The sparrow’s dying song: a possible path toward natural, stem cell-based repair of human brain diseases

Songbird research? How the heck could studying tweeting birds lead to advancements in human health?

At a first glance, biological research in other organisms like bacteria, yeast, flies, mice and birds can seem frivolous and a waste of taxpayer money. Yet it’s astonishing how we humans share very similar if not identical functions at a cellular level with our fellow creatures on Earth. So unraveling underlying biological processes in less complex animals is essential to better understanding human biology and to finding possible paths for treating human disease.

Gambel's White-crown sparrow: could its song unlock methods for repairing the brain? (photo courtesy Lip Kee, wikimedia commons)

Gambel’s White-crown sparrow: could its song unlock methods for repairing the brain? (photo courtesy Lip Kee, wikimedia commons)

Case in point: research published in the Journal of Neuroscience last week suggests that studying brain stem cells in song birds could one day lead to methods for naturally repairing neurodegenerative disorders such as Alzheimer’s disease in humans.

The University of Washington team behind the report studies the seasonal song behavior of Gambel’s white-crown sparrows. During the spring breeding season, the population of cells in the sparrow’s brain that are responsible for singing double in number. This cell growth helps the bird to be at its peak singing performance for attracting mates and staking its territory. As breeding season recedes, these brain cells die away naturally and the sparrow’s song, no longer needed, deteriorates. When the next spring arrives the brain cells will grow again.

Audio tracing's of the sparrow's song show its degradation after breeding season each year. (T. Larson/Univ. of Washington)

Audio tracings of the sparrow’s song show its degradation after breeding season each year. (image: T. Larson/Univ. of Washington)

The team’s fascinating discovery is that the dying brain cells themselves appear to provide a signal that tells brain stem cells to multiply for the next breeding season. The scientific term for the cell die-off is called programmed cell death, or apoptosis (pronounced A-POP-TOE-SIS). There are chemicals available to block apoptosis signals. And when the research team administered these anti-apoptosis chemicals at the end of the breeding season, there was a significant reduction in newly dividing brain stem cells. This result shows that new brain stem cell growth depends on the death of brain cells associated with song.

The next step in the project is to identify the signal from the dying cells that stimulates new brain stem cell growth. Once identified, that signal could be harnessed to naturally stimulate new brain stem growth to help repair the loss of brain cells seen in aging, Parkinson’s or Alzheimer’s disease.

As he mentions in a university news story, Dr. Eliot Brenowitz, the senior author of the report, is optimistic about their prospects:

“There’s no reason to think what goes on in a bird brain doesn’t also go on in mammal brains, in human brains. As far as we know, the molecules are the same, the pathways are the same, the hormones are the same. That’s the ultimate purpose of all this, to identify these molecular mechanisms that will be of use in repairing human brains.”

To learn about CIRM-funded projects related to neurodegenerative disorders, visit our Alzheimer’s and Parkinson’s online fact sheets.

Stem cell stories that caught our eye: heart disease, premature infants and incontinence

Here are some stem cell stories that caught our eye this past week. Some are groundbreaking science, others are of personal interest to us, and still others are just fun.

Decoding heart health and genetics in Asians. A study from CIRM grantee Joseph Wu at Stanford may point the way to using stem cells to solve problems caused by too many drugs being tested predominantly on white males. Ethnic variations to drug response too often get ignored in current clinical trials.

The Stanford team has used iPS type stem cells to create a disease-in-a-dish model of a genetic mutation that effects 500 million people, but mostly East Asians. The mutation disables the metabolic protein called ALDH2 and results in increased risk of heart disease and increases the risk of death after a heart attack. By growing heart muscle from stem cells made from the skin of patients with the mutation his team found that the defect alters the way the heart cells react to stress.

Wu suggests that drug companies one day may keep banks of iPS cells from various ethnic groups to see how their responses to drugs differ. Science Daily ran the university’s press release.

Stem cells may treat gut disease in premies.
A laundry list of medical challenges confronts premature babies, but few are as deadly as the intestinal disease that goes by the name NEC, or necrotizing enterocolitis. It strikes with no notice and can kill within hours.

140925100256-largeA team at the University of Ohio reports they have developed what may be a two-pronged attack on the disease. First, they found a biomarker that can predict which infants might develop NEC, and second they have tested stem cells for treating the intestinal damage done by the disease. In an animal model they found that a type of stem cell found in bone marrow, mesenchymal stem cells, can reduce the inflammation that causes the damage and that neural stem cells can repair the nerve connections disrupted by the inflammation.

While this explanation sounds straight forward, getting to that potential intervention was anything but a simple path. The university wrote an extensive feature detailing the many years and many steps the research team took to unravel this who-done-it that involves the gut’s extensive “brain” and immune system. Science Daily picked up the piece.

We recently posted a video about a project we fund using stem cells to develop a treatment for irritable bowel disease.

Fat stem cells tested in incontinence. For far too many older women laughing and coughing can lead to embarrassing bladder leaks. Several groups are working with various types of stem cells to try to strengthen the urinary sphincter and help patients lead a more normal life. A team at Cleveland Clinic now reports some positive results using the most easily accessed form of stem cells, those in fat.

They harvested patients’ own fat stems cells, grew them in the lab for three weeks and then mixed them with a collagen gel from cows to hold them in place before injecting them into the sphincter. Three of five patients passed “the cough test” after one year. Good results, but clearly more work needs to be done to yield more uniform results. Stem Cells Translational Medicine published the research and issued this press release.

Some researcher suspect starting with an earlier stage, more versatile stem cell might yield better results. One of our grantees is developing cells to treat incontinence starting with reprogrammed iPS type stem cells.

New course looks at where fact and fiction overlap. I am a big fan of almost any effort to blend science and the arts. A professor at the University of Southern California seems to agree. CIRM grantee Gage Crump will be teaching a course next spring about science fiction and stem cells.

The university says the course, Stem Cells: Fact and Fiction, will range from babies born with three biological parents to regrown body parts. The course will explore the current state of stem cell biology as it closes the gap between reality and the sci fi visions of authors such as Margaret Atwood and Philip K. Dick. Crump describes it as:

“a mad scientist type of course, where we go through some real science but also [think] about what’s the future of science.”

Don Gibbons

Museum exhibit explaining stem cell super heroes opens in Canada today, due in California in 2016

7108285_origAn international touring exhibit using super hero cells as guides to explain the many roles of stem cells in our lives opens today at the Sherbrooke Museum of Nature and Science in Canada. Its five-year tour will include further displays in Canada, the United Kingdom and three stops on California—the San Francisco Bay area, Los Angeles and San Diego—in 2016.

Super Cell logoDesigned for the general public, with a special eye to children, the exhibit uses hands-on and interactive modules to show just how important stem cells are not only to our early development but also to our daily lives. CIRM was a partner in the development of the exhibit, but the primary mover behind it has been Canada’s Stem Cell Network, and within the network, Lisa Willemse who has really pushed its two-year gestation.

The earliest steps in the development involved visits to children in schools to tease out their points of interest. In a press release she explained some of what they learned:

“How does a lizard grow a new tail? Where does disease come from? How do we start little and get big? These were the kinds of questions the kids asked us, which shows a real interest in the mysteries of the body—mysteries that are largely the domain of stem cells.”

“Much of it is easy to explain, once they understand that stem cells have the ability to make all the kinds of cells in the body. For example, you can tell them that every second, stem cells in your bone marrow make about 2 million new red blood cells. You snap your fingers, and just like that, another 2 million cells were made. Soon they all start snapping their fingers, knowing that every time they do it, something remarkable and vital to life has happened in their own body.”

In Canada, the four modules have explanations in English and French. In California, they will be in English and Spanish. In Spanish the exhibit title “Super Cells: The Power of Stem Cells” becomes Celulas Fantasticas: El Poder Del Las Celulas Madre. I love the concept of a mother cell.

Additional partners in the project included the Centre for Commercialization of Regenerative Medicine in Canada and the UK’s Cell Therapy Catapult.

Don Gibbons

Cells’ Knack for Hoarding Proteins Inadvertently Kickstarts the Aging Process

Even cells need to take out the trash in order to maintain a healthy clean environment. And scientists are now uncovering the harmful effects when cells instead begin to hoard their garbage.

Cells' penchant for hoarding proteins may spur the cellular aging process, according to new research.

Cells’ penchant for hoarding proteins may spur the cellular aging process, according to new research. [Labyrinth (1986)]

Aging, on the cellular level is—at its core—the increasing inability for cells to repair themselves over time. As cells begin to break down faster than they can be repaired, the risk of age-related diseases escalates. Cancer, heart disease and neurological conditions such as Alzheimer’s disease are some of aging’s most deadly effects.

As a result, scientists have long searched for ways to give our cells a little help and improve our quality of life as we age. For example, recent research has pointed to a connection between fasting (restricting calories) and a longer lifespan, though the molecular mechanisms behind this connection remain somewhat cryptic.

But now Dr. Daniel Gottschling, a scientist at the Fred Hutchinson Cancer Research Center and an aging expert, has made extraordinary progress toward solving some of the mysteries of aging.

In two studies published this month in the Proceedings of the National Academy of Sciences and eLife, Gottschling and colleagues discover that a particular long-lasting protein builds up over time in certain cell types, causing the buildup of a protein hoard that damages the cell beyond repair.

Clearing out the Cobwebs

Some cells, such as those that make up the skin or that reside in the gut, are continually replenished by a stockpile of adult stem cells. But other cells, such as those found in the eye and brain, last for years, decades and—in some cases—our entire lifetimes.

Within and surrounding these long-lived cells are similarly long-lived proteins which help the cell perform essential functions. For example, the lens of the human eye, which helps focus light, is made up of these proteins that arise during embryonic development and last for a lifetime.

Dr. Daniel Gottschling is looking to unlock the mysteries behind cellular aging.

Dr. Daniel Gottschling is looking to unlock the mysteries behind cellular aging. [Image courtesy of the Fred Hutchinson Cancer Research Center]

“Shortly after you’re born, that’s it, you get no more of that protein and it lives with you the rest of your life,” explained Gottschling.

As a result, if those proteins degrade and die, new ones don’t replace them—the result is the age-related disease called cataracts.

But scientists weren’t exactly sure of the relationship between these dying proteins and the onset of conditions such as cataracts, and other disease related to aging. Did these conditions occur because the proteins were dying? Or rather because the proteins were building up to toxic levels?

So Gottschling and his team set up a series of experiments to find out.

Stashing Trash

They developed a laboratory model by using yeast cells. Interestingly, yeast cells share several key properties with human stem cells, and are often the focus of early-stage research into basic, fundamental concepts of biology.

Like stem cells, yeast cells grow and divide asymmetrically. In other words, a ‘mother’ cell will produce many ‘daughter’ cells, but will itself remain intact. In general, yeast mother cells produce up to 35 daughter cells before dying—which usually takes just a few days.

 Yeast “mother” cells budding and giving birth to newborn “daughter” cells.  [Image courtesy of Dr. Kiersten Henderson / Gottschling Lab]

Yeast “mother” cells budding and giving birth to newborn “daughter” cells.
[Image courtesy of Dr. Kiersten Henderson / Gottschling Lab]

Here, the research team used a special labeling technique that marked individual proteins that exist within and surrounding these mother cells. These microscopic tracking devices then told researchers how these proteins behaved over the entire lifespan of the mother cell as it aged.

The team found a total of 135 long-lived proteins within the mother cell. But what really surprised them was what they found upon closer examination: all but 21 of these 135 proteins appeared to have no function. They appeared to be trash.

“No one’s ever seen proteins like this before [in aging],” said Nathanial Thayer, a graduate student in the Gottschling Lab and lead author of one of the studies.

Added Gottschling, “With the number of different fragments [in the mother cell], we think they’re going to cause trouble. As the daughter yeast cells grow and split off, somehow mom retains all these protein bits.”

This startling discovery opened up an entirely new set of questions, explained Gottschling.

“It’s not clear whether the mother’s trash keeper function is a selfless act designed to give her daughters the best start possible, or if she’s hanging on to them for another reason.”

Hungry, Hoarding Mother Cells

So Gottschling and his team took a closer look at one of these proteins, known as Pma1.

Recent work by the Gottschling Lab found that cells lose their acidity over time, which itself leads to the deterioration of the cells’ primary energy source. The team hypothesized that Pma1 was somehow intricately tied to corresponding levels of pH (high pH levels indicate an acidic environment, while lower pH levels signify a more basic environment).

In the second study published in eLife, led by Postdoctoral Fellow Dr. Kiersten Henderson, the team made several intriguing discoveries about the role of Pma1.

First, they uncovered a key difference between mother and daughter cells: daughter cells are born with no Pma1. As a result, they are far more acidic than their mothers. But when they ramped up Pma1 in the mother cells, the acidity levels in subsequent generations of daughter cells changed accordingly.

“When we boosted levels of the protein, daughter cells were born with Pma1 and became more basic (they had a lower pH), just like their mothers.”

Further examination uncovered the true relationship between Pma1 and these cells. At its most fundamental, Pma1 helps the mother cells eat.

“Pma1 plays a key role in cellular feeding,” said Gottschling. “The protein sits on the surface of cells and helps them take in nutrients from their environment.”

Pma1 gives the mother cell the ability to gorge herself. The more access to food she has, the easier it is for her to produce more daughter cells. By hoarding Pma1, the mother cell can churn out more offspring. Unfortunately, she is also signing her own death certificate—she’s creating a more basic environment that, in the end, proves toxic and contributes to her death.

The hoarding, it turns out, may not all be due to the mother cells’ failure to ‘take out the trash.’ Instead, she wants to keep eating and producing daughters—and hoarding Pma1 allows her to do just that.

“There’s this whole trade off of being able to divide quickly and the negative side is that the individual, the mother, does not get to live as long.”

Together, the results from these two studies provide a huge boost for researchers like Gottschling who are trying to unravel the molecular mysteries of aging. But the process is incredibly intricate, and there will likely be no one simple solution to improving quality of life as we get older.

“The whole issue of aging is so complex that we’re still laying the groundwork of possibilities of how things can go awry,” said Gottschling. “And so we’re still learning what is going on. We’re defining the aging process.”

New Cellular Tracking Device Tests Ability of Cell-Based Therapies to Reach Intended Destination

Therapies aimed at replacing damaged cells with a fresh, healthy batch hold immense promise—but there remains one major sticking point: once you have injected new, healthy cells into the patient, how do you track them and how do you ensure they do the job for which they were designed?

New tracking technique could improve researchers' ability to test potential cell therapies.

New tracking technique could improve researchers’ ability to test potential cell therapies.

Unfortunately, there’s no easy solution. The problem of tracking the movement of cells during cell therapy is that it’s hard to stay on their trail they enter the body. They can get mixed up with other, native cells, and in order to test whether the therapy is working, doctors often have to rely on taking tissue samples.

But now, scientists at the University of California, San Diego School of Medicine and the University of Pittsburgh have devised an ingenious way to keep tabs on where cells go post injection. Their findings, reported last week in the journal Magnetic Resonance in Medicine, stand to help researchers identify whether cells are arriving at the correct destination.

The research team, lead by UCSD Radiology Professor Dr. Eric Ahrens, developed something called a periflourocarbon (PFC) tracer in conjunction with MRI technology. Testing this new technology in patients receiving immune cell therapy for colorectal cancer, the team found that they were better able to track the movement of the cells than with traditional methods.

“This is the first human PFC cell tracking agent, which is a new way to do MRI cell tracking,” said Ahrens in a news release. “It’s the first example of a clinical MRI agent designed specifically for cell tracking.”

They tagged these cells with atoms of fluorine, a compound that normally occurs at extremely low levels. After tagging the immune cells, the researchers could then see where they went after being injected. Importantly, the team found that more than one-half of the implanted cells left the injection site and headed towards the colon. This finding marks the first time this process had been so readily visible.

Ahrens explained the technology’s potential implications:

“The imaging agent technology has been shown to be able to tag any cell type that is of interest. It is a platform imaging technology for a wide range of diseases and applications.”

A non-invasive cell tracking solution could serve as not only as an attractive alternative to the current method of tissue sampling, it could even help fast-track through regulatory hurdles new stem cell-based therapies. According to Ahrens:

“For example, new stem cell therapies can be slow to obtain regulatory approvals in part because it is difficult, if not impossible, with current approaches to verify survival and location of transplanted cells…. Tools that allow the investigator to gain a ‘richer’ data set from individual patients mean it may be possible to reduce patient numbers enrolled in a trial, thus reducing total trial cost.”

What are the ways scientists see stem cells in the body? Check out our Spotlight Video on Magnetic Particle Imaging.

New Videos: Living with Crohn’s Disease and Working Towards a Stem Cell Therapy

Note: the two videos below are also available on our website

She doesn’t want your sympathy. She doesn’t want your admiration. She just wants your understanding.

Rachel Bonner, a sixteen-year-old high school student and founder of the Hope for Crohn’s charity, spoke to the CIRM governing Board on September 10th about what it’s like living with Crohn’s disease. In the eight years since her diagnosis, Rachel has come a long way in talking publicly about her condition:

“I never thought I’d stand up here and admit to wearing a diaper while being in middle school. But Crohn’s turns from a secret struggle to something I want to share with other people. And ultimately have others understand the life of a Crohn’s patient just a bit more. “

Crohn’s disease is a type of inflammatory bowel disease (IBD) in which the intestines are chronically inflamed. Symptoms of Crohn’s include a frequent need to pass bowel movements, constant diarrhea, rectal bleeding, fatigue and loss of appetite.

In a healthy individual, the friendly bacteria living in the gut are ignored by the immune system. But in the case of IBD, the immune cells attack these bacteria as foreign invaders, causing an inflammatory response. The sustained inflammation eventually damages the gut wall causing the symptoms of IBD.

Current therapies for IBD focus solely on treating the inflammation. Dr. Ophir Klein, a CIRM grantee and UCSF researcher, also spoke to the governing Board and described another treatment avenue:

“There’s another component that’s been under-explored and potentially has a lot of impact therapeutically which is the regenerative aspects of the condition because after the inflammation occurs in the gut, the gut needs to heal, and that healing comes from stem cells. “

In his presentation to the Board, Dr. Klein detailed his lab’s work to understand how stem cells regulate the healing of the intestine and to eventually find cures for IBD.

Although Rachel and her doctors have found a treatment sweet spot, which has kept her Crohn’s at bay, she still holds out hope that a cure, perhaps from a stem-cell based therapy, is not too far away:

“Everyday I go to sleep hoping that this treatment sweet spot will work until they find a cure”

Stem cell stories that caught our eye: a good review at the NY Times, expanding cord blood and leukemia

Here are some stem cell stories that caught our eye this past week. Some are groundbreaking science, others are of personal interest to us, and still others are just fun.

Review paints picture of the field today.
A writer I have respected for many years, Karen Weintraub, wrote a nice review of the current state of stem cell clinical trials in the Tuesday Science Times in the New York Times. She discusses the steady, methodical progress being made:

“Researchers have been slowly learning how to best use stem cells, what types to use and how to deliver them to the body — findings that are not singularly transformational, but progressive and pragmatic.”

She quotes our senior VP Ellen Feigal about the safety seen so far in clinical trials and notes that CIRM should have 10 clinical trials enrolling patients by the end of the year. She also covers the dangers of clinics offering unproven therapies and the power of using iPS-type stem cells to model diseases in the laboratory. Overall, a nicely balanced piece.

Making mitochondrial disease and 3-parent embryos personal. A little newspaper in Oregon called the Willamet Week has published a story that makes the issues around so-called “three-parent” babies very personal. The controversial procedure aims to allow women with rare mitochondrial diseases to have normal children.

Mitochondria, known as the powerhouses of the cell, have the unusual trait of being the only part of the cell besides the nucleus to have any DNA. It is these few genes in the mitochondria that we inherit solely from our mothers because when the DNA from the egg and sperm fuse, the mother’s mitochondria stay in the fluid outside the nucleus. So, to avoid passing along faulty mitochondrial genes, a team in Oregon devised a way to insert the DNA from the mother’s nucleus into a donor egg that had its nucleus removed, a process called nuclear transfer.

Guided by a microscope researchers insert the nucleus from one woman into the egg of another

Guided by a microscope researchers insert the nucleus from one woman into the egg of another

The paper provides a long read—nearly 4,000 words—that goes into great detail about the procedure, the ethics, the research team’s views on the ethics, and the personal story of a patient living with a disease of exhaustion she calls “mitochondrial crash.” The writer lets the patient have the last word on ethics:

“To me it’s win-win because you’re not messing with God’s child. You’re just taking out the bad parts. I don’t want to pick out a blond-haired, blue-eyed tall kid, picking your child’s traits, but to rule out a potentially lethal chronic illness brings in a whole different story.”


Cord blood might now save more adult cancer patients.
Umbilical cord blood is a literal lifesaver for many pediatric cancer patients allowing them to withstand harsh chemotherapy and be rescued by the stem cells in the cord blood. But the procedure is used in few adults because the vast majority of cord blood samples don’t have enough stem cell for an adult requiring the use of two cord samples and doubling the chance for potentially deadly immune reactions.

A team at the University of Montreal screened more than 5,000 molecules looking for one that would let them expand the number of stem cells from one sample in the lab. They hit upon one that they say could allow a 10-fold increase in the number of single cord samples suitable for adults. They expect to begin clinical trials in December.

Science News ran a brief review of the work and the blog Science 2.0 ran the university’s press release with a bit more detail.

Trial begins with cancer drug named for CIRM
Researchers at the University of California, San Diego, announced this week that they had begun a clinical trial with leukemia patients using a drug named for our agency cirmtuzumab. This molecule, in the class of drugs called antibodies, disables a protein that cancer stem cells use to accelerate the growth of cancer.

This trial, for patients with recurrence of their chronic lymphocytic leukemia, became the third CIRM funded team this month announcing plans to start clinical trials. In addition to our blog post the San Diego Union Tribune wrote about the latest trial, and we issued press releases on the trials for spinal cord injury and diabetes.

Don Gibbons

New formula a more efficient way to reprogram adult cells to become like embryonic stem cells

Shinya Yamanaka won the Nobel Prize for developing a recipe of genetic factors that can turn back the clock of adult cells and make them behave like embryonic stem cells. But he would be the first to tell you his recipe ultimately may not be the best one for making these stem cells called iPS cells.

Virtually from the day he published his groundbreaking work, teams around the world have tried to develop new formulas that get around some problems with the original. One issue is the low efficiency of getting true stem cells. Another is the high rate of genetic aberrations that can be produced in the resulting stem cells.

Now, a team pairing researchers at the Hebrew University in Jerusalem and the Whitehead Institute in Cambridge, Massachusetts, has published a new recipe that seems to yield many more true stem cells, ones that are called pluripotent because they can make all cell types. The new cells also seem to have fewer genetic alterations, which could make them safer for clinical use in people.

They made the improved cells by moving from OSKM to SNEL—from the original genetic factors, Oct4, Sox2, Klf4 and Myc, to Sall4, Nanog, Esrrb and Lin28. An elaborate computer analysis of the function of genes helped them come up with the formula.

This work used mouse cells, so up next on their agenda is coming up with a similar formula that works in human cells. HealthCanal ran the university’s press release and Genetic Engineering & Biotechnology News ran a slightly more technical analysis of the work.

Don Gibbons

Harder, Better, Faster, Stronger: Scientists Work to Create Improved Immune System One Cell at a Time

The human immune system is the body’s best defense against invaders. But even our hardy immune systems can sometimes be outpaced by particularly dangerous bacteria, viruses or other pathogens, or even by cancer.

Salk Institute scientists have developed a new cellular reprogramming technique that could one day boost a weakened immune system.

Salk Institute scientists have developed a new cellular reprogramming technique that could one day boost a weakened immune system.

But what if we could give our immune system a boost when it needs it most? Last week scientists at the Salk Institute for Biological Sciences devised a new method of doing just that.

Reporting in the latest issue of the journal Stem Cells, Dr. Juan Carlos Izpisua Belmonte and his team announce a new method of creating—and then transplanting—white blood cells into laboratory mice. This new and improved method could have significant ramifications for how doctors attack the most relentless disease.

The authors achieved this transformation through the reprogramming of skin cells into white blood cells. This process builds on induced pluripotent stem cell, or iPS cell, technology, in which the introduction of a set of genes can effectively turn one cell type into another.

This Nobel prize-winning approach, while revolutionary, is still a many months’ long process. In this study, the Salk team found a way to shorten the cellular ‘reprogramming’ process from several months to just a few weeks.

“The process is quick and safe in mice,” said Izpisua Belmonte in a news release. “It circumvents long-standing obstacles that have plagued the reprogramming of human cells for therapeutic and regenerative purposes.”

Traditional reprogramming methods change one cell type, such as a skin cell, into a different cell type by first taking them back into a stem cell-like, or ‘pluripotent’ state. But here, the research team didn’t take the cells all the way back to pluripotency. Instead, they simply wiped the cell’s memory—and gave it a new one. As first author Dr. Ignacio Sancho-Martinez explained:

“We tell skin cells to forget what they are and become what we tell them to be—in this case, white blood cells. Only two biological molecules are needed to induce such cellular memory loss and to direct a new cell fate.”

This technique, which they dubbed ‘indirect lineage conversion,’ uses the molecule SOX2 to wipe the skin cell’s memory. They then use another molecule called miRNA 125b to reprogram the cell into a white blood cell.

These newly generated cells appear to engraft far better than cells derived from traditional iPS cell technology, opening the door to therapies that more effectively introduce these immune cells into the human body. As Sanchi-Martinez so eloquently stated:

“It is fair to say that the promise of stem cell transplantation is now closer to realization.”

Stories of Hope: Stroke

Six months after surviving a stroke, Sonia Olea wanted to die. Her right leg was weak, her right arm useless. She had trouble speaking and even small tasks were challenging. Just making a phone call was virtually impossible. One morning, she woke up with her arm pinned in an awkward, painful position. After finally repositioning it, she wanted to call her fiancé, but knew she couldn’t get the words out. That’s when it hit her.

Sonia has seen first hand how a stroke can rob you of even your most basic abilities.

Sonia has seen first hand how a stroke can rob you of even your most basic abilities.

“I thought, I’m only 32,” says Sonia. “How could this be happening to me?”

Nobody really had an answer. A stroke occurs when a blood clot blocks a vessel in the brain and cuts off blood flow. Brain cells begin to die within minutes when they are deprived of oxygen and nutrients. Stroke rates are on the rise for young adults for a variety of reasons but no one could pinpoint specifically what caused hers.

Slowly, Sonia fought back from her depression and realized she could do this. She would find a way to recover. Just one year later, she got a call from Stanford University; asking if she would be willing to participate in a cutting-edge, stem cell-based clinical trial.

Was she ever. The answer, says Sonia, was a no-brainer.

Rescuing Brain Cells
Led by CIRM grantee Gary Steinberg, M.D., Ph.D., chairman of the Department of Neurosurgery at Stanford School of Medicine, the early phase clinical trial tested the safety of transplanting bone marrow stem cells into the brain. It was a revolutionary approach.

“The old notion was that you couldn’t recover from a stroke after around three months,” says Steinberg. “At that point, the circuits were completely dead—and you couldn’t revive them.”

While this was partially true, it was thought that brain cells, or neurons, just outside the stroke damage might be saved. Steinberg and collaborators at the University of Pittsburgh recognized that stem cells taken from bone marrow wouldn’t transform into functioning neurons. However, the transplanted cells could release molecules that might rescue neurons that were impaired, but not yet dead.

Brain Surgery
Sonia had surgery to transplant bone marrow stem cells into her brain in late May 2013. The improvement was almost instantaneous. “When I woke up, my speech was strong, I could lift up my feet and keep them in the air, I even raised my right hand,” says Sonia. Though the trial was primarily designed to study the stem cell therapy’s safety, researchers were also interested in its effectiveness.

“Sonia was one of our two remarkable patients who got better the day after surgery and continued to improve throughout the year,” says Steinberg. 18 patients in total were treated in that study.

Although Sonia’s treatment results are still very preliminary, they bode well for a separate CIRM-funded stroke research project also led by Steinberg. In this study, cells grown from embryonic stem cells will be turned into early-stage neuron, or brain, cells and then transplanted into the area of stroke damage. The team has found that transplanting these neural cells into mice or rats after a stroke helps the animals regain strength in their limbs. The team is busy working out the best conditions for growing these neural cells in order to take them into clinical trials.

In the meantime, Sonia continues to improve. “My leg is about 95 percent better and my arm is around 60 percent there,” says Sonia. “My speech isn’t perfect, but I can talk and that’s something I never could have done before the surgery.”

The added function has made a huge difference in her quality of life. She can walk, run, drive a car, call a restaurant to make a dinner reservation—simple things she took for granted before having a stroke. But most importantly, she has confidence in the future.

“Everything is good,” says Sonia, “and it’s only going to get better.”

To learn about CIRM-funded stroke research, visit our Stroke Fact Sheet. Read more about Sonia’s Story of Hope on our website.