a whole group to be treated with this therapy; thousands have been treated around the world.Stephan Grupp, MD, PhD

A combination of persistence and perfect timing provided the magic bullet. It was just the day before that CHOP received approval to treat their first pediatric relapsed/refractory ALL patient with CAR T cells in a trial. And standing right there, on the threshold of history, was that deathly sick little girl named Emily.

At that time, only a scant few terminal adult patients had ever received the treatment, which is now FDA-approved as tisagenlecleucel and developed in cooperation with CHOP and the University of Pennsylvania. When three adults were treated, two experienced quick and complete remission of their cancers. Could CAR T-cell therapy perform a miracle for Emily? A lot would ride on the answer.

On March 1, 2012, Emily was transferred to CHOP and a few days later an apheresis catheter was placed in her neck; her T cells were extracted and sent to a lab. Emily received more chemotherapy, which knocked out her existing immune system, and she was kept in isolation for 6 weeks. Waiting.

Finally, over 3 days in April, Emily's re-engineered T cells, weaponized with chimeric antigen receptors, were infused back into her weakening body. But Emily did not rise like a Phoenix from the ashes of ALL. Instead, she sunk into the feverish fire of cytokine release syndrome (CRS), and experienced a worse-than-anticipated reaction. The hope for a swift victory seemed to be disappearing.

I can still see Emily's blood pressure dropping down to 53/29, her fever going up to 105F, her body swelling beyond recognition, her struggle to breathe, said Tom, of the most nightmarish period of his life. Doctors induced a coma, and Emily was put on a ventilator. For 14 days, her death seemed imminent. Doctors told us Emily had a one in a thousand chance of surviving, said Tom. They said she could die at any moment. But she didn't.

Medical team members who fought alongside the young patient are unwavering heroes in Emily's story. But at the time of her massive struggle, they too were exhausted and battle-scarred, descending into the quicksand of what could have been a failing trial, grasping for some life-saving branch of stability. They knew if CRS could be overcome, the CAR T cells might work a miracle as they had done for those earlier adult patients. But the CRS was severe. There was no obvious antidote; time was running out.

I recall Dr. June saying he believed Emily was past the point where she could come back and recover, said her father. And he said if she didn't turn around, this whole immunotherapy revolution would be over.

The Whiteheads enjoy Penn State football games not far from their hometown. The family has often taken part in Penn State's THON, a 48-hour dance marathon that raises funds for childhood cancer.

June confirmed to Oncology Times that he and Grupp believed Emily would not survive the night. It was mentioned to the Whiteheads that perhaps they should just concentrate on comfort care measures and stop all the ICU interventions, he recalled. I believed she was going to die on the trial due to all the toxicity. I even drafted a letter to our provost to give a heads up.

When the first patient in a trial dies, that's called a Grade 5 toxicity, June noted. That closes the trial as well. It goes right into the trash bin and you have to start all over again. But fortunately, that letter never left my outbox. We decided to continue one more day, and an amazing event happened.

Grupp, offering context to the mysterious amazing event, said it was clear that Emily's extreme CRS was caused by the infusion of cells that he himself had placed in her fragile body. He said he felt an enormous sense of responsibility and incredible urgency as he watched the child struggle to live.

It was not until the CHOP/Penn team received results from a test profiling cytokines in Emily's body that a new flicker of hope sparked. Though Emily had many cytokine abnormalities, the one most strikingly abnormal, interleukin-6 (IL-6), caught the team's attention. It is not made by T cells, and should not have been part of the critical mix. Though there were very few cytokines that had drugs to target them individually, IL-6 was one that did. So the doctors decided to repurpose tocilizumab, an arthritis drug, as a last-ditch effort at saving their young patient.

We treated Emily with tocilizumab out of desperation, June admitted. Steve [Grupp] has told me that when he went to the ICU with tocilizumab as a rescue attempt for CRS, the ICU docs called him a cowboy. The ICU docs had given up hope for Emily. But she turned aroundunbelievably rapidly. Today, tocilizumab is the standard of care for CRS, and the only drug approved by the FDA for that complication. Emily's recovery was huge for the entire field.

Grupp reflected on the immensity of the moment. If things had gone differently, if Emily had experienced fatal toxicity, it would have been devastating to her family and to the medical team. And it might have ended the whole research endeavor. It would have set us back years and years. The impact that Emily and her family had on the field is nothing short of transformational, he declared.

Since we treated Emily, we have treated more than 420 patients with CAR T cells at CHOP. She launched a whole group to be treated with this therapy; thousands have been treated around the world, Grupp noted. And, if not for Emily, we wouldn't be in the position we are in todaywith five FDA-approved [CAR T-cell] products: four for adults and one for kids. And I think it also important to point out that the very first CAR-T approval, thanks to Emily, was in pediatric ALL.

June noted that between 2010 and the time of Emily's treatment in 2012, My work was running like a shoestring operation. I had to fire people because I couldn't get grants to support the infrastructure of the research. It was thought there was no way beyond an academic enterprise to actually make customized T cells, then mail and deliver them worldwide, he recalled.

But then everything changed. We experienced that initial success; it was totally exciting. It was a career-defining moment and the culmination of decades of research. It led to a lot of recognition, both for my contribution and for the team here at the University of Pennsylvania and at CHOP.

Today, hundreds of pharmaceutical and biotech companies are developing innovations. Hundreds of labs are making next-generation approaches to improve in this area, June noted. Today, I'm a kid in a candy shop because all kinds of things are happening. We have funding thanks to the amazing momentum from Emily. She literally changed the landscape of modern cancer therapy.

Grupp said the continuing CAR T-cell program at CHOP offers evidence of success in a broad perspective. There are two things to look at, he offered. The first is how well patients do with their therapy in terms of getting into remission. A month after getting their cells, are they in remission or not? A study with just CHOP patients showed that more than 90 percent met that bar (N Engl J Med 2014; doi: 10.1056/NEJMoa1407222). Worldwide, the numbers appear to be in the 80 percent range (N Engl J Med 2018; doi: 10.1056/NEJMoa1709866). So, I would say it is a highly successful therapy.

We now have trials using different cell types, like natural killer cells, monocytes, and stem cells, noted Carl June, MD, at Penn's Perelman School of Medicine. An entirely new field has opened because of our initial success. This is going to continue for a long time, making more potent cells that cover all kinds of cancer.

The other big question, Grupp noted: How long does remission last? We are probably looking at about 50 percent of patients remaining in remission long-term, which is to say years after the infusion. The farther out we go, the fewer patients there are to look at because it just started with Emily in 2012, reminded Grupp. We have Emily now 10 years out, and other patients who are at 5, 6, 7, 8 years out, but most were treated more recently than that. We need to follow them longer.

June said registries of patients treated with CAR T-cell therapy are being kept worldwide by various groups, including the FDA. CAR T-cell therapy happened fastest in the U.S., but it's gained traction in Japan, Europe, Australia, and they all have databases. The U.S. database for CAR T cells will probably be the best that exists, because the FDA requires people treated continue follow-up for at least 15 years, he explained.

This will provide important information about any long-term complications, and the relapse rate. If patients do get cancer again, will it be a new one or related to the first one we treated? We will follow the outcomes, he noted. Clinicians are teaching us a lot about how to use the informationat what stage of the disease the therapy is best used, and which patients are most likely to respond. This can move us forward.

June mentioned that Grupp is collaborating with the Children's Oncology Group ALL Committee led by Mignon Loh, MD, at the University of California in San Francisco.

They are conducting a national trial to explore using CAR T cells as a frontline therapy in newly diagnosed patients, he detailed. Emily was treated when she had pounds and pounds of leukemia in her body; ideally we don't want to wait so long. There are a lot of reasons to believe it would work as a frontline therapy and spare patients all the complications of previous chemotherapy and/or radiation. The good news is that the clinical trial is under way, and I suspect we may know the answer within 2 years.

The only true measure of success in Emily's case is the state of her health. When asked if she is considered cured, June said, All we can do is a lot of prognostication. We know with other therapies in leukemia, the most similar being bone marrow transplants, if you go 5 years without relapsing, basically you are considered cured. We don't know with CAR T cells because Emily is the first one. We have no other history. But she's at a decade now, and in lab data we cannot find any leukemia in her. So by all of the evidence we haveand by looking in the magic eight ballI believe Emily is cured.

One might think that going through such a battle for life would be enough for any one person, any one family. But for Emily and her parents, her survival was just the beginning of a larger assault. All of them saw the experience as a way to provide interest in continuing research, education for patients as well as physicians, and an extension of hope to other patients about to succumb to a cancerous enemy.

Tom thought back to one particular occasion, all those years ago, when Emily finally slept peacefully through the night in her hospital bed. I should have felt nothing but relief, but I heard a mother crying in the hallway. Her child, who has been in the room next door, had died that morning, he recalled. I am constantly reminded of how fortunate we are. There are so many parents fighting for their children who do not have a good outcome.

As soon as Emily regained her strength and resumed normal childhood activities, the family began travelling with members of the medical team, joining in presentations at meetings and conferences throughout the world. They wanted to give a human face to the potential of CAR T-cell therapy, and as such they willingly became a powerful tool to raise understanding and essential research dollars. In 2016, the Whiteheads founded the Emily Whitehead Foundation (www.emilywhiteheadfoundation.org) ...to help fund research for new, less toxic pediatric treatments, and to give other families hope.

We decided to hold what we called the Believe Ball in 2017. We asked lots of companies to sponsor a child who had received CAR T-cell treatment to come with their family to the ball at no cost to them. Each company's representative would be seated with the child and family they sponsored, and would meet the doctors and scientists involved in the research, as well as members of industry and pharma, to see exactly where research dollars are going. We implored these companies to move the cancer revolution forward with sponsorship. When it all shook out, we had around 35 CAR T-cell families together for the first time, said Tom.

He noted proudly that since the foundation's debut, donations have been consistent and now have totaled an impressive $1.5 million.

When the Emily Whitehead Foundation had a virtual gala recently, it awarded a $50,000 grantthe Nicole Gularte Fight for Cures Ambassador Awardto a young researcher working to get another trial started. The award is named for a woman who found her way to CAR T-cell trials at Penn through the Whitehead Foundation. The treatment extended her life by 5 years during which time Gularte became an advocate for other cancer patients, travelled with the Whiteheads, and made personal appearances whenever she thought she could be of help or inspiration. Eventually, she would relapse and succumb, but she assured Tom Whitehead, These were 5 of the best years of my life. I think my time here on Earth was meant to help cancer research move forward.'

While raising funds for progress is important, the Whiteheads' work is not just about bringing in money. It's also about education.

We want to send a message to all oncologists; they need to be more informed about these emerging treatments when their patients ask for help, Tom noted. In the beginning of CAR T-cell therapy, a lot of doctors were against it. It's hard to believe, but some still are, though not as much. We need more education so that oncologists give patients a chance to get to big research hospitals for cutting-edge treatments before everything else has failed.

June said he regularly interacts with patients Tom or the foundation refer to him. Such unawareness happens with all new therapies, he noted. The people most familiar with them are at academic medical centers. But only about 10 percent of patients actually go to academic centers, the rest are in community centers where newer therapies take much longer to roll out, he explained.

So much of Emily's life has been chronicled through the eyes of observers. But since her watershed medical intervention, she has grown into a well-travelled, articulate young woman who talks easily about her life. I used to let my father do all the talking, but I am finding my own voice now, she said, having granted an interview to Oncology Times.

I'm currently 16 years old and I'm a junior at high school. Just like when I was younger, cows are my favorite animals, she offered with a laugh. I still love playing with our chihuahua, Luna. In school, I love my young adult literature class because I really like reading. Besides that, I like art and film. And I'm in really good health today.

She mentioned her health casually, almost as an afterthought. I really don't have any memory of my treatment at this point, she revealed, but, the experiences that I've had since then have really shaped who I am. Traveling is a huge part of my life now and something I look forward to. We've been to conferences at a lot of distant places. I'm so grateful that I get to travel with my family and make these memories that I will have forever, while still being able to advocate for less toxic treatment options and raising money for cancer research. All of that is really important to me.

Reminded that she has already obtained fame as pediatric patient No. 1 for CAR T-cell therapy, Emily considered her status for a moment then commented, I don't really like to base the progress of the therapy on my story and what I went through. Instead, I like to take my experience and use it to advocate for all patients so that what happened to me does not have to be repeated and endured by another family. My hope is that CAR T-cell therapy will become a frontline treatment option and be readily available, so pediatric patients can get back to a normal life as soon as possible. I want to tell people if conventional treatments do not work, other options do exist. Overall, I am grateful that I can encourage others to keep fighting. That's the main thing; I am grateful.

After a brief pause, Emily continued, I always tell oncologists and scientists that the work they are doing is truly saving children's lives. It allows these kids to grow up, be with their friends and families, take vacations, play with their dogs, and someday go to college, just like me. They are not only saving patients' lives, they are saving families. The work they do does not go unnoticed or unappreciated. Again, I am really so grateful.

Appreciation is a two-way street, and June said he and his team appreciate and draw inspiration from Emily on a daily basis. Her picture hangs on the wall of our manufacturing center, June stated. Some of the technicians who were in high school when Emily was infused are now manufacturing CAR T cells. They learned so much from Emily's experience; she continues to be a big motivator. She's helped my team galvanize and see that the work can really benefit people.

Grupp said the success that is embodied in Emily Whitehead has spurred additional successes, and new inroads in CAR T-cell therapy. There are more applications now, especially in other blood cancerslymphoma and myeloma, in addition to leukemia. We've seen a lot of expansion there.

He noted a national trial is under way for an FDA-approved therapy called idecabtagene vicleucel, which can benefit multiple myeloma patients. All other CAR Ts target the same target, CD19. But this goes after an entirely different target, BCMA. The fact that we now have approval in something that isn't aimed at CD19 is very exciting. And there are others coming right behind it.

The field also has seen further expansion ...into adults being treated safely, because initially there was concern that these drug therapies were too powerful for safe treatment in older adults, detailed Grupp. Now we know that is clearly not the case, and that is great news, particularly because multiple myeloma most often occurs in people over 60.

The use of CAR T cells in solid tumors continues to be challenging, although Grupp noted, We have certainly seen hints of patients with solid tumors having major responses and going into remission with CAR T cells. It is still a small handful of patients, so we haven't perfected the recipe for solid tumors yet. But I am absolutely confident we will have the answers in a very short numberperhaps 2-4of years.

June said, since Emily's infusion, CAR T cells have matured and gotten better. There are many ways that has happened, he informed. We have different kinds of CAR designs to improve and increase the response rates, to decrease the CRS, or to target other kinds of bone marrow cancers. One that is not curable with a lot of therapies is acute myeloid leukemia (AML), so we have a huge group at Penn and CHOP working on AML specifically. And there is the whole field of solid cancer; we have teams working on pancreatic, prostate, breast, brain, and lung cancer now.

In addition to targeting different types of cancer, June said contemporary research is also exploring the use of different types of cells. Our initial CAR T trial used T cells, and that is what all the FDA-approved CARs are. But we now have trials using different cell types, like natural killer cells, monocytes, and stem cells. An entirely new field has opened because of our initial success. This is going to continue for a long time, making more potent cells that cover all kinds of cancer, not just leukemia and lymphoma.

Is this the beginning of the end of cancer? Is this that Holy Grail called a cure to cancer? It's a question June has pondered.

Some people do think that, he answered. They believe the immune system is the solution. And that's a huge statement. President Biden has made a big investment in this work, with the Cancer Moonshot. He's accelerated this research at the federal level. But we just don't know how long it is going to take. Fortunately, a lot of good minds are working hard to make an end to cancer a reality.

As the battle grinds on, June said he applies something he's learned over time, with reinforcement from Tom and Kari Whitehead. They were bulldogs. When it came to getting treatment for Emily, they just wouldn't take no for an answer. They demonstrated the importance of never giving up. That's what happened; they would not surrender. I think that is why Emily is alive today.

Valerie Neff Newitt is a contributing writer.

The Emily Whitehead Foundation and the Whitehead family take extraordinary advantage of a variety of media to reach patients and physicians and optimize educational opportunities.

See the original post here:
The Incredible Story of Emily Whitehead & CAR T-Cell Therapy : Oncology Times - LWW Journals

Ray Resection as a Personalized Surgical Technique for Progressive Hand Macrodactyly in a 60-Year-Old Patient: A Case Report and Literature Review -…

Hand macrodactyly is a very scarce deformity. It was first described over 200 years ago and was characterized as local gigantism of one or multiple digits. Benign bone overgrowth, massive increase of soft tissue volume, and nerve involvement are associated with hand macrodactyly have been consistently reported in the literature. Often, macrodactyly affects one or more digits and is further classified as static or progressive, depending on the growth pattern, and as sporadic or syndromic, according to its genetic predisposition. Surgical treatment for hand macrodactyly remains a complex issue even for expert hand surgeons. In most of the cases, macrodactyly is diagnosed during early childhood and can be appropriately managed with minimal and well affordable surgical approaches that stabilize its fast progression. However, adults with progressive hand macrodactyly develop advanced deformities leading to severe functional deterioration and aesthetic hand dysmorphia. The purpose of this report is to document the management and surgical approach of the oldest published case, a 60-year-old adult patient with neglected progressive hand macrodactyly despite previous surgical attempts for disease stabilization. A personalized preoperative planning was created, which included ray resection involving the fourth metacarpal and fourth finger along with extensive debulking of the overgrown fatty soft tissue and carpal tunnel release. At six months follow-up, the patient reported an excellent aesthetic and functional outcome.

The term macrodactyly is a descriptive term derived from the Greek words macro meaning long and dactyl meaning finger. Macrodactyly of the hand represents a very rare congenital deformity of unknown etiology, which constitutes less than 1% of congenital disorders in the upper extremity [1]. To our knowledge, hand macrodactyly affects approximately 1 out of 100,000 live births and can appear either as a sporadic (isolated form) or as part of a hereditary deformity syndrome (syndromic form) [2]. There are two distinct types of macrodactyly depending on the functional status of the hand. Static macrodactyly is the first type of macrodactyly, with affected fingers being roughly one and a half times the size compared to a normal finger [3]. Besides, static macrodactyly is present at birth and abnormal fingers grow in line with normal fingers [3]. Progressive macrodactyly constitutes the second type, causing continuous bony overgrowth even after skeletal maturity, with digits growing at a much faster rate compared to normal ones [3]. Oftentimes, the dysmorphic appearance of hand macrodactyly causes functional disability in the majority of cases along with cultural stigma, which might have a negative psychological impact on the patient [4].

A plethora of surgical interventions have been described mainly in young patients to cure static and prevent progressive hand macrodactyly, such as debulking procedures, epiphysiodesis, and osteotomies [1]. The purpose of this report is to document the personalized strategy for surgical reconstruction of a neglected progressive hand macrodactyly in an elder 60-year-old male patient - the oldest individual that we are aware of to have been surgically treated based on our literature search - and our efforts to obtain a functional hand with a good aesthetic outcome.

A 60-year-old male individual was presented to our department to seek consultation for his hand macrodactyly. The patients condition was diagnosed in early childhood as a sporadic isolated anomaly affecting moderately the middle and severely the ring fingers. According to his medical records, he underwent two minimal soft tissue debulking surgeries on the third interdigital space when he was 4 and 37 years old, respectively. Initially, he reported that his hypertrophic left middle and ring fingers became more painful and less functional recently, albeit he could manage it until then. The thumb, index, and little fingers were normal. Despite his hand deformity, the patient has been a professional guitar player for at least 40 years. During the last three years, he was complaining of progressive disproportionate growth of his middle and ring fingers (Figure 1). He was further experiencing numbness, tingling, and ache at the tip of all his left digits accompanied with a painful sensation of fullness on the affected fingers. Phalens test was positive, and electrophysiological tests were indicative of carpal tunnel syndrome due to median nerve compression. However, the ulnar nerve was not found entrapped through Guyon's canal. Likewise, the movements of middle and ring fingers were extremely restricted due to soft tissue hypertrophy and stiffness of his metacarpophalangeal and interphalangeal joints. The flexion of his two gigantic fingers was severely deteriorated due to malalignment of joints and angled phalanges. X-rays revealed excessive hand osteoarthritis with the presence of large bone spurs (osteophytes) and joint space narrowing between all phalanges of the third and fourth fingers. Therefore, a debulking reconstruction surgery of the overgrown fatty tissue and ray resection of the most enlarged fourth finger were performed along with carpal tunnel release.

Under axillary block anesthesia and application of a pneumatic tourniquet, a racket-shaped incision was made around the base of the fourth metacarpal. The extensor and flexor tendons (flexor digitorum superficialis and flexor digitorum profundus) as well as interosseous and lumbrical muscles of the fourth finger were detached and transected carefully to prevent tendon injuries of the remaining normal digits. Exposure of the digital neurovascular bundle of the third and fifth fingers was well visualized, mobilized, and preserved. Radial and ulnar digital nerves of the fourth finger were found enlarged and were easily recognized due to their increased thickness. Subsequently, nerve endings were carefully implanted within the surrounding soft tissue to avoid as much as possible the formation of painful neuromas. With a surgical oscillating saw, a transection at the base of the fourth metacarpal was performed. Subsequently, surgical debulking of the extensive soft tissue was implemented (Figure 2), and hemostasis of blood vessels leakage was achieved with an electrocautery. Eventually, median nerve decompression was achieved with a palmar incision to divide the transverse carpal ligament. The surgical procedure was performed by a senior consultant hand surgeon and a hand fellow orthopaedic surgeon. The postoperative plan consisted of temporary splinting and early supervised physiotherapy.

Postoperatively, the patient was complaining of phantom pain, which was well tolerated with a two-week prescription of acetaminophen and NSAIDs. Eventually, the phantom pain was resolved entirely eight weeks after surgery [5]. At the six-month follow-up, the patient reported a great recovery with excellent functional and aesthetic satisfaction (Figure 2). His grip strength and hand mobility improved with at least 30 of better flexion range for third metacarpophalangeal joint and so did abduction-adduction for his index, ring, and small fingers.

Until recently, pathogenesis of osseous and fibrofatty overgrowth in hand macrodactyly is not clearly identified and still no consensus exists on its treatment. Therefore, hand macrodactyly poses a significant surgical challenge. which often requires the expertise of experienced hand surgeons to manage effectively [6].

It has been reported that significant nerve enlargement is usually observed during surgical approach of affected digits with morphological and neurophysiological impairment of the median nerve that requires carpal tunnel release [7]. In the present case, the third and fourth rays were affected with simultaneous enlargement of the radial and ulnar digital nerve of the fourth finger. However, electrophysiological testing depicted entrapment of the median nerve but not of the ulnar nerve. Lipomatosis of peripheral nerves (fibrolipomatous proliferation within the nerve) accompanied with osseous enlargement and hypertrophic changes on osteochondral tissue leads not only to compressive neuropathy but also to disabling ankylosis of innervated joints [8].

Genetic studies have demonstrated that hand macrodactyly can be a clinical manifestation of three major overgrowth syndromes: the Proteus syndrome (mosaic mutations in the AKT1 gene), the PIK3CA-related overgrowth syndrome (mutations in the PIK3CA oncogene), and the PTEN hamartoma tumor syndrome (mutations in somatic PTEN tumor suppressor gene). According to Cui et al., somatic mutations in PIK3CA oncogene were observed within bone marrow stem cells from patients diagnosed with hand macrodactyly [9]. These specific mutations enhance activation of the PI3K/AKT/mTOR pathway and deregulate bone homeostasis, leading to hyperplastic bone formation [9]. Moreover, Cui et al. demonstrated that downregulation of distal-less homeobox 5 gene (DLX5) which induces Runx2-mediated osteogenesis and P13K-mediated bone overgrowth could be inhibited by the administration of BYL719 [9]. Subsequently, the administration of this novel therapeutic agent in early-stage disease could appropriately reverse progressive hand macrodactyly [9].

Progressive macrodactyly is an extremely challenging disease as no surgery is able to cure the underlying condition. Most patients, even if operated early in life, require multiple debulking procedures in accordance with the present case report. Concurring to the literature, only few published reports demonstrate potential strategies and surgical treatment options for progressive hand macrodactyly without a clear consensus on treatment guidelines, as shown in Table 1 [5,10]. Children with static hand macrodactyly can be appropriately treated with minimal surgical interventions such as stripping or resection of the local nerve, debulking, closing-wedge osteotomies, and phalangeal epiphysiodesis [10,11]. A very innovative surgical technique was proposed by Kobraei et al. to prevent fast skeletal overgrowth and avoid digits amputation in progressive hand macrodactyly [12]. According to the authors, a radical dissection of the diseased gross digital nerve in two cases with thumb (radial digital nerve) and ring finger (ulnar digital nerve) overgrowth was performed until healthy nerve stump was found [12]. The gaps were reconstructed with a processed nerve allograft between normal edges [12]. To authors view, an early application of this novel surgical approach could yield functional and aesthetic digits with remarkable sensory outcomes and significant deceleration of the disease [12].

The largest case series study considering clinical characteristics and surgical management of 90 hand macrodactyly cases was conducted by Wu et al. [2]. According to their study, multiple digit involvement is up to 2.6 times more frequent than a single-digit disease, which is in line with our patient who had his middle and ring fingers enlarged. In the case series report by Wu et al., most of the affected digits (79.4%) involved were in the median nerve innervation surface [2]. However, in the present case report, the patient had a ring finger macrodactyly, which corresponds to the ulnar nerve area, with no signs of ulnar nerve compression. In addition, the study by Wu et al. included young patients aged between six months and five years. The vast majority of patients were treated with soft tissue reconstruction or minimal phalangeal osteotomies and only two out of 90 cases had an amputation [2]. Consequently, function-preserving surgeries are performed instead of amputation when hand macrodactyly is effectively treated during early-stage compared to advanced-stage disease.

Based on recent bibliography, Jacobs et al. presented the most advanced case of a 55-year-old female patient diagnosed with Proteus syndrome and macrodactyly of her right-hand thumb, middle, and index fingers [5]. The individualized surgical plan included amputation of the thumb and index rami and removal of trapezoid, trapezium, and scaphoid bones [5]. Consequently, the resulted wrist instability was treated with transosseous ligament reconstruction [5]. Good aesthetic and functional results were comparable to that in our patient who was treated with a lesser ray resection technique. To the best of our knowledge, the present case report depicts the surgical management of the oldest patient (60-year-old male) with progressive isolated macrodactyly among the published cases in recent literature. In addition, we strongly believe that efficient stabilization during early-stage disease would have prevented the development of severe chronic osteoarthritis in the metacarpophalangeal, proximal interphalangeal, and distal interphalangeal joints of the third and fourth fingers, which add more disability to a macrodactyly hand.

Hand macrodactyly is usually visible at birth and patients experience overgrowth symptoms during early childhood. Consequently, an early and effective surgical management is strongly recommended to prevent chronic progression and development of severe secondary degenerative bone changes in macrodactyly fingers, such as ankyloses, narrowing of joints, and formation of osteophytes. Patients suffering from hand macrodactyly can significantly benefit from early surgical stabilization of the condition instead of late and more aggressive interventions such as amputation.

In the present case, surgical interventions at an early stage proved ineffective, and the patient developed severe and disabling hand deformities due to the progressive subtype of hand macrodactyly. Most of the macrodactyly cases seem to stabilize at skeletal maturity, and it is unusual to see this degree of progressive bony overgrowth. Nevertheless, a personalized surgical technique including ray resection and debulking reconstruction surgery was proposed for this neglected case with great aesthetic and functional outcomes.

More here:
Ray Resection as a Personalized Surgical Technique for Progressive Hand Macrodactyly in a 60-Year-Old Patient: A Case Report and Literature Review -...

European Wellness Collaborates with Heidelberg University Germany to Conduct Efficacy Studies of Peptides and Cell Therapy Research – WILX-TV

Published: Oct. 27, 2021 at 4:30 AM EDT|Updated: 21 hours ago

FRANKFURT, Germany, Oct. 27, 2021 /PRNewswire/ European Wellness Academy (EWA), the educational arm of European Wellness Biomedical Group (EWG), has signed an agreement to carry out joint scientific research on the efficacy of peptides, cell therapy, exosomes and cell reprogramming for rejuvenation in premature murine aging models.

EWA was represented by its Group Chairman, Prof. Dr. Mike Chan, while Heidelberg University was represented by its Commercial Managing Director, Katrin Erk and its Head of Institute of Anatomy and Cell Biology III, Prof. Dr. Thomas Skutella.

The cutting-edge therapeutics used for the studies include precursor (progenitor) stem cells (PSC), precursor cells (Frozen Organo Crygenics (FOC)), Mito Organelle (MO), Nano Organo Peptides (NOP) and exosomes.

Their studies include in vitro experiments concentrating on the effects of the products on the aging of somatic cells and cellular senescence, which is known to contribute to disease onset and progression. Investigated exosomes include neuronal stem cells (NSCs), mesenchymal stem cells (MSCs), cardiomyocytes, kidney progenitors and hepatocytes.

EWA and Heidelberg University will also conduct in vivo experiments to demonstrate both safety and efficacy of the therapeutics, whereby the proof of effectivity will be recorded in the life span, histopathological and molecular criteria of neurodegeneration including Alzheimer/dementia, and system degeneration disorders including those affecting the immune system, skin, cardio, lung, kidney, liver, stomach/intestine/gut, eye, and muscular dystrophy.

Other criteria included are cartilage/joint/bone regeneration including knees/joints/hips, cervical, thoracic, lumbar, pelvic and musculoskeletal disorder, as well as endocrine disorders like endocrinal dysfunction due to over and underproduction of hormones and other activity pattern under the sleep wake cycle.

The ongoing specially designed studies are coordinated and designed by Prof. Dr. Thomas Skutella of Heidelberg University, a world-renowned research university and one of Germany's Top 3, Prof. Dr.Mike Chan and scientists of EWG.

European Wellness Academy

Located in Germany, Switzerland, Greece and Malaysia, EWA is a UK CPD authorised body with a premium training and development wing that revolves around cutting-edge Bio-Regenerative Medicine modalities for practitioners and researchers. The Academy has extensive years of combined clinical experience and a core academic team comprising of qualified clinicians and scientists with multiple international affiliations and accreditations.

https://ewacademy.eu https://european-wellness.eu/

View original content to download multimedia:

SOURCE European Wellness Biomedical Group

The above press release was provided courtesy of PRNewswire. The views, opinions and statements in the press release are not endorsed by Gray Media Group nor do they necessarily state or reflect those of Gray Media Group, Inc.

See original here:
European Wellness Collaborates with Heidelberg University Germany to Conduct Efficacy Studies of Peptides and Cell Therapy Research - WILX-TV

What are the different kinds of stem cells? | American for …

There are three types of stem cells: adult stem cells, embryonic (or pluripotent) stem cells, and induced pluripotent stem cells (iPSCs).

Adult stem cells, or tissue stem cells, can come from different parts of the adult body. They are specific to a certain kind of tissue in the body: for instance, liver stem cells can regenerate liver tissue, and muscle stem cells can regenerate muscle fibers. But adult stem cells are limited to only becoming more of their specialized tissueliver stem cells cannot make new muscle fibers, nor can muscle stem cells make new liver tissue.

The thousands of different cell types that make up our bodies all came from one single master builder cell, called a pluripotent stem cell.

Pluripotent stem cells can be thought of as blank slates, because of their ability to build any cell type in the bodyskin cells, brain cells, muscle cells, etc. Unlike tissue stem cells, pluripotent stem cells are not limited to only becoming more of a certain tissue. Pluripotent stem cells primarily consist of embryonic stem cells, but the term now also encompasses another type of cells, called induced pluripotent stem cells. More on that later.

Induced pluripotent stem (iPS) cells are pluripotent cells that are derived from adult tissue using new scientific technology. They share characteristics with embryonic stem cells in that they can become any cell type in the body.

Reprogramming stem cells to create iPSCs involves some genetic manipulation, and this may cause some differences that are not present in cells that are already embryonic in nature. It is essential to continue research using all cell types. Because the field of stem cell research is so new, it is critical to explore all avenues of stem cell research, from pluripotent to tissue stem cells.

The process of generating an iPS cell line takes time and resources in a lab. To do so in a sterile and safe way in which the cells can be transplanted back into someone is even more expensive. It is also necessary that these cells undergo tests to ensure that they have not mutated or changed in any detrimental way through the reprogramming process. It is a cool idea that everyone could have their own iPS cell line that could be used to make a personalized therapy product for themselves, but in practice this is very time consuming and expensive to do it on a per-person basis. In embryonic stem cell therapies, the generation of the cells has already been performed in the proper ways, and the expensive tests can be performed on a single stem cell line, rather than a different line for every individual.

It is possible that one day iPSCs may prove to be equivalent to embryonic stem cells (ESCs) and could be used in the same way we use ESCs now. However, because iPSCs are a very new discovery (2006), it is still to be determined iPS cells are are equivalent to embryonic stem cells in all ways. Scientists are working hard on understanding the differences that may exist between embryonic stem cells and iPS cells, and we still have yet to determine which cell type will be the most useful for regenerative medicine.

What Are Stem Cells and Why Are They So Important? Stem cells are the builders

Research using pluripotent stem cells is legal in the United States. Federal courts, including the

Proposition 71 created the California stem cell program, formally titled the California Institute of Regenerative

See more here:
What are the different kinds of stem cells? | American for ...

The translatome of neuronal cell bodies, dendrites, and axons – pnas.org

Significance

Proteins are the key drivers of neuronal synaptic function. The regulation of gene expression is important for the formation and modification of synapses throughout the lifespan. The complexity of dendrites and axons imposes unique challenges for protein supply at remote locations. The discovery of messenger RNAs (mRNAs) and ribosomes near synapses has shown that local protein synthesis represents an important solution to this challenge. Here we used RNA sequencing and ribosome sequencing to determine directly the population of mRNAs that is present and in the process of translation in neuronal cell bodies, dendrites, and axons. Thousands of transcripts were differentially translated between the cell body and synaptic regions with over 800 mRNAs exhibiting more translation in the dendriticaxonal compartment.

To form synaptic connections and store information, neurons continuously remodel their proteomes. The impressive length of dendrites and axons imposes logistical challenges to maintain synaptic proteins at locations remote from the transcription source (the nucleus). The discovery of thousands of messenger RNAs (mRNAs) near synapses suggested that neurons overcome distance and gain autonomy by producing proteins locally. It is not generally known, however, if, how, and when localized mRNAs are translated into protein. To investigate the translational landscape in neuronal subregions, we performed simultaneous RNA sequencing (RNA-seq) and ribosome sequencing (Ribo-seq) from microdissected rodent brain slices to identify and quantify the transcriptome and translatome in cell bodies (somata) as well as dendrites and axons (neuropil). Thousands of transcripts were differentially translated between somatic and synaptic regions, with many scaffold and signaling molecules displaying increased translation levels in the neuropil. Most translational changes between compartments could be accounted for by differences in RNA abundance. Pervasive translational regulation was observed in both somata and neuropil influenced by specific mRNA features (e.g., untranslated region [UTR] length, RNA-binding protein [RBP] motifs, and upstream open reading frames [uORFs]). For over 800 mRNAs, the dominant source of translation was the neuropil. We constructed a searchable and interactive database for exploring mRNA transcripts and their translation levels in the somata and neuropil [MPI Brain Research, The mRNA translation landscape in the synaptic neuropil. https://public.brain.mpg.de/dashapps/localseq/. Accessed 5 October 2021]. Overall, our findings emphasize the substantial contribution of local translation to maintaining synaptic protein levels and indicate that on-site translational control is an important mechanism to control synaptic strength.

At neuronal synapses, more than 2,500 proteins (1, 2) (the synaptic proteome) act as sensors and effectors to control neuronal excitability, synaptic strength, and plasticity. The elaborate morphology and functional compartmentalization of the individual neuron imposes unique logistical challenges to maintain and modify the synaptic proteome at locations remote from the transcription source (i.e., the nucleus). To fulfill the local demand for new protein, neurons localize messenger RNAs (mRNAs) and ribosomes near synapses to produce proteins directly where they are needed (1). Using high-throughput sequencing, several groups have reported the localization of thousands of transcripts to axons and dendrites (the local transcriptome) (37). In many cell types, however, it has been shown that the transcript levels do not always predict protein levels (8), suggesting that mRNA translation is a highly regulated process. Since proteins, rather than mRNAs, drive cellular function, it is imperative to determine directly which transcripts are translated into proteins in dendrites and/or axons in vivo (the local translatome). Importantly, it remains unknown which transcripts exhibit differential levels of translation between somatic and synaptic regions.

A given transcripts translation level is determined by the rate of ribosome recruitment to the start codon during initiation and the velocity of ribosome translocation during polypeptide elongation. For most mRNAs, translation initiation is considered rate limiting (9): Initiation is regulated by elements within the mRNAs untranslated regions (UTRs) that bind RNA-binding proteins (RBPs) or miRNAs (1012). In addition, the elongation rate also plays a regulatory role in determining the amount of protein produced from a transcript (13). Although disrupted translational control has been linked to a number of neurological disorders (14), little is known about the magnitude and mechanisms for transcript-specific translational regulation in neuronal compartments.

In this study, we combined deep sequencing of ribosome-protected fragments (ribosome sequencing [Ribo-seq]) and RNA sequencing (RNA-seq) of microdissected hippocampal rodent brain sections to provide a comprehensive analysis of the mRNA translational landscape both in the somata (enriched in cell bodies) and the neuropil (enriched in neuronal dendrites/axons). Thousands of mRNAs were translated in the somatic and synaptic regions. Many transcripts exhibited differential translation levels between somatic and synaptic regions. Many of these translational changes likely resulted from differences in the RNA levels between the somata and neuropil. Furthermore, we found evidence for pervasive translational regulation of synaptic proteins in both neuronal compartments. We provide a dynamic query-based web interface for exploring mRNA transcripts and their translation in neuronal compartments (15). Together, our results reveal an unprecedented capacity for local protein production in vivo to maintain and modify the pre- and postsynaptic proteome.

To discover the mRNA species localized and translated in cell bodies as well as dendrites and axons we carried out a genome-wide analysis of the transcriptome and translatome of the somata and neuropil from microdissected hippocampal slices (16). Ribosome footprints were obtained from somata and neuropil lysates to assess the number and position of translating ribosomes on a transcript (Ribo-seq) (17). In parallel, transcript levels were quantified by performing RNA-seq from the somata and neuropil (Fig. 1A) (16). The RNA- and Ribo-seq libraries from the somata and neuropil were highly reproducible among the three biological replicates (SI Appendix, Fig. S1 A and B). Furthermore, the Ribo-seq samples exhibited the expected depletion of footprint read densities in the UTRs and introns of transcripts (SI Appendix, Fig. S1 C and D), as well as three-nucleotide phasing (SI Appendix, Fig. S1 E and F) (17).

Many transcripts display differential translation between the somata and neuropil. (A) Experimental workflow. Microdissection of the CA1 region of the rat hippocampus. RNA-seq and Ribo-seq were conducted simultaneously for the somata (enriched in pyramidal neuron cell bodies) and the neuropil (enriched in dendrites and axons) layers. A neuronal filter was applied to enrich for excitatory neuron transcripts in downstream analyses. (B) Volcano plot comparing the translational level of 7,850 transcripts between compartments (neuropil:somata Ribo-seq ratio [log2FC]). FDR < 0.05 using DESeq2 (Experimental Procedures). Colored dots highlight the transcripts significantly more translated in the somata (somata [smt]-translation-up, n = 2,945, orange) or neuropil (neuropil [npl]-translation-up, n = 807, teal). (C) Coverage tracks representing the average neuropil (Top) or somata (Bottom) ribosome footprint coverage for candidate smt-translation-up (Gria2, Neurod6, and Hpca) and npl-translation-up (Shank1, Map2, and Dgkz) transcripts. The y axis indicates the number of normalized reads. (D) Schematic depicting in vivo ribosome run-off following harringtonine incubation of rat hippocampal cultures. (E) Elongation rates for smt-translation-up (orange), npl-translation-up (teal), and other (gray) transcripts inferred from the slope of the linear fit shown in SI Appendix, Fig. S4 are plotted with their SE (n = 3). P = 0.5738, One-way ANOVA. Har, harringtonine; Chx, cycloheximide; ns, not significant.

We detected 13,055 and 12,371 transcripts with one count per million (CPM) in two of three neuropil (SI Appendix, Fig. S2A) or somata (SI Appendix, Fig. S2B) Ribo-seq replicates, respectively. Using the Ribo-seq datasets, we found substantial overlap between our translatome data and a previously published neuropil (SI Appendix, Fig. S2A) and somata (SI Appendix, Fig. S2B) transcriptome (3). The somata and neuropil of the hippocampus contain excitatory neuron cell bodies and their processes, as well as glia and interneurons. We created a pipeline to focus on excitatory neuron genes by minimizing the contribution of other cell types via bioinformatic filtering. To obtain a comprehensive set of glia-enriched transcripts, we prepared hippocampal neuron- and glia-enriched cultures (SI Appendix, Fig. S2C and Dataset S1). Because the somata and neuropil do not only contain glia but also interneurons, we additionally compiled lists of transcripts enriched in nonexcitatory neuron cell types in the hippocampus. To do so, we identified the transcripts significantly deenriched in the hippocampi of two different RiboTag mouse lines that target primarily excitatory neurons: Camk2Cre::RiboTag mice (SI Appendix, Fig. S2D), as well as the microdissected somata (SI Appendix, Fig. S2E) and neuropil (SI Appendix, Fig. S2F) from Wfs1Cre::RiboTag mice (16). Combining these datasets, we obtained a list of contaminant nonexcitatory neuron genes (SI Appendix, Fig. S2G).

The number of ribosomes loaded on a transcript indicates how much it is translated. To identify transcripts that exhibit differential translation between the somata and neuropil, we computed neuropil:somata Ribo-seq ratios (DESeq2) (18) (Experimental Procedures). After subtraction of the contaminant genes, we detected 7,850 neuronal transcripts (SI Appendix, Fig. S2H) (19) that were translated in both the somata and neuropil (Fig. 1B). Of these, 807 transcripts exhibited significantly increased translation levels in the neuropil compared to the somata (neuropil-translation-up) (Fig. 1B and Dataset S2). The neuropil-translation-up transcripts included, for example, Shank1, Map2, and Dgkz (Fig. 1 B and C). In contrast, 2,945 transcripts showed increased translation in the somata, including Gria2, Neurod6, and Hpca (somata-translation-up) (Fig. 1 B and C and Dataset S2). Both neuropil- and somata-translation-up transcripts exhibited three-nucleotide periodicity arising from the codon-by-codon translocation of ribosomes along mRNAs during translation in the neuropil and somata, respectively (SI Appendix, Fig. S3 A and B). Consistent with previous findings (12), the neuropil-translation-up transcripts displayed significantly longer 3 UTRs (SI Appendix, Fig. S3C).

Previous studies suggested that mRNAs present in dendrites and/or axons might be translationally silenced, via the pausing of ribosomes at the level of elongation (13, 20). To address this, we asked whether the neuropil- and somata-translation-up transcripts exhibited differences in the speed of translation elongation. We performed a time series of ribosome run-off by incubating cultured hippocampal neurons for 15, 30, 45, or 90 s with harringtonine, a drug that immobilizes ribosomes immediately after translation initiation, resulting in a progressive run-off of ribosomes over time (Fig. 1D and SI Appendix, Fig. S4). We analyzed the rate of ribosome progression (elongation) from the 5 end of neuropil- and somata-translation-up transcripts (SI Appendix, Fig. S4). The neuropil- and somata-translation-up transcript subsets displayed a similar elongation rate of 4 codons per second (Fig. 1E and SI Appendix, Fig. S4), a value that is within the range measured in other cell types (3 to 10 codons per second) (2124). Together, these findings indicate that neuropil-translation-up mRNAs are globally not significantly more paused than other transcripts.

To examine whether particular protein function groups are encoded by transcripts that exhibit increased translation levels in either compartment, we performed a gene ontology (GO) analysis (Fig. 2 A and B). An enrichment of terms associated with synaptic function was found for both somata- and neuropil-translation-up transcripts (Fig. 2 A and B). For the somata-translation-up transcripts, we observed a significant overrepresentation of the term perikaryon as well as many membrane-related terms such as integral component of postsynaptic density membrane, presynaptic membrane, or synaptic vesicle membrane (Fig. 2A). On the other hand, mostly postsynaptic functions were significantly associated with the neuropil-translation-up transcripts, including for example dendritic spine and postsynaptic density (Fig. 2B). To understand better the synaptic function of the neuropil- and somata-translation-up transcripts, we analyzed the neuropil:somata Ribo-seq fold changes of excitatory synaptic proteins (Fig. 2C). We noted that ionotropic and metabotropic glutamate receptor subunits (AMPARs, NMDARs, and mGluRs) mostly displayed greater translation levels in the somata (Fig. 2C). In contrast, many glutamate receptor-associated accessory (e.g., Cnih2) or scaffold proteins (e.g., Shank1, Dlg4, and Homer2) exhibited increased translation levels in the neuropil (Fig. 2C). Also, we found that many presynaptic proteins exhibited greater protein synthesis rates in the somata (Fig. 2C). Interestingly, we identified several nuclear-encoded mRNAs related to mitochondrial function that exhibited enhanced translation levels in the neuropil (e.g., Timm8a1 and Mrpl40) (Fig. 2C).

Functional segregation of transcripts differentially translated between the somata and neuropil. (A and B) GO terms representing the top five highest significantly enriched (FDR < 0.05) protein function groups for somata-translation-up (A) and neuropil-translation-up (B) transcripts. (C) Scheme depicting proteins of glutamatergic synapses. Ribo-seq neuropil:somata ratios (log2FC) are color coded from orange (more somata-translated) to teal (more neuropil-translated). Interacting proteins are displayed in closer proximity. Proteins with similar functions are grouped together and the synaptic vesicle cycle is indicated by arrows.

The mRNA transcript and translation profiles in the somata and neuropil are available for download and exploration at a searchable web interface (https://public.brain.mpg.de/dashapps/localseq/). This interactive database allows viewers to compare transcript and mRNA translation levels between neuronal compartments.

The translation level of a given transcript is proportional to its abundance and its ribosome density. We thus asked whether differential translation of somata- and neuropil-translation-up transcripts was associated with between-compartment changes in RNA levels (Dataset S3). Indeed, neuropil-translation-up transcripts displayed significantly higher neuropil:somata RNA-seq ratios compared to somata-translation-up genes (Fig. 3A). In order to validate these observations in situ in hippocampal slices, we performed high-resolution fluorescence in situ hybridization (FISH) for 14 candidate transcripts with significantly different translation levels between the somata and neuropil (Fig. 3 BD). The in situ hybridization signal detected was highest in expected compartment (i.e., somata for somata-translation-up, Fig. 3 B and D, and neuropil for neuropil-translation-up, Fig. 3 C and D). Taken together, both the RNA-seq and FISH analyses revealed that increased translation in the somata or neuropil was accompanied by higher RNA levels in the same neuronal compartment.

Differential translation of neuropil- and somata-translation-up genes is accompanied by between-compartment changes in RNA levels. (A) Box plot representing the neuropil:somata RNA-seq ratio (log2FC) for somata (smt)-translation-up (orange) and neuropil (npl)-translation-up (teal) genes (DESeq2; Experimental Procedures). (B and C) (Top) Neuropil:somata RNA-and Ribo-seq ratios (log2FC) for candidate smt-translation-up genes (Gria2, Cacng8, Uchl1, Sv2b, Syp1, Gria1, and Snap25) (B) and npl-translation-up genes (Aco2, Dlg4, Hpcal4, Cnih2, Ddn, Eef2, and Camk2a) (C). (Bottom) FISH signal in the CA1 region of rat hippocampal slices using probes against smt- (B) and npl-translation-up (C) candidate genes. The dendrites were immunostained with an anti-MAP2 antibody (purple). (Scale bar, 50 m.) (D) Neuropil:somata ratio of mRNA puncta relative to the mean neuropil:somata ratio of the smt-translation-up genes (***P < 2.2e-16, MannWhitney U Test between all smt-translation-up and all npl-translation-up genes).

We next compared gene-level translation efficiencies (TEs) between the neuropil and somata by computing the ratio of ribosome footprints (from Ribo-seq) to mRNA fragments (from RNA-seq) (17) in both compartments (Fig. 4A and Dataset S4). We observed a good correlation between the somata and neuropil TE values, indicating that most transcripts exhibit similar translational regulation in both neuronal compartments (Fig. 4A, R2 = 0.92, P < 2.2e-16). For instance, Syngap1 exhibited low footprint-to-mRNA ratios in both somata and neuropil, indicating the relatively poor translational efficiency of this transcript (Fig. 4 A and B). In contrast, Camk2a was found translated with high efficiency (high footprint-to-mRNA ratio) in both neuronal compartments (Fig. 4 A and B). We also identified a handful of mRNAs that displayed significantly higher TE values in the somata, including, for example, Kif5c (Fig. 4 A and B). Thus, many but not all of the between-compartment differences in ribosome footprint levels can be accounted for by differences in the amount of mRNA present.

Most transcripts exhibit similar translational efficiency in the somata and neuropil. (A) Correlation of the translational efficiencies (TE; log2Ribo-Seq/RNA-seq) in the neuropil and somata (R2 = 0.92, P < 2.2e-16). Highlighted are genes with significantly higher (TEhigh, yellow) or lower (TElow, blue) TE than log2 1.5 (FDR < 0.05, DESeq2) in both somata and neuropil. Genes with significantly differential TE between somata and neuropil are shown in red. DESeq2 with FDR <0.05. Marginal rug (gray) represents the distribution of the TE values in the somata (x axis) and neuropil (y axis). (B) Coverage tracks representing the average ribosome footprint or RNA coverage for candidate genes (Syngap1, Kif5c, and Camk2a) in the neuropil and somata. The y axis indicates reads per million (RPM). (C and D) GO terms representing significantly enriched (FDR < 0.05) protein function groups for TElow (C) and TEhigh (D) transcripts. (E) Empirical cumulative distribution frequency (Ecdf) of the TE (log2FC) of SFARI autism associated (yellow) and other (black) genes. P = 2.579e-05, KolmogorovSmirnov test.

In both neuronal compartments, we observed a wide distribution of translation efficiencies, with a greater than 1,000-fold difference between the most and least efficiently translated transcripts in the neuropil (Fig. 4A). We identified 730 and 592 transcripts exhibiting significantly high or low translational efficiencies, respectively, in both somata and neuropil (Fig. 4A and Dataset S4). We identified gene features associated with these two groups which we call TElow and TEhigh. GO analysis revealed an enrichment of terms such as spindle and microtubule organizing center for TElow genes (Fig. 4C). In contrast, TEhigh genes were associated with terms such as intrinsic component of synaptic vesicle membrane and intrinsic component of postsynaptic membrane (Fig. 4D). As a group, TElow transcripts had longer coding sequences (CDS), consistent with previous observations (2527) (SI Appendix, Fig. S5A). Because autism risk factor genes have been described to be exceptionally long (2830), we analyzed the TE values of Simons Foundation Autism Research Initiative (SFARI) transcripts. We found that SFARI transcripts displayed overall lower TE values compared to other genes (Fig. 4E). The efficiency of mRNA translation is also influenced by elements within the UTRs that serve as binding platforms for regulatory RBPs (10, 12). Because longer UTRs harbor more cis-acting elements (10, 12), we examined the 5 and 3 UTR length of the translationally regulated transcripts. We found that TElow genes exhibited significantly longer 5 and 3 UTRs (Fig. 5 A and B). To identify potential RBPs for the neuropil UTRs, we searched for known RBP consensus motifs (31) and determined whether transcript groups sharing the same motifs were associated with higher or lower TE values in the neuropil (Experimental Procedures). A total of 131 3 UTR motifs targeted by 52 RBPs (Dataset S5) were associated with transcripts displaying significantly higher TE values in the neuropil (Fig. 5C; for somata see SI Appendix, Fig. S5B and Dataset S6). For example, consistent with their described role as translational enhancers (3234), HNRNPK and MBNL1 motifs were detected in transcripts exhibiting significantly higher TE values (Fig. 5C). On the other hand, 155 3 UTR motifs targeted by 90 RBPs (Dataset S5) were associated with transcripts exhibiting significantly lower neuropil TE values in the neuropil (Fig. 5C). Among these, we identified, for example, the CPEB, Hu (Elav), and PUF/Pumilio RBP families, all known for their repressive action on translation in neuronal processes (35). We note that none of the RBP motifs we detected within neuropil 5 UTRs were associated with transcripts displaying significantly higher or lower neuropil or somata TE (Datasets S7 and S8). Our results thus reveal the identity of potentially novel regulators that bind the 3 UTR and control translation, either directly or indirectly for example via the regulation of polyadenylation (34) or mRNA decay (35).

Features of translationally regulated transcripts in the somata and neuropil. (A and B) Box plots of 5 UTR (A) and 3 UTR (B) length (log10 nucleotides (nts) for TEhigh (yellow), TElow (blue), and other (gray) genes. Bars indicate 1.5*IQR. *P < 0.05, ****P < 0.0001; one-way ANOVA test followed by pairwise t test with BenjaminiHochberg P value adjustment. (C) Shown are RBP motifs within 3 UTRs associated with significantly lower (blue) or higher (yellow) neuropil TE values (q values < 0.05; Wilcoxon rank sum test) (Experimental Procedures). (D) Detection of translated uORFs in hippocampal neurons. Translation initiation sites were mapped using the drug harringtonine (har), which accumulates ribosomes at start codons. A total of 766 uORF-containing neuronal transcripts were detected in the somata and neuropil. (E) Coverage tracks representing the average ribosome footprint reads along the UTRs (gray), detected uORFs (orange), or the main protein coding sequence (blue) of Dlg4, Gria2, Taok1, and Ppp1r9b in the neuropil. The y axis indicates reads per million (RPM). (F) Observed-to-expected ratio of TEhigh (teal), TElow (blue), and other (gray) transcripts containing uORFs. **P < 0.01, ***P < 0.001, ****P < 0.0001; hypergeometric test. (G) Neuropil TE (log2FC) measurements of transcripts containing translated uORFs (uORF) or not (no uORF). ****P < 0.0001; Welch two-sample t test. (H) GO terms representing the top eight significantly (FDR < 0.05) enriched protein function groups for uORF-containing transcripts in the neuropil.

Upstream open reading frames (uORFs) also play an important role in regulating the translation of the main protein coding sequence (36). While most uORFs are believed to exert a negative effect on the translation of downstream ORFs (36), a few examples of positive-acting uORFs have been reported (37, 38). We identified translated uORFs in neuronal compartments using an integrated experimental and computational approach. To map upstream translation initiation sites within neuronal transcripts, we performed Ribo-seq on neurons treated with the drug harringtonine, which causes the accumulation of ribosomes at start codons (21) (Fig. 5D and Experimental Procedures). We then used the ORF-RATER pipeline to identify and quantify translated uORFs in the neuropil- and somata Ribo-seq data (Experimental Procedures) (39). In total, we identified 766 uORF-containing mRNAs in neuronal compartments (Fig. 5D and Dataset S9), including novel (e.g., Gria2, Taok1, Dlg4, and Ppp1r9b) (Fig. 5E and SI Appendix, Fig. S5C) and previously described (e.g., Atf4 and Ppp1r15b) (38, 40) (SI Appendix, Fig. S5D) transcripts. A comparison of TElow and TEhigh transcripts revealed an overrepresentation of uORF-containing transcripts in the TElow group and an underrepresentation of uORF-containing transcripts in the TEhigh group (Fig. 5F). Additionally, uORF-containing transcripts displayed a significantly lower neuropil median TE value when compared with non-uORF-containing mRNAs (Fig. 5G and SI Appendix, Fig. S5E for the somata). Using the neuropil Ribo-seq data, we next computed a relative uORF to CDS ribosome density for each uORF. Of interest, the relative uORF:CDS ribosome densities ranged from 0.1 to 1,000, indicating a wide spread in the uORF-mediated translational repression in the neuropil (SI Appendix, Fig. S5F). Many uORFs displayed uORF:CDS ribosome density ratios greater than 1, indicating that uORFs often act as CDS translational repressors. A GO analysis indicated that above described uORF-containing neuropil and somata mRNAs were significantly enriched for terms like positive regulation of synapse assembly, regulation of membrane potential, and behavior (Fig. 5H). These findings highlight uORFs as an important translational regulatory element present in many transcripts in somatic and synaptic regions.

Using ribosome profiling, we detected thousands of mRNA species that are translated in synaptic regions, dramatically expanding the contribution of ongoing local protein synthesis to the protein pool detected in dendrites, axons, or synapses (4144). Indeed, among the locally translated mRNAs, we identified most protein families, including signaling molecules (kinases or phosphatases), ion channels, metabotropic and ionotropic receptors, cell adhesion molecules, scaffold proteins, as well as regulators of cytoskeleton remodeling or translation.

Many transcripts were found differentially translated between neuronal compartments. An open question in the field has concerned the contribution of local synthesis to the total pool of a particular protein. Our data indicate that most proteins are synthesized in both compartments. We note that over 800 mRNAs displayed enhanced translation levels in the neuropil, suggesting that most of these proteins arise from a local source. For many transcripts, the abundance of the mRNA was positively associated with the translation level differences between somata and neuropil, as observed previously in developing neurons derived from mouse embryonic stem cells (45). Notably, the neuropil-translation-up transcripts often encoded signaling and scaffold proteins that play an important role in the maintenance and modification of synaptic strength. Of interest, we detected several mitochondrial mRNAs that displayed enhanced neuropil translation. Recently, it has been shown that endosomes can act as platforms for the local translation of candidate mitochondrial mRNAs (46). It is thus tempting to hypothesize that local translation plays a role in sustaining mitochondria, which in turn fuel protein synthesis near synapses during plasticity (47). Together, our results suggest that the increased translation levels of a specific transcript subset in the neuropil likely provide a means to ensure the efficient production of key synaptic proteins at very remote locations from the cell body.

In contrast the transcripts with increased translation levels in the somata often encoded transmembrane proteins. This protein class is typically processed through multiple membrane-bound organelles (including the endoplasmic reticulum [ER] and Golgi apparatus [GA]), where they are folded, assembled, and biochemically modified prior to their delivery to the neuronal cell surface (48). However, recent studies reported that hundreds of neuronal surface proteins (e.g., the AMPAR subunit GluA1) bypass GA maturation and likely travel directly from the ER to the neuronal cell surface (49, 50). Thus, although the bulk synthesis and posttranslational modification of transmembrane proteins might occur in the somatic ER and GA, a small residual fraction of this protein class could undergo on demand local translation to fine tune synaptic strength.

Using a combination of microdissection with Ribo-and RNA-seq, we found that most transcripts exhibit similar translational regulation in the somata and neuropil. In both neuronal compartments, we detected widespread translational regulation, with an unexpectedly high dynamic range in the translation efficiencies of transcripts. Among the mechanisms that regulate the synthesis of proteins in somatic and synaptic regions, we identified uORF-mediated translational control. This finding is in good agreement with previous studies revealing the role of uORFs in the translational regulation of two candidate transcripts in neuronal processes (51, 52). uORF-mediated translational control is often fine tuned by the phosphorylation of eukaryotic initiation factor 2 (eIF2) (53). The phosphorylation of eIF2 inhibits global translation while leading to a paradoxical increase in the translation of a subset of uORF-bearing transcripts (54). Many manipulations of cellular and synaptic activity modulate the phosphorylation status of eIF2 in neurons in vivo and in vitro (5457). Thus, activity-driven eIF2 phosphorylation could act as a switch to enhance the local translational efficiency of uORF-containing transcripts encoding key plasticity-related proteins. It is noteworthy that the translational regulation of some uORF-containing transcripts is insensitive to changes in the eIF2 phosphorylation status (e.g., the protein phosphatase 1 regulatory subunit CReP [Ppp1r15b]) (40).

Electron microscopy (EM) studies have shown that the distribution of the ribosomes along neuronal processes is heterogeneous, with a selective localization of protein-making machines (i.e., polyribosomes, more than three ribosomes per mRNA) beneath synapses, while only a few polyribosomes could be observed in CA1 dendritic shafts (58, 59). Dendritic shafts could be mostly populated by monosomes (i.e., single ribosome per mRNA) that cannot be visualized by EM but also represent active protein making machines in synaptic regions (16). Indeed, a recent superresolution study which likely detects both monosomes and polysomes identified a greater ribosome density in dendrites compared to EM studies (60). These observations raise intriguing questions about the definition of local translation compartments: Are different protein species synthesized within distinct subregions of neuronal processes (e.g., spines vs. dendritic shafts)? And: Could the translation efficiency of the same transcript vary depending on whether it is localized beneath synapses or in other dendritic regions? These questions set the stage for future studies characterizing the translational landscape in neuronal subregions with greater spatial resolution using, for example, proximity-specific ribosome profiling.

Timed pregnant specific-pathogen-free (Charles River Laboratories) female rats were housed in Max Planck Institute for Brain Research animal facility for 1 wk on a 12/12-h light/dark cycle with food and water ad libitum until the litter was born. Cultured neurons were derived from P0 (postnatal day 0) Sprague-Dawley rat pups (both male and female, research resource identifier: 734476). Pups were killed by decapitation. The housing and killing procedures involving animal treatment and care were conducted in conformity with the institutional guidelines that are in compliance with national and international laws and policies (Directive 2010/63/EU; German animal welfare law; Federation of European Laboratory Animal Science Associations guidelines). The animals were killed according to annex 2 of 2 Abs. 2 Tierschutz-Versuchstier-Verordnung. Animal numbers were reported to the local authority (Regierungsprsidium Darmstadt, approval numbers: V54-19c20/15-F126/1020 and V54-19c20/15-F126/1023).

Total Ribo-seq (including monosomes and polysomes) and RNA-seq libraries from microdissected rat somata and neuropil of three biological replicates were generated previously (16) (SI Appendix, Table S1). In short, somata and neuropil were microdissected from 4-wk-old male rats. The tissue samples were homogenized in polysome lysis buffer (20 mM Tris pH 7.5, 150 mM NaCl, 5 mM MgCl2, 24 U/mL TurboDNase, 100 g/mL cycloheximide, 1 mM dithiothreitol (DTT), 1% Triton X-100, and protease inhibitor mixture [Roche]) by douncing in a glass homogenizer. After triturating the lysate 10 times using a 23-gauge syringe, samples were chilled on ice for 10 min and cleared by two centrifugations at 16,100 g for 6 min. From the somata and neuropil lysates Ribo-seq and RNA-seq libraries were prepared simultaneously. For Ribo-seq, neuropil and somata lysates containing equal amounts of total RNA were digested with 0.5 U/g RNase I (Epicentre), shaking for 45 min at 400 rpm at 24C. Nuclease digestion reactions were promptly cooled and spun, and 10 L of SUPERaseIN*RNase inhibitor was added. Samples were then layered onto a 34% sucrose cushion, prepared wt/vol in gradient buffer supplemented with 20 U/L of SUPERaseIN*RNase inhibitor. 80S particles were pelleted by centrifugation in a SW55Ti rotor for 3 h 30 min at 55,000 rpm at 4C. Ribo-seq libraries were prepared according to ref. 61 with the modifications described in ref. 16. Total RNA was isolated from tissue lysates using the Direct-zol RNA micro prep kit (Zymo). RNA integrity was assessed using the Agilent RNA 6000 Nano kit. Rat neuropil and somata total RNA-seq libraries were prepared from an equal amount of total RNA using the TruSeq stranded total RNA library prep gold kit (Illumina) (16). Libraries were sequenced on an Illumina NextSeq500, using a single-end 52- and 75-bp run for Ribo-seq and RNA-seq, respectively.

Neuron-enriched and glia-enriched cultures were prepared from the same litter as described previously (12). The hippocampi of P0-d-old rat pups were isolated and triturated after digestion with papain. Both cultures were plated on 60-mm cell culture dishes. For the preparation of hippocampal neuron-enriched cultures, cells were plated onto poly-d-lysine-coated 60-mm cell culture dishes and treated as described above with Ara-C (Sigma) at a final concentration of 5 M for 48 h. After 48 h, the medium was replaced with preconditioned growth medium and cells were cultured until 21 d in vitro (DIV). For the preparation of glia-enriched cultures, cells were plated onto uncoated 60-mm cell culture dishes in conditioned minimal essential medium (minimal essential medium, 10% horse serum, 0.6% glucose [wt/vol]). At 7 DIV, the medium was replaced with preconditioned growth medium and cells were cultured until 21 DIV. Four independent biological replicates were prepared. RNA was isolated using the Direct-zol RNA micro prep kit (Zymo). RNA integrity was assessed using the Agilent RNA 6000 Nano kit. mRNA-seq libraries were prepared starting from 200 ng of total RNA, using the TruSeq stranded mRNA library prep kit (Illumina). Libraries were sequenced on an Illumina NextSeq500, using a single-end, 75-bp run.

The input- and translating ribosome affinity purification (TRAP)-seq libraries from hippocampi of Camk2a-Cre-RiboTag or somata/neuropil sections of Wfs1-Cre-RiboTag mice were generated previously (16) (SI Appendix, Table S1).

Dissociated rat hippocampal neurons were prepared from P0-d-old rat pups as described previously (62). Hippocampal neurons were plated at a density of 31,250 cells/cm2 onto poly-d-lysine-coated 100-mm dishes and cultured in preconditioned growth medium (Neurobasal-A, B27, GlutaMAX, 30% glia-culture supernatant, 15% cortex-culture supernatant) for 21 DIV. At 1 DIV, cells were treated with Ara-C (Sigma) at a final concentration of 5 M to prevent the overgrowth of nonneuronal cells. After 48 h, the medium was replaced with preconditioned growth medium and cells were cultured until 21 DIV. Cells were fed with 1 mL of preconditioned medium every 7 d. Three independent biological replicates were prepared. At 24 h before drug treatment, cell medium was adjusted to 8 mL per dish. In appropriate experiments, harringtonine (LKT Laboratories) was added to a final concentration of 2 g/mL from a 5 mg/mL stock in 100% ethanol. Cells were returned to the incubator at 37C for 15, 30, 45, 90, or 150 s. Cycloheximide was added to a final concentration of 100 g/mL from a stock of 50 mg/mL in 100% ethanol. After drug addition, cells were returned to the incubator at 37C for 1 min. After the incubation with cycloheximide, the cells were immediately placed on ice and washed twice with ice-cold phosphate-buffered saline (PBS) plus 100 g/mL cycloheximide and scraped in polysome lysis buffer (20 mM Tris pH 7.5, 150 mM NaCl, 5 mM MgCl2, 24 U/mL TurboDNase, 100 g/mL cycloheximide, 1 mM DTT, 1% Triton-X-100, and protease inhibitor mixture [Roche]) (21). After scraping, the lysates were triturated 10 times using a 23-gauge syringe; samples were chilled on ice for 10 min and then cleared by centrifugation at 16,100 g for 10 min. Ribo-seq libraries from rat hippocampal neuron cultures treated for 0, 15, 30, 45, 90, and 150 s with harringtonine were prepared as described above. The 0-, 30-, and 90-s datasets were previously published in ref. 16 (SI Appendix, Table S1).

Four-week-old male rats were perfused with 1 RNase-free PBS and fixative solution (4% (vol/vol) paraformaldehyde (PFA), 4% (wt/vol) sucrose in 1 RNase-free PBS). Brains were dissected and fixed for another hour at room temperature. Brains were cryoprotected for two consecutive days at 4C. In 15% (wt/vol) sucrose in RNase-free 1 PBS on day 1, followed by 30% (wt/vol) sucrose in RNase-free 1 PBS on day 2. Hippocampi were cryosectioned at 30-m thickness.

Fluorescence in situ hybridization was performed using the QuantiGene ViewRNA kit (Thermo Fisher) mostly following the manufacturers instructions. In brief, hippocampal slices were postfixed for 10 min at room temperature in fixative solution (4% [vol/vol] PFA, 5.4% [wt/vol] glucose, 0.01 M sodium metaperiodate in 1 lysine-phosphate buffer). The manufacturer recommended proteinase K treatment was omitted to preserve the integrity of the dendrites. Slices were permeabilized for 20 min using the kits detergent buffer. Detection probes were incubated overnight at 40C. Preamplification, amplification, and label probes were incubated for 60 min at 40C, respectively, washing three times for 5 min between each step. After completion of in situ hybridization, slices were washed with 1 PBS and incubated in blocking buffer (4% [vol/vol] goat serum 1 PBS) for 1 h at room temperature. The primary antibody (gp-anti-MAP2, SYSY 188004, 1:1,000) was incubated overnight in blocking buffer at 4C. Slices were washed five times for 10 min in 1 PBS and the secondary antibody (gt-anti-gp Alexa 647, Thermo Fisher A21450, 1:500) was incubated in blocking buffer for 5 h at room temperature. Slices were washed in 1 PBS and nuclei were stained with DAPI for 3 min at room temperature. Slices were mounted in AquaPolyMount.

Slices were imaged using a Zeiss LSM780 confocal microscope and a 40 oil objective (numerical aperture [NA] 1.3). Z stacks spanning the entire slice volume were obtained using appropriate excitation laser lines and spectral detection windows. The mRNA signal was dilated for better visualization. The raw, nondilated images were used for analysis.

An in-house Python script was used to count mRNA puncta in the somata and the neuropil layer, respectively. In the neuropil, puncta colocalizing with DAPI signal (arising from glia or interneurons) were excluded from the analysis. Counts were normalized by area and a neuropil-to-somata ratio was computed for each slice. The mean neuropil-to-somata ratio was calculated for somata-translation-up target genes. All neuropil-to-somata ratios were divided by this average.

Sequencing adapters were trimmed using the Cutadapt software version 1.15 (63) with the following arguments: cut 1minimum-length 22 discard-untrimmed overlap 3 -e 0.2. An extended unique molecular identifier (UMI) was constructed from the two random nucleotides (nts) of the reverse transcription primer and the five random nucleotides of the linker and added to the FASTQ description line using a custom Perl script. To remove reads originating from noncoding RNA (ncRNA, i.e., rRNA), trimmed reads were aligned to rat ncRNA using Bowtie2 version 2.3.5.1 (very-sensitive) (64) and aligned reads were discarded. The remaining reads were aligned to the rat genome (rn6) with the split-aware aligner STAR version 2.7.3.a (65) with the following arguments: twopassMode Basic twopass1readsN -1 seedSearchStartLmax 15 outSJfilterOverhangMin 15 8 8 8 outFilterMismatchNoverReadLmax 0.1. To retrieve transcript coordinates, STARs quant mode (quantMode) was used. Throughout the study, genome alignments were used for differential expression analyses and genomic feature analyses. Transcriptome alignments were used for all other analyses. The STAR genome index was built using annotation downloaded from the University of California Santa Cruz (UCSC) table browser (66). PCR duplicates were suppressed using a custom Perl script and alignments flagged as secondary alignment were discarded before analysis. Only footprints with sizes between 24 and 34 nts were used for analyses.

Sequencing adapters and low-quality nucleotides were trimmed using the Cutadapt software version 1.15 (63) with the following arguments: minimum-length 25nextseq-trim = 20. The trimmed reads were aligned to the rat (rn6) or the mouse (mm10) genome with STAR version 2.7.3a (65).

The coordinates of genomic features (CDS, 3 UTR, 5 UTR, intron) were downloaded from the UCSC table browser in BED format (66). Bedtools version 2.26.0 (67) was used to convert BAM into BED files and to identify reads overlapping with the individual features.

P-site offsets were defined for different footprint lengths. Each footprint start position defined the footprint frame in reference to the annotated start codon. The footprint reads were virtually back projected over the start codon and the offsets from the start and the end of the read were calculated. We used every read of a given length and accumulated the most probable offset and frame. Next, the P-site position per footprint read was deduced from its length and the previously determined offset. All P-site positions were plotted for 100 nucleotides around the start and stop codons, and the center of a transcript. To correct for differences in translation rates between genes, the P-site coverage of each gene was normalized to its mean footprint coverage. The nucleotide coverage at the 0, 1, and 2 frame positions were assessed. A one-way analysis of variance (ANOVA) was used to determine if the observed frame fraction was different from the expected frame fraction. A significant P value rejected the null hypothesis that all frames featured the expected P-site coverage.

Footprint alignments were converted into the BedGraph file format using Bedtools version 2.26.0 and visualized as custom tracks on the UCSC Genome Browser (68). Footprint coverages were corrected for sequencing depth.

For both total RNA sequencing and ribosome footprint libraries from the somata and neuropil, the software featureCounts version 2.0.0 (69) was used to calculate counts per gene from reads that were aligned to the rat genome. All annotated transcript isoforms were considered. Raw counts were fed into DESeq2 version 1.30.1 and log fold change (LFC) shrinkage was used (18). Only genes with an adjusted P value are displayed in Fig. 1B.

The software featureCounts version 2.0.0 (69) was used to calculate counts per gene from reads mapped to the genome (mm10, rn6). All annotated transcript isoforms were considered. Raw counts were fed into DESeq2 version 1.30.1 and LFC shrinkage was used (18).

Gene ontology analysis was performed for neuropil- and somata-translation-up genes. All detected genes (baseMean greater than zero and with an adjusted P value), without the contaminants, were used as background. GO enrichment analysis was performed for the complete cellular component annotation using the PANTHER overrepresentation test (70, 71). The Fisher exact test was used and only GO terms with a false discovery rate (FDR) smaller than 0.05 were considered. The most specific GO terms per branch were retained. The top five GO terms with the highest enrichment scores were visualized.

Gene ontology analysis was performed for uORF-containing transcripts. All detected genes in the neuropil and the somata (baseMean greater than zero), without the contaminants, were used as background. GO enrichment analysis was performed for the complete biological process annotation using the PANTHER overrepresentation test (70, 71). The Fisher exact test was used and only GO terms with an FDR smaller than 0.05 were considered. The most specific GO terms per branch were retained. All significant GO terms were visualized.

Gene ontology analysis was performed for TEhigh and TElow transcripts. All detected genes in the neuropil and the somata (baseMean greater than zero), without the contaminants, were used as background. GO enrichment analysis was performed for the complete cellular component annotation using the PANTHER overrepresentation test (70, 71). Only GO terms with at least 50 genes in the background set were used in the analysis. The Fisher exact test was used and only GO terms with an FDR smaller than 0.05 were considered. The most specific GO terms per branch were retained. All significant GO terms were visualized.

The number of ribosomes per transcript was estimated by integrating Ribo-seq and RNA-seq libraries to calculate TE values in the neuropil. Raw Ribo-seq and RNA-seq counts, falling into gene CDS, were fed into DESeq2 version 1.30.1 and LFC shrinkage was used (18). TE values that were either significantly higher than log2(1.5) in the neuropil and the somata or smaller than log2(1.5) in the neuropil and the somata were assigned to TEhigh and TElow, respectively [lfcThreshold = log2(1.5) with an FDR < 0.05]. Only genes with a baseMean greater than 10 in the neuropil and the somata were considered. An interaction term was added to the experimental design to compare TE values between the neuropil and the somata (72).

Genes known to be associated with autism spectrum disorders were downloaded from the SFARI Gene database (https://www.sfari.org). Human gene symbols were converted into rat gene symbols. Genes with an SFARI score of 1 and 2 were considered as autism genes.

RBP motifs (human, rat, and mouse) were downloaded as position weighted matrices from the public ATtRACT database (31). The FIMO tool from the MEME suite version 5.1.1 was used to scan 5 and 3 UTRs for motif occurrences, using the default threshold (P value = 1e-4) and a precalculated nucleotide background model derived from query sequences (73). Only genes with an RBP motif occurrence were considered for analysis. For each identified RBP motif, the motif-containing genes were grouped and a median TE value was calculated. A Wilcoxon rank sum test was conducted to test if the median TE of a given RBP motif group differed from the median TE of all genes that do not contain the motif.

The ORF-RATER pipeline (https://github.com/alexfields/ORF-RATER) was run as previously described (39), starting with the harringtonine 150 s as well as the neuropil and somata BAM files. Note, that it is possible that a translated uORF may be assigned a low score, as ORF-RATER is tuned to indicate the highest-confidence sites of translation, at the expense of an increased false negative rate (74). The following parameters were used: --codons NTG for ORF types, --minrdlen 28 --maxrdlen 34 for harringtonine-treated samples, --minrdlen 27 --maxrdlen 34 for neuropil and somata samples. Only uORFs with a score of at least 0.7, a length of at least three codons, and at least one count in each of the neuropil and the somata replicates were considered.

The ribosome density of a uORF or CDS was computed as the number of ribosome footprints divided by the uORF or CDS length, respectively. The relative ribosome density was computed as the uORF ribosome density divided by the CDS ribosome density.

The 5 and 3 UTR lengths were calculated based on the Rattus norvegicus annotation version 6 (rn6). The 3 UTR lengths were corrected in accordance with newly identified 3 UTR isoforms described in ref. 12. For genes with multiple 5 UTR isoforms the longest 5 UTR sequence was chosen, giving priority to curated isoforms. For genes with multiple 3 UTRs, the most-expressed 3 UTR isoform was chosen (12).

For the comparison of 5 UTR lengths between TEhigh, TElow, and others only 5 UTRs with a minimum length of 10 nts and a maximum length of 5,000 nts were considered. For the comparison of 3 UTR lengths between TEhigh, TElow, and others, only 3 UTRs with a minimum length of 50 nts and a maximum length of 10,000 nts were considered.

For the comparison of 3 UTR lengths between neuropil-translation-up and somata-translation-up genes, the 3 UTR isoform with the highest expression in the hippocampus per gene family was considered (12).

The coverage of each gene was projected along the CDS in transcript coordinates (only exons). Genes with CDS lengths shorter than 440 codons were omitted from analysis. Each metagene profile was scaled by the average coverage between codon 400 and 20 codons before the stop codon. For each time point, the metagene profiles were smoothed with a running average window of 30 codons. For each group, the coverage tracks were accumulated, averaged, and normalized to the 0-s condition. A baseline coverage track was defined as 85% of the nontreated sample coverage track. The first positive crossing between the harringtonine-treated coverage track and the baseline coverage track determined the crossing position in codons. Elongation rates were calculated as the slope of a linear regression between the harringtonine incubation times for each track and the crossing position in codons.

Statistical significance and the tests performed are indicated in the figure legends. Statistical analysis was performed using MATLAB and R.

Details about data availability can be found in SI Appendix, Table S1. The accession number for the raw sequencing data published previously in ref. 16 is National Center for Biotechnology Information (NCBI) BioProject: PRJNA550323. The accession number for the raw sequencing data reported in this paper is NCBI BioProject: PRJNA634994. All bioinformatic tools used in this study are contained in one modular C++ program called RiboTools. The source code and further notes on the algorithms can be found on our GitHub repository (DOI: 10.5281/zenodo.3579508). Other analysis scripts and codes are available upon request.

We thank Elena Ciirdaeva for help with mRNA library preparation. A.B. is supported by a European Molecular Biology Organization (EMBO) long-term postdoctoral fellowship (EMBO ALTF 331-2017). E.M.S. is funded by the Max Planck Society, an Advanced Investigator award from the European Research Council (Grant 743216), Deutsche Forschungsgemeinschaft (DFG) Collaborative Research Centre (CRC) 1080: Molecular and Cellular Mechanisms of Neural Homeostasis, and DFG CRC 902: Molecular Principles of RNA-Based Regulation.

Author contributions: C.G., A.B., and E.M.S. designed research; C.G., A.B., B.N.-A., A.K., I.B., and S.t.D. performed research; C.G., A.B., and G.T. analyzed data; and A.B. and E.M.S. wrote the paper.

Reviewers: C.M.A., The University of Edinburgh Centre for Genomic and Experimental Medicine; and E.K., New York University.

The authors declare no competing interest.

See online for related content such as Commentaries.

This article contains supporting information online at https://www.pnas.org/lookup/suppl/doi:10.1073/pnas.2113929118/-/DCSupplemental.

Read the original:
The translatome of neuronal cell bodies, dendrites, and axons - pnas.org

Virtual Care Market to Witness Exponential Growth by 2031 – BioSpace

Global Virtual Care Market: Snapshot

Virtual care refers to a technique that allows for the treatment of patients dealing with different health issues with the help of advanced technologies such as audio, video, or written communication. Moreover, it also includes virtual visits performed using communication devices held by patients as well as physicians from diverse places.

Read Report Overview - https://www.transparencymarketresearch.com/virtual-care-market.html

TMRs upcoming research report provides comprehensive study of all factors influencing the development of the global virtual care market. Thus, it gives inclusive assessment of important facets such as drivers, trends, restraints, challenges, and growth opportunities in the market. In addition, this assessment document offers dependable statistics on sales, volume, revenues, and shares of the market for virtual care.

The report analysts have performed segmentation of the global virtual care market on the basis of several important parameters such as consultation type, end-user, and region. On the basis of consultation type, the market is classified into audio consultation, kiosks, and video consultation.

Request Brochure of Report - https://www.transparencymarketresearch.com/sample/sample.php?flag=B&rep_id=83552

Key Drivers of Virtual Care Market Growth

In the healthcare sector, there is notable growth in the application of different advanced technologies such as virtual care owing to the flexibility provided by the connected devices. Moreover, people today are inclining toward the use of virtual care services as they get an opportunity to gain second opinions from qualified healthcare professionals through online channels.

Virtual care is utilized by patients for performing varied activities such as consultations, meetings, check-ins, and checking the status of their reports. In addition, this technique can be utilized in the management of diseases that need continual follow ups. Thus, increased number of individuals suffering from critical health issues such as hypertension and diabetes is expected to support in the rapid expansion of the global virtual care market in the years to come.

Request for Analysis of COVID-19 Impact on Virtual Care Market- https://www.transparencymarketresearch.com/sample/sample.php?flag=covid19&rep_id=83552

Virtual care solutions are increasingly adopted across major parts of the globe as they offer a wide range of advantages such as accessibility to doctors or healthcare providers with the help of video conferencing, which can be a prominent option in case of medical emergencies in remote areas.

The virtual care technique is adopted by healthcare specialists as they can focus on critical cases, as the technology gives them direct access to the patient medication room or to the hospital even if they are not physically present at that particular place.

Request for Custom Research - https://www.transparencymarketresearch.com/sample/sample.php?flag=CR&rep_id=83552

What Key Strategies are Utilized by Companies in Global Virtual Care Market to Stay Ahead in Competition

The global virtual care market is fragmented in nature and its competitive landscape is highly intense. Players are utilizing diverse strategies to maintain their prominent market positions. Some of the key strategies utilized by market enterprises are partnerships, collaborations, and mergers and acquisitions.

Pre Book Virtual Care Market Report - https://www.transparencymarketresearch.com/checkout.php?rep_id=83552&ltype=S

North America Demand Outlook for Virtual Care

In terms of region, the global virtual care market shows existence in many regions such as Europe, Asia Pacific, North America, South America, and Middle East and Africa. Among all regions, North America is one of the dominant regions of the market for virtual care.

The North America virtual care market is estimated to maintain its dominant position in the forthcoming years due to early adoption of advanced technologies in the region.

More Trending Reports by Transparency Market Research:

Polymer Implantables Market Implantables, commonly known as prosthetics, are medical devices which are used to replace missing or abnormal body parts or organs. Various biomaterials are used for the development of implants such as metals, polymers, ceramics, and composites.

Microneedle Drug Delivery System Market Microneedles are used for micro-projections of drugs. Microneedles are the pathways to biological membranes which can create transport pathways of micron dimensions for sampling body fluids, such as for measuring the blood glucose levels in diabetic therapy.

Induced Pluripotent Stem Cells Market Induced pluripotent stem cells (iPSCs or iPS cells) are a form of pluripotent stem cells, generated directly from somatic (adult) cells. Pluripotent stem cells have significant potential in regenerative medicine. These possess the potential and capability to form various types of adult cells such as liver, heart, neurons, and pancreatic cells.

About Us

Transparency Market Research is a next-generation market intelligence provider, offering fact-based solutions to business leaders, consultants, and strategy professionals.

Our reports are single-point solutions for businesses to grow, evolve, and mature. Our real-time data collection methods along with ability to track more than one million high growth niche products are aligned with your aims. The detailed and proprietary statistical models used by our analysts offer insights for making right decision in the shortest span of time. For organizations that require specific but comprehensive information we offer customized solutions through ad hoc reports. These requests are delivered with the perfect combination of right sense of fact-oriented problem solving methodologies and leveraging existing data repositories.

TMR believes that unison of solutions for clients-specific problems with right methodology of research is the key to help enterprises reach right decision.

Contact Mr. Rohit Bhisey Transparency Market Research State Tower, 90 State Street, Suite 700, Albany NY - 12207 United States USA - Canada Toll Free: 866-552-3453 Email: sales@transparencymarketresearch.com Website: https://www.transparencymarketresearch.com/

Visit link:
Virtual Care Market to Witness Exponential Growth by 2031 - BioSpace

The Rise of Longevity Therapeutics – Pharmaceutical Executive

Aging is the ultimate risk factor for most diseases, such as cancer, neurodegenerative, cardiovascular, diabetes, degenerative fibrosis and many others. When we are young, we are typically healthy, despite a predisposition that will lead inevitably to a specific degenerative condition. However, the degenerative processes do not kick in until a certain age, when we are older. It looks like when we are younger, the body can compensate cumulative stress and damage caused to our cells in the tissues, allowing to maintain that equilibrium, called homeostasis, that keeps our organs functional and healthy. However, over time this buffering capacity becomes thinner and thinner, until things wear off: our tissues stop working as they used to. These changes are typically caused by an initial small number of rare but bad cells, that progressively increase over time, causing additional damage to the good cells that eventually stop working efficiently, causing a vicious cycle. Eventually the bad cells take over leading to the onset of a disease.

Our body is equipped with a number or regenerative and healing functions. Some are intrinsic in every cell, such as DNA repair mechanisms that are triggered when something compromises the integrity of our genomic structures. These are important functions that enable a cell, for example when it replicates, to repair errors and other damages that might have happened to our DNA. For example, two large proteins called ATM and ATR, involved in the cellular response to DNA damage, are responsible to maintain genomic instability caused by intrinsic and external DNA-damaging agents, such as UV light or various chemicals and toxins. A lack of functions of these proteins results in progressive neurodegeneration, immunodeficiency, predisposition to malignancy or radiation sensitivity. Mutations on the genes encoding these proteins can cause premature aging and premature development of these diseases, but this occurs also naturally, over time.

Cells also have an intrinsic immune system, producing factors called interferons employed by the cells as antiviral agents and to modulate other immune functions. It can be triggers by a viral infection so when a cell is infected will release interferons, protecting the neighbor cells against potential infection. Interferons can also suppress growth of blood vessels preventing tumors to get nutrients and growing. They can also activate immune cells so they can better fight viruses, tumors and others agents. Unfortunately, an age-related decline or impaired innate interferon functions in the cells results in a number of negative consequences in the body, such as increased susceptibility of the elderly to infections, tumors and damage.

In the body there are several cell types responsible to keep the tissues in check. The immune system is specialized to recognize remove and remember damaging agents. Those could be external, such as virus, bacteria or parasites, or internal, such as tumorigenic cells or senescent cells (see below). The immune system is a very sophisticated network of cell types, intercommunicating with each other to maintain the body clean from damaging factors. As we age the immune system also ages and loses capacity to recognize or responding to these damaging agents. It also become exhausted by an increasing chronic inflammation that progressively accumulate as we age, phenomenon also called inflammaging.

Another important repairing mechanism is the regenerative tissue functions, driven by the stem cells. Those cells are progenitor cells, often dormant in a quiescent state in the tissue and waiting to be activated by some damage. Stem cells are critical because once activated they can generate a progeny of daughter cells capable of re-growing the damaged tissue back to its original structure and function. Stem cells have another important function: they can regenerate themselves, in a process called self-renewal. This is important so that the new repaired tissue can repeat the process if a new damage occurs. The regenerative capacity of our body is remarkable, allowing our tissues to keep their integrity, health and functions. However, over time also stem cells age or respond to the aged microenvironment where they live (called the niche), and they become less efficient to repair tissues or to self-renewing. As a result, our tissues change, become atrophic, fibrotic or dysfunctional leading eventually to diseases.

In regenerative medicine, the application of stem cells resulted of the generation of multiple new therapeutic opportunities. A promising area uses stem cells to generate bioengineering strategies to grow new tissues in a petri dish to be then transplanted in the body to repair damaged tissues. Some applications are already in clinical use, such as for skin grafts. Many others are on their way, either in preclinical development or in clinical trials for many different tissue types and for different clinical indications.

Another promising stem cells application is the direct transplantation into damaged tissues, where they can grow and engraft repairing. However, as we age stem cells become less efficient. What if we If we could rejuvenate them? We could restore their capacity to repair our tissues and maintain homeostasis. Promising and exciting strategies are advancing in that direction. For example, we and others showed that it is possible to reprogram epigenetically a cell so it can become the younger and healthier version of itself (Sarkar et al., 2020). This is a mechanism that every cell has encoded in its DNA, but normally works only in the germline (the sperm and the egg) during the embryogenesis to make sure that the cellular clock is turned back to zero, before initiating the cellular programs to generate the embryo. This important for example to prevent making old newborn babies. This intrinsic rejuvenative mechanism is locked in the other somatic cells of the body. We found it is possible to re-activate it transiently and safely, without changing the identity of the cell, enabling to push back the cellular clock of aged human cells to make them healthier and restore their functions. These technologies are under development to be translated into therapeutics with the promise that one day could rejuvenate the aged cells in the body so they can become the younger version of themselves, repeating the process over time when needed.

Among many of the drivers of the aging process, there is one that seems to stands out as the lower hanging fruit among the emerging space of the longevity therapeutics. This is cellular senescence. Every damage that occurs to the cells in our body can push the cells to stop what they are doing and activate a safety mechanism that locks them into an arrested state called cellular senescence. Senescent cells cannot replicate anymore preventing them to cause additional damage, such as becoming cancer cells. All sort of damage can trigger this response leading to cellular senescence such as, oxidative stress, mitochondrial dysfunctions, DNA damage, viral infection, cigarette smoking, pollutions, chemicals, etc. They all can induce that safety lock and push damage cells to become senescent.

Senescent cells dont die easily but they stick around in the tissue, accumulating slowly over time. Importantly, cellular senescence is a pleiotropic mechanism, meaning it can be both good or bad. When we are young, we can efficiently get rid of senescent cells. The body uses them positively such as for tissue repair, wound healing or tissue remodeling. However, as we age, and our immune system ages (partially trough cellular senescence, a phenomenon called immune-senescence), our body become less efficient in removing senescent cells, which then start to accumulate.

Being able to make a new generation of drugs that are very selective for senescent cells, will enable the promise to achieve rejuvenative clinical results in humans similarly to what we found in preclinical results. On that end, we recently published a targeted strategy with the goal to advance the field in that direction (Doan et al., 2020). Using a prodrug, we engineered a small molecule to generate a selective senolytic compound to develop a targeted therapy. This prodrug is inactive in non-senescent cells but activated by senescent cells, taking advantage of an enzymatic function of those cells. In geriatric mice this prodrug showed to be well tolerated but also efficacious to clear senescent cells, resulting in restored cognitive functions, muscle functions, stem cells functions, vitality and overall health. As we advance senolytic drugs to the clinic to treat age-related diseases, it is very important to be mindful that elderly individuals, who are frail, with co-morbidities and exposed to multiple medications, will not well tolerate drugs that are not safe and effective. Importantly, not all senescent cells are the same. They are rare, interspersed in the tissues but are also very heterogeneous. Being able to hit the right senescent cells, in the right diseased tissue will be key to enable effective therapies. Developing drugs that are very potent, selective and potent and safe will be pivotal.

The longevity therapeutics space is emerging, but is already disrupting the medical industry. The goal of longevity therapeutics is not just to add years to life, extending lifespan. The true goal is to add life to years and extend health span. A target that gets closer every day.

Marco Quarta is CEO, Rubedo Life Sciences.

See the original post:
The Rise of Longevity Therapeutics - Pharmaceutical Executive

Tooth Regeneration Market Size | COVID-19 Impact Analysis | Forecast to 2027 investigated in the latest research – WhaTech

Tooth Regeneration Market Global Trends, Market Share, Industry Size, Growth, Opportunities and Market Forecast - 2021 to 2027. The tooth regeneration market size is expected to grow significantly from 2021 to 2027.

The tooth regeneration marketsize is expected to grow significantly from 2021 to 2027.

Tooth regeneration is a stem cell-based regenerative medical procedure used in the fields of tissue engineering and stem cell biology. The tooth regeneration procedure replaces damaged or lost teeth by growing on autologous stem cells.

Somatic cells are collected and reprogrammed to derive pluripotent stem cells and tooth layers with the help of resorbable biopolymers.

(Getthis Report)

A full report of Global Tooth Regeneration Market is available at: http://www.orionmarketreports.com/tooth-rket/55394/

Market Segments

By Type

By Process

By End-use

Key Players

Key players operating in the global tooth regeneration market include Unilever, Ocata Therapeutics, Integra LifeSciences, CryoLife, Inc., BioMimetic Therapeutics, Inc. (Wright Medical Group, Inc.), Cook Medical, and StemCells Inc.

Scope of the Report

The research study analyzes the global Tooth Regeneration industry from 360-degree analysis of the market thoroughly delivering insights into the market for better business decisions, considering multiple aspects some of which are listed below as:

Recent Developments

o Market Overview and growth analysis o Import and Export Overview o Volume Analysis o Current Market Trends and Future Outlook o Market Opportunistic and Attractive Investment Segment

Geographic Coverage

o North America Market Size and/or Volume o Latin America Market Size and/or Volume o Europe Market Size and/or Volume o Asia-Pacific Market Size and/or Volume o Rest of the world Market Size and/or Volume

Key Questions Answered by Tooth Regeneration Market Report

1. What was the Tooth Regeneration Market size in 2019 and 2020; what are the estimated growth trends and market forecast (2021-2027).

2. What will be the CAGR of the Tooth Regeneration Market during the forecast period (2021-2027)?

3. Which segments (product type/applications/end-user) were most attractive for investments in 2021? How these segments are expected to grow during the forecast period (2021-2027).

4. Which manufacturer/vendor/players in the Tooth Regeneration Market was the market leader in 2020?

5. Overview on the existing product portfolio, products in the pipeline, and strategic initiatives taken by key vendors in the market.

The report covers the following objectives:

About Us:

Orion Market Reports (OMR) endeavors to provide an exclusive blend of qualitative and quantitative market research reports to clients across the globe. Our organization helps both multinational and domestic enterprises to bolster their business by providing in-depth market insights and the most reliable future market trends.

Our reports address all the major aspects of the markets providing insights and market outlook to global clients.

This email address is being protected from spambots. You need JavaScript enabled to view it.

Original post:
Tooth Regeneration Market Size | COVID-19 Impact Analysis | Forecast to 2027 investigated in the latest research - WhaTech