Category Archives: Embryonic Stem Cells

Embryonic Stem Cells: 5 Misconceptions – livescience.com

Fertility treatments could be a factor that will result in declining fecundity (potential for fertility, such as regular menstrual cycles) across the generations, some researchers say. Others point to the depression that can come with infertility as a reason to offer medicalized pregnanies. Image

Last week President Obama lifted restrictions on federal funding for embryonic stem cell research and asked the National Institutes of Health to come up with a funding game plan within 120 days. Yet while the field of stem cell research holds great promise, hype and misconceptions cloud the picture. Here are a five such misconceptions.

1. George W. Bush killed research on embryonic stem cells.

Wrong. Bush actually was the first president to allow federal funding. Bill Clinton had chickened out. A very brief history follows.

In 1974, Congress banned federal funding on fetal tissue research and established the Ethics Advisory Board to study the nascent field of in vitro fertilization. In 1980 Ronald Reagan killed the Board, which was friendly to embryonic research, resulting in a de facto moratorium on funding. Congress tried to override the moratorium in 1992, but George H.W. Bush vetoed it. Bill Clinton lifted the moratorium in 1993 but reversed his decision in 1994 after public outcry. In 1995, Congress passed the Dickey-Wicker Amendment, banning federal funding on any research that destroys human embryos.

In 2001 Bush enabled limited funding on embryonic stem cell lines already derived from discarded embryos; the life or death decision already had been made, he said. He thought more than 60 lines existed, but within months scientists realized that only about 20 were viable, not enough to do substantial research.

2. Bush spurred development of alternative sources of embryonic stem cells.

Sure, in the same way his disastrous invasion of Afghanistan and Iraq spurred the development of treatment for massive head trauma, or the way his economic policies have encouraged all of us to do more with less. One doesn't advance a scientific field by handicapping researchers.

Regardless, the biggest advance in recent years has come from Japan by a researcher not affected by U.S. research funding rules. U.S. federal funding could have led to even more advances of alternative sources, because funding stem cell research in general can have a synergetic effect across the various research specialties.

3. Embryonic stem cells are no longer needed.

Wrong. In 2007, Shinya Yamanaka of Kyoto University in Japan announced a breakthrough in which adult skin cells could be coaxed back into an embryonic state and thus regain the ability to branch into any kind of human cell, such as heart, pancreas or spinal cord nerve cell. While a major advance, the work itself is in an embryonic state, years from practical application.

The work on these so-called induced pluripotent stem (iPS) cells complements embryonic stem cell research; it doesn't replace it. The iPS cells have a greater tendency to become cancerous. Work on "real" embryonic stem cells is needed, at a minimum, to understand what iPS cells lack. Many view Yamanaka's technique as brilliant yet worry that his four-gene manipulation of adult cells might be too simplistic.

Research on iPS cells is particularly exciting because it opens the possibility of using one's own cells say, from skin to produce pancreas cells to cure diabetes, whereas embryonic stem cells would introduce DNA from a stranger.

4. Cures are around the corner.

Wrong. Stem cell research is dominated by hype. Remember gene therapy, the insertion of genes into human cells to cure all types of diseases? Nearly two decades after the first gene therapy procedure, the technique remains highly experimental and problematic. Stem cell research faces a similar future.

5. Obama's executive order means "all systems go."

Unlikely. The new rule eliminates red tape, for now researchers can study any established embryonic stem cell line. Previously, stem cell researchers receiving private and public funding needed to keep detailed records of spending, down to which microscope is used for which kind of stem cell. That's history.

But the Dickey-Wicker Amendment (see No. 1 above) is the law of the land, meaning federally funded researchers cannot create new embryonic stem cells lines. They can work only on those new lines created with private funding, which aren't that plentiful. Also, some scientists worry that crucial private funding will dry up with the poor economy and false reassurances that federal funding is in place.

The furor over stem cells focuses on the definition of human life, which many believe begins when sperm meets eggs. Yet inevitably lines will be blurred in coming years when babies are born with the DNA of two sperms or ova transplanted into an egg. Just as humans evolved from non-humans with no precise generation in which a non-human gave birth to a human we may come to understand that all of nature is a continuum.

Christopher Wanjek is the author of the books "Bad Medicine" and "Food At Work." His column, Bad Medicine, appears each Tuesday on LiveScience.

The rest is here:
Embryonic Stem Cells: 5 Misconceptions - livescience.com

Lab grown human colons change study of GI disease – Medical Xpress

June 22, 2017 This confocal microscopic image shows a human colon organoid generated in the laboratory with human pluripotent stem cells. The organoid is shown after it was transplanted into a mouse. The engineered colon secreted proteins found in natural human colon. Researchers report study results in Cell Stem Cell. Credit: Cincinnati Children's

Scientists used human pluripotent stem cells to generate human embryonic colons in a laboratory that function much like natural human tissues when transplanted into mice, according to research published June 22 in Cell Stem Cell.

The study is believed to be the first time human colon organoids have been successfully tissue engineered in this manner, according to researchers at Cincinnati Children's Hospital Medical Center who led the project.

The technology allows diseases of the colon to be studied in unprecedented detail in a human modeling system. It also comes with the potential to one day generate human gastrointestinal (GI) tract tissues for transplant into patients, according to James Wells, PhD, senior study investigator and director of the Cincinnati Children's Pluripotent Stem Cell Center.

"Diseases affecting this region of the GI tract are quite prevalent and include ailments like colitis, colon cancer, Irritable Bowel Syndrome, Hirschsprung's disease and polyposis syndromes," Wells said. "We've been limited in how we can study these diseases, including the fact that animal models like mice don't precisely recreate human disease processes in the gastrointestinal tract. This system allows us to very effectively model human diseases and human development."

Building the GI Tract

In a series of studies published since 2009, researchers in Wells' laboratory used human pluripotent stem cells (hPSCs) to grow embryonic-stage small intestines with a functioning nervous system, and the antrum and fundus regions of the human stomach.

The researchers - including Jorge Munera, PhD, first author and postdoctoral fellow in the Wells laboratory - note in their current paper the colon has been more difficult to generate than other parts of the GI tract.

Part of the challenge to identifying the correct genetic and molecular programming to coax hPSCs in to colonic organoids has been a lack of data about embryonic development of the organ, according to the authors. They addressed this by conducting a series of molecular and genetic screens of developing hindgut tissues in animal models. The hindgut is the portion of the developing gut that gives rise to the entire large intestine - which includes the cecum, colon and rectum.

They also mined public databases (GNCPro, TiGER, Human Protein Atlas) to identify molecular markers of the hindgut in the adult colon.

Frogs and Mice at Forefront

To develop a model for generating the human colon, scientists first identified SATB2 (special AT-rich sequence-binding protein 2) as a definitive molecular marker for hindgut in frogs, mice and in humans.

SATB2 is a DNA-binding protein that facilitates structural organization of chromosomes in the nucleus of cells.

The protein sequence of SATB2 is remarkably similar between frogs, mice and humans. This led the authors to the hypothesis that molecular signals regulating SATB2 in frogs and mice could be used to make human colon organoids that express the protein.

The authors also noticed that signaling from the growth factor BMP (bone morphogenetic protein) was highly active in the SATB2-expressing region of the gut tube. The researchers learned during their analysis of frog, mouse and human stem-cell derived intestine that signaling by BMP is needed to establish SATB2 in the developing hindgut. With SABT2 as a marker, the researchers show BMP signaling is required for development of tissues specific to the posterior gut region of frogs and mice where the colon develops.

When BMP protein was added for three days in human pluripotent stem cell-derived gut tube cultures, it induced a posterior HOX code. HOX includes a critical set of genes that help control the embryo's development plan from head to toe. Researchers report the posterior HOX helps control the formation of SATB2-expressing human colon organoids.

Testing Translational Potential

To see how human GI tissues perform in a living organism - and to test their future therapeutic potential - the research team included collaborators from the Division of Surgery, led by Michael Helmrath, MD, a pediatric surgeon and director of the Surgical Research program.

The tissue-engineered colonic organoids were transplanted into the kidney capsules of immunocompromised mice for six to 10 weeks. During observation and analysis of the now in vivo organoids, study authors looked for signs of posterior region enteroendocrine cells, which make hormones found in naturally developed human colon.

Researchers report that following transplant, the human colonic organoids assumed the form, different structures and molecular and cell properties of the human colon.

Munera, study first author, pointed to a number of new ways that human colon organoids could be used study disease.

"By exposing human colonic organoids to inflammatory triggers, we can now learn how the cell lining of the colon and the supporting cells beneath cooperate to respond to inflammation," Munera said. "This could be very relevant for patients with Crohn's disease or ulcerative colitis. And because the microbiome, the organisms that live in our guts, are most concentrated in the colon, the organoids potentially could be used to model the human microbiome in health and disease."

Like other parts of the GI tract grown by the researchers, the human colon organoids also create a potential new platform for testing new drugs before the start of clinical trials. Most oral drugs are absorbed by the body through the gut.

Explore further: Human tissue model developed to test colon cancer drugs

More information: Cell Stem Cell (2017). DOI: 10.1016/j.stem.2017.05.020

New research overturns long-held views on a basic messaging system within living cells. Key cellular communication machinery is more regionally constrained within the cell than previously thought. The findings suggest new ...

The leading cause of acute gastroenteritis linked to eating raw seafood disarms a key host defense system in a novel way: It paralyzes a cell's skeleton, or cytoskeleton.

It's a tiny marine invertebrate, no more than 3 millimeters in size. But closely related to humans, Botryllus schlosseri might hold the key to new treatments for cancer and a host of vascular diseases.

Scientists used human pluripotent stem cells to generate human embryonic colons in a laboratory that function much like natural human tissues when transplanted into mice, according to research published June 22 in Cell Stem ...

Paracetamol is popular for relieving pain. But if you are pregnant, you should think twice before popping these pills according to the researchers in a new study. In an animal model, Paracetamol, which is the pain-relieving ...

Fathers-to-be, take note: You may be more useful in the labor and delivery room than you realize.

Please sign in to add a comment. Registration is free, and takes less than a minute. Read more

See original here:
Lab grown human colons change study of GI disease - Medical Xpress

Congressmen seek pro-life focus at NIH – OneNewsNow

A move is afoot in Congress to change the direction of the National Institutes of Health to more valuable and humane research projects.

Under the direction of Dr. Francis Collins, NIH has directed millions of dollars toward research on human embryos.

David Prentice of the Charlotte Lozier Institute tells OneNewsNow that Congressmen Jim Banks and Dan Lipinski have introduced the Patients First Act to change the agency's course.

The whole point of the bill," says Prentice, "is to prioritize NIH funding for adult stem cells that are going to be able to help patients, first, in the near term, be able to get them into the clinic and help these people. And that, again, is adult stem cells, not embryonic stem cells."

Dr. Collins is famous for leading the decade-long Human Genome Project, which mapped DNA sequences.

Once an atheist, Collins is also known for being an outspoken Christian in the scientific community, where faith is often mocked and dismissed. Yet the embryonic research at NIH has caused disappointment among pro-life activists, and some have called for his firing at NIH.

The proposed legislation could mean hundreds of millions of dollars would be directed toward adult stem cells, which are already being used to treat medical conditions for an estimated 1.5 million patients around the world.

Shifting research to stem cells is going to help patients, not just mean playing in the laboratory, which is what happens with embryonic stem cells as well as fetal tissue and fetal stem cells, Prentice adds. Embryonic and fetal stem cells have been failures.

Prentice says the latter forms of research are unethical, so instead the money could be spent to accelerate research on something of proven value.

Charlotte Lozier Institute would also like to see bills dealing with other ethical concerns at NIH including animal/human hybrids and research using aborted baby tissue.

We moderate all reader comments, usually within 24 hours of posting (longer on weekends). Please limit your comment to 300 words or less and ensure it addresses the article - NOT another reader's comments. Comments that contain a link (URL), an inordinate number of words in ALL CAPS, rude remarks directed at other readers, or profanity/vulgarity will not be approved.

Go here to see the original:
Congressmen seek pro-life focus at NIH - OneNewsNow

There are millions of protein factories in every cell. Surprise, they’re not all the same – Science Magazine

Ribosomes, which build a protein (black) from an RNA strand (blue), may specialize in making particular sets of proteins.

V. ALTOUNIAN/SCIENCE

By Mitch LeslieJun. 21, 2017 , 11:00 AM

The plant that built your computer isn't churning out cars and toys as well. But many researchers think cells' crucial protein factories, organelles known as ribosomes, are interchangeable, each one able to make any of the body's proteins. Now, a provocative study suggests that some ribosomes, like modern factories, specialize to manufacture only certain products. Such tailored ribosomes could provide a cell with another way to control which proteins it generates. They could also help explain the puzzling symptoms of certain diseases, which might arise when particular ribosomes are defective.

Biologists have long debated whether ribosomes specialize, and some remain unconvinced by the new work. But other researchers say they are sold on the finding, which relied on sophisticated analytical techniques. "This is really an important step in redefining how we think about this central player in molecular biology," says Jonathan Dinman, a molecular biologist at the University of Maryland in College Park.

A mammalian cell may harbor as many as 10 million ribosomes, and it can devote up to 60% of its energy to constructing them from RNA and 80 different types of proteins. Although ribosomes are costly, they are essential for translating the genetic code, carried in messenger RNA (mRNA) molecules, into all the proteins the cell needs. "Life evolved around the ribosome," Dinman says.

The standard view has been that a ribosome doesn't play favorites with mRNAsand therefore can synthesize every protein variety. But for decades, some researchers have reported hints of customized ribosomes. For example, molecular and developmental biologist Maria Barna of Stanford University in Palo Alto, California, and colleagues reported in 2011 that mice with too little of one ribosome protein have short tails, sprout extra ribs, and display other anatomical defects. That pattern of abnormalities suggested that the protein shortage had crippled ribosomes specialized for manufacturing proteins key to embryonic development.

Definitive evidence for such differences has been elusive, however. "It's been a really hard field to make progress in," says structural and systems biologist Jamie Cate of the University of California (UC), Berkeley. For one thing, he says, measuring the concentrations of proteins in naturally occurring ribosomes has been difficult.

In their latest study, published online last week in Molecular Cell, Barna and her team determined the abundances of various ribosome proteins with a method known as selected reaction monitoring, which depends on a type of mass spectrometry, a technique for sorting molecules by their weight. When the researchers analyzed 15 ribosomal proteins in mouse embryonic stem cells, they found that nine of the proteins were equally common in all ribosomes. However, four were absent from 30% to 40% of the organelles, suggesting that those ribosomes were distinctive. Among 76 ribosome proteins the scientists measured with another mass spectrometry-based method, seven varied enough to indicate ribosome specialization.

Barna and colleagues then asked whether they could identify the proteins that the seemingly distinctive ribosomes made. A technique called ribosome profiling enabled them to pinpoint which mRNAs the organelles were readingand thus determine their end products. The specialized ribosomes often concentrated on proteins that worked together to perform particular tasks. One type of ribosome built several proteins that control growth, for example. A second type churned out all the proteins that allow cells to use vitamin B12, an essential molecule for metabolism. That each ribosome focused on proteins crucial for a certain function took the team by surprise, Barna says. "I don't think any of us would have expected this."

Ribosome specialization could explain the symptoms of several rare diseases, known as ribosomopathies, in which the organelles are defective. In Diamond-Blackfan anemia, for instance, the bone marrow that generates new blood cells is faulty, but patients also often have birth defects such as a small head and misshapen or missing thumbs. These seemingly unconnected abnormalities might have a single cause, the researchers suggest, if the cells that spawn these different parts of the body during embryonic development carry the same specialized ribosomes.

Normal cells might be able to dial protein production up or down by adjusting the numbers of these specialized factories, providing "a new layer of control of gene expression," Barna says. Why cells need another mechanism for controlling gene activity isn't clear, says Cate, but it could help keep cells stable if their environment changes.

He and Dinman say the use of "state-of-the-art tools" makes the results from Barna's team compelling. However, molecular biologist Harry Noller of UC Santa Cruz doubts that cells would evolve to reshuffle the array of proteins in the organelles. "The ribosome is very expensive to synthesize for the cell," he says. If cells are going to tailor their ribosomes, "the cheaper way to do it" would entail modifying a universal ribosome structure rather than building custom ones.

See the original post:
There are millions of protein factories in every cell. Surprise, they're not all the same - Science Magazine

3D-Printed Human Embryonic Stem Cells Created for First Time

Researchers have developed a 3D printer that prints human embryonic stem cells.

Imagine if you could take living cells, load them into a printer, and squirt out a 3D tissue that could develop into a kidney or a heart. Scientists are one step closer to that reality, now that they have developed the first printer for embryonic human stem cells.

In a new study, researchers from Heriot-Watt University in Edinburgh have created a cell printer that spits out living embryonic stem cells. The printer was capable of printing uniform-size droplets of cells gently enough to keep the cells alive and maintain their ability to develop into different cell types. The new printing method could be used to make 3D human tissues for testing new drugs, grow organs, or ultimately print cells directly inside the body.

Human embryonic stem cells (hESCs) are obtained from human embryos and can develop into any cell type in an adult person, from brain tissue to muscle to bone. This attribute makes them ideal for use in regenerative medicine repairing, replacing and regenerating damaged cells, tissues or organs. [Stem Cells: 5 Fascinating Findings]

In a lab dish, hESCs can be placed in a solution that contains the biological cues that tell the cells to develop into specific tissue types, a process called differentiation. The process starts with the cells forming what are called "embryoid bodies." Cell printers offer a means of producing embryoid bodies of a defined size and shape.

The new 3D cell printer used compressed air to squirt out "bio-inks" containing cells and nutrient-rich fluid.

In the new study, the cell printer was made from a modified CNC machine (a computer-controlled machining tool) outfitted with two "bio-ink" dispensers: one containing stem cells in a nutrient-rich soup called cell medium and another containing just the medium. These embryonic stem cells were dispensed through computer-operated valves, while a microscope mounted to the printer provided a close-up view of what was being printed.

The two inks were dispensed in layers, one on top of the other to create cell droplets of varying concentration. The smallest droplets were only two nanoliters, containing roughly five cells.

The cells were printed onto a dish containing many small wells. The dish was then flipped over so the droplets now hung from them, allowing the stem cells to form clumps inside each well. (The printer lays down the cells in precisely sized droplets and in a certain pattern that is optimal for differentiation.)

Tests revealed that more than 95 percent of the cells were still alive 24 hours after being printed, suggesting they had not been killed by the printing process. More than 89 percent of the cells were still alive three days later, and also tested positive for a marker of their pluripotency their potential to develop into different cell types.

Biomedical engineer Utkan Demirci, of Harvard University Medical School and Brigham and Women's Hospital, has done pioneering work in printing cells, and thinks the new study is taking it in an exciting direction. "This technology could be really good for high-throughput drug testing," Demirci told LiveScience. One can build mini-tissues from the bottom up, using a repeatable, reliable method, he said. Building whole organs is the long-term goal, Demirci said, though he cautioned that it "may be quite far from where we are today."

Others have created printers for other types of cells. Demirci and colleagues made one that printed embryonic stem cells from mice. Others have printed a kind of human stem cells from connective tissues, which aren't able to develop into as many cell types as embryonic stem cells. The current study is the first to print embryonic stem cells from humans, researchers report in the Feb. 5 issue of the journal Biofabrication.

Editor's Note:This article was corrected at 2:45 p.m. Eastern Time, April 3 to correct the researchers' affiliation.

Follow LiveScience on Twitter @livescience. We're also on Facebook& Google+.

Original post:
3D-Printed Human Embryonic Stem Cells Created for First Time

Hyderabad team grows miniature eyes using stem cells – The Hindu


The Hindu
Hyderabad team grows miniature eyes using stem cells
The Hindu
The iPS cells are produced by genetically manipulating human skin cells to produce embryonic-like stem cells that are capable of forming any cell types of the body. Small portions of the corneal tissue were separated from the miniature eyes and used ...

Read this article:
Hyderabad team grows miniature eyes using stem cells - The Hindu

China plans embryonic stem cell trials for Parkinson’s and blindness – BioNews

Two teams of doctors in China are to administer embryonic stem cell therapy from fertilised human embryos to treat different degenerative diseases.

One trial testing ESC therapy in Parkinson's diseasewill be the first clinical trial of human embryonic stem cells (ESCs) in China, as well as the first trial in the world to examine ESCs for the treatment of Parkinson's, according to a report in Nature News. Both studies will be led by stem cell specialist Dr Qi Zhou at the First Affiliated Hospital of Zhengzhou University.

The only previous study of this type started in Australia last year:patients with the degenerative disease are being treated with cells derived from parthenogenetic embryos. These stem cells were harvested from unfertilised eggs induced into embryonic development, sidestepping many of the ethical issues with using viable embryos. ESCs are pluripotent stem cells taken from the inner cell mass of human embryos and have the potential to develop into any of the 200-plus specialised cell types in the adult body.

In Parkinson's disease, a specialised type of brain cell that produces the neurotransmitter dopamine are lost. Dr Zhou's team will inject four million immature cells derived from ESCs into the striatum area of patients' brains with the aim of reducing symptoms. This follows an unpublishedfour-year study led by Dr Zhou which showed promising results in 15 monkeys.

A second team will treat patients with age-related macular degeneration (AMD), in which vision is compromised by the loss of pigmented retinal epithelia in the eye. Building on pre-clinical trials carried out in South Korea and the US, ESC-derived retinal epithelial cells will be injected into the retinas of AMD patients in a bid to stop the disease progressing.

Using ESCs intreatment is controversialon ethical grounds, as well as fears that they could cause tumours. Some scientists are concerned that theParkinson's patients are being injected with cells thatmay not become the desired type of neurone. 'Not knowing what the cells will become is troubling.' said Dr Jeanne Loring at the Scripps Research Institute, California.However, pre-clinical work for the Australian trial found that 97 percentof the ESC-derived precursor cells developed into dopaminergic neurons.

In 2015, China announced new regulations for stem cell therapies aiming both to enable legitimate human trials, and curb administration of unapproved treatments. Zhou's clinical trials will be fully compliant, using government-certified ESC lines and the clinicians have been granted approval by a central government committee for their use.

Committee member Dr Pei Xuetao, a stem-cell scientist at the Beijing Institute of Transfusion Medicine, called the study a'major new direction for China'. Other groups of scientists are already planning trials for the testing of ESC therapy for other targeted treatments, such as encouraging growth and repair following spinal cord injuries.

Read more from the original source:
China plans embryonic stem cell trials for Parkinson's and blindness - BioNews

Parkinson’s target of China’s first clinical trial using embryonic stem … – Genetic Literacy Project

In the next few months, surgeons in the Chinese city of Zhengzhou will carefully drill through the skulls of people with Parkinsons disease and inject 4 million immature neurons derived from human embryonic stem cells into their brains.

This will mark the start of the first clinical trial in China using human embryonic stem (ES) cells, and the first one worldwide aimed at treating Parkinsons disease using ES cells from fertilized embryos. In a second trial starting around the same time, a different team in Zhengzhou will use ES cells to target vision loss caused by age-related macular degeneration.

It will be a major new direction for China, says Pei Xuetao, a stem-cell scientist at the Beijing Institute of Transfusion Medicine who is on the central-government committee that approved the trials. Other researchers who work on Parkinsons disease, however, worry that the trials might be misguided.

Jeanne Loring, a stem-cell biologist at the Scripps Research Institute in La Jolla, California, is concerned that theChinese trials use neural precursors[, which] can turn into other kinds of neurons, and could accumulate dangerous mutations during their many divisions, says Loring. Not knowing what the cells will become is troubling.

The GLP aggregated and excerpted this blog/article to reflect the diversity of news, opinion, and analysis. Read full, original post:Trials of embryonic stem cells to launch in China

Visit link:
Parkinson's target of China's first clinical trial using embryonic stem ... - Genetic Literacy Project

Similarities in human and pig embryos provide clues to early stages of development – Phys.Org

June 8, 2017 Developing human primordial germ cells (each small green and red cell is a PGC). Credit: Walfred Tang/Surani lab

Scientists have shown how the precursors of egg and sperm cells the cells that are key to the preservation of a species arise in the early embryo by studying pig embryos alongside human stem cells.

In research published today in Nature, researchers at the University of Cambridge and the University of Nottingham demonstrate how pig embryos and human embryonic cells show remarkable similarities in the early stages of their development. By combining these two models, they hope to improve our understanding of the origins of diseases such as paediatric germ cell tumours and fetal abnormalities.

Primordial germ cells, the precursors of eggs and sperm, are among the earliest cells to emerge in human embryos after implantation, appearing around day 17, while the surrounding cells go on to form the rest of the human body. However, little is understood about how they originate. Currently, the law prohibits culture of human embryos beyond 14 days, which prevents investigations on this and subsequent events such as gastrulation, when the overall body plan is established.

Now, researchers have used a combination of human and pig models of development to shed light on these events. They have shown for the first time that the interplay between two key genes is critical for the formation of the germline precursors and that this 'genetic cocktail' is not the same in all species.

First, by using human pluripotent embryonic stem cells in vitro, scientists led by Professor Azim Surani at the Wellcome Trust/Cancer Research UK Gurdon Institute established a model that simulates genetic and cellular changes occurring up to gastrulation. Human pluripotent embryonic stem cells are 'master cells' found in embryos, which have the potential to become almost any type of cell in the body.

As these stem cells can be multiplied and precisely genetically manipulated, the model system provides a powerful tool for detailed molecular analysis of how human cells transform into distinct cell types during early development, and which changes might underlie human diseases.

The work shows that when an embryo progresses towards gastrulation, cells temporarily acquire the potential to form primordial germ cells, but shortly afterwards lose this potential and instead acquire the potential to form precursors of blood and muscle (mesoderm) or precursors of the gut, lung and the pancreas (endoderm). The model also tells us that while the genes SOX17 and BLIMP1 are critical for germ cell fate, SOX17 subsequently has another role in the specification of endodermal tissues.

For an accurate picture of how the embryo develops, however, it is necessary to understand how cells behave in the three-dimensional context of a normal embryo. This cannot be achieved by studies on the most commonly used mouse embryos, which develop as egg 'cylinders', unlike the 'flat-disc' human embryos. Pig embryos, on the other hand, develop as flat discs (similar to human embryos), can be easily obtained, and are ethically more acceptable than working with non-human primate (monkey) embryos.

Researchers from the University of Nottingham dissected whole flat discs from pig embryos at different developmental stages and found that development of these embryos matches with the observations on the in vitro human model, as well as with non-human primate embryonic stem cells in vitro. For example, pig germ cells emerge in the course of gastrulation just as predicted from the human model, and with the expression of the same key genes as in human germ cells. Human and pig germ cells also exhibit key characteristics of this lineage, including initiation of reprogramming and re-setting of the epigenome modifications to our DNA that regulate its operations and have the potential to be passed down to our offspring which continues as germ cells progress towards development into sperm and eggs.

The combined human-pig models for early development and cell fate decisions likely reflect critical events in early human embryos in the womb. Altogether, knowledge gained from this approach can be applied to regenerative medicine for the derivation of relevant human cell types that might be used to help understand and treat human diseases, and to understand how mutations that perturb early development can result in human diseases.

Dr Ramiro Alberio, from the School of Biosciences at the University of Nottingham, says: "We've shown how precursors to egg and sperm cells arise in pigs and humans, which have similar patterns of embryo development. This suggests that the pig can be an excellent model system for the study of early human development, as well as improving our understanding of the origins of genetic diseases."

Dr Toshihiro Kobayashi in the Surani lab at the Gurdon Institute, adds: "We are currently prevented from studying human embryo development beyond day 14, which means that certain key stages in our development remain a mystery. The remarkable similarities between human and pig development suggest that we may soon be able to reveal the answers to some of our long-held questions."

Explore further: Mapping pluripotency differences between mice, monkeys, and humans

More information: Toshihiro Kobayashi et al. Principles of early human development and germ cell program from conserved model systems, Nature (2017). DOI: 10.1038/nature22812

Journal reference: Nature

Provided by: University of Cambridge

Not too shabby, humans. New research shows that certain primate stem cells have pluripotency superior to some types derived from mice. The study, published in Nature, maps how pluripotency differs among mice, monkeys, and ...

Researchers at the Babraham Institute have investigated the early stages of the development of cells called primordial germ cells and developed strategies to generate 'lookalike' cells in the lab. The generation of human ...

Scientists at the University of Cambridge working with the Weizmann Institute have created primordial germ cells - cells that will go on to become egg and sperm - using human embryonic stem cells. Although this had already ...

An important international summit on human gene editing recently recommended that researchers go ahead with gene editing human embryos, but keep revisiting how and when such modifications would be appropriate in the clinic. ...

Scientists at the University of Cambridge have managed to create a structure resembling a mouse embryo in culture, using two types of stem cells - the body's 'master cells' - and a 3D scaffold on which they can grow.

Researchers at Karolinska Institutet have identified cell surface markers specific for the very earliest stem cells in the human embryo. These cells are thought to possess great potential for replacing damaged tissue but ...

(Phys.org)A small team of researchers from Austria and Sweden has found that ravens are able to remember people who trick them for at least two months. In their paper published in the journal Animal Behavior, the group ...

Sex-changing fish exhibit differences in androgen receptor (AR) expression in muscles that are highly sensitive to androgens (male sex hormones) and essential for male courtship behavior, according to a Georgia State University ...

It's well known that young babies are more interested in faces than other objects. Now, researchers reporting in Current Biology on June 8 have the first evidence that this preference for faces develops in the womb. By projecting ...

Paleontologists investigating the sea bed off the coast of southern California have discovered a lost ecosystem that for thousands of years had nurtured communities of scallops and shelled marine organisms called brachiopods.

A Cornell study, published May 26 in the journal Nature Ecology and Evolution, describes how shifts in diets in Europeans after the introduction of farming 10,000 years ago led to genetic adaptations that favored the dietary ...

Wild capuchin monkeys readily learn skills from each otherbut that social learning is driven home by the payoff of learning a useful new skill. It's the first demonstration of "payoff bias" learning in a wild animal, and ...

Please sign in to add a comment. Registration is free, and takes less than a minute. Read more

Visit link:
Similarities in human and pig embryos provide clues to early stages of development - Phys.Org

Sowing Stem Cells: Lab-Grown Organoids Hold Promise for Patient Treatments – UCSF News Services

Illustration by Camila Carlow

Ophir Klein is growing teeth,which is just slightly less odd than what Jeffrey Bush is growing tissues that make up the face. Jason Pomerantz is growing muscle; Sarah Knox is growing salivary glands; and Edward Hsiao is printing 3-D bone using a machine that looks about as complex as a clock radio.

Together, these members of the UC San Francisco facultyare cultivating organs of the craniofacial complex the skull and face which too often go terribly wrong during fetal development. Deformities of these bones or soft tissues, the most common of birth defects, can cut life short by blocking the airway or circulation. Or they can disfigure a face so profoundly that a child strugglesto see, hear, or talk. Perhaps most painful of all, such deformities render children physically other, potentially leading to a lifetime of corrective surgeries and social isolation.

As director of the UCSF Program in Craniofacial Biology, Klein orchestrates a multisite research endeavor to translate basic science findings in tissue regeneration into improved treatments for these kids. Using stem cells from patients with craniofacial deformities, Klein, Bush, Pomerantz, Knox, Hsiao, and their colleagues are growing tiny functioning segments of organs, called organoids, to figure out exactly when and how in fetal development such design flaws occur.

Read a digital flipbook of the entire summer issue of UCSF Magazine, featuring this and other stories.

They are among scientists across UCSF who are cultivating cellular systems such as miniature brains and breasts from patient cells. They serve as dioramas of disease models derived from human cells either displacing or complementing the mouse models that have served science well, though inexactly, for manyyears. The effort is one of the most obvious and viable payoffs to date from stem cell science. With these organoids, physicians and scientists can not only trace the pathways of normal and abnormal development, but also test drugs and other treatments for their effectiveness in humans. Organoids are also one tiny step toward the ultimate goal of generating complete organs, as a way to circumvent rejection issues and save the lives of those who now die waiting for transplants.

As the reservoirs of human development, stem cells take it upon themselves to tirelessly renew and differentiate into the myriad cell types required to build out a body from an embryo. In creating an organoid, typical construction metaphors do not apply. There are no building blocks to nail, stack, or solder and no job-site supervisor barking orders. Thats not how biology works, says Zev Gartner, PhD, an associate professor of pharmaceutical chemistry.

It is a self-organizing process, he explains, a process that starts in the womb with embryonic stem cells (ESCs) or, in the case of organoids, induced pluripotent stem cells (iPSCs). iPSCs are mature cells that are stripped back to their earliest stage of development using a process devised by UCSF Professor of Anatomy Shinya Yamanaka, MD, PhD, who won a Nobel Prize for discovering the process. To make organoids, iPSCs are put through a series of solutions, then added to a gel that mimics the squishy 3-D cellular matrix of the embryo. The gel provides the right conditions for them to get to work.

Take an organ like the lung. Its basic functional units are a tube and a sac, and outside that sac are capillaries that allow gas exchange. Hundreds of millions of tubes and sacs make a lung, explains Gartner. You can make the little sacs and the tubes in a dish as an organoid model. But we dont know how to drive the self-organization of those units into much more complex, elaborate, highly ramified structures. The fundamental limitation of organoids is that they lack the vasculature that brings nutrient-laden blood to fuel the evolution of the larger structure.

Gartner notes that people who work with stem cells tend to focus on either regenerative medicine or disease modeling. Those interested in disease make models of tissues so that they can understand how diseases work, while those interested in regenerative medicine try to make models of healthy tissue that couldbe transplanted. Gartner straddles both camps. He grows breast organoids. The mammary gland is great because we can simultaneously think about these two phenomena as two sides of the same coin, he says. One is regenerative medicine through self-organization, and the other is understanding the progression of breast cancer through a breakdown in self-organization.

So theres potentially a triple payoff in stem cell science: By deducing how a breast forms itself, Gartner might figure out how to grow the entire organ. By tracing how cancer throws a wrench in the works, he may be able to target ways to stop that process. And by growing a human organ in a dish, he avoids making cross-species assumptions or putting animals or humans at risk in testing potential drugs to cure breast cancer, greatly accelerating the push toward a cure.

On Kleins team, Jeffrey Bush, PhD, an assistant professor of cell and tissue biology, looks at organoids through the lens of disease.

The organoids he grows model craniofrontonasal syndrome a birth defect that is caused by a mutation in a single gene and that dramatically impacts the shape of the face and head. He knows from studies reproducing craniofrontonasal syndrome in mice that the first place something goes wrong is in a cell type called the neuroectoderm. To create an organoid to study this, he obtained skin cells from Pomerantz, an associate professor of surgery, who has patients with the syndrome who were willing to donate tissue samples. Such collaborations between basic scientists and clinicians are key to bringing research out of the lab and into patient care.

We studied this simple system to see how this mutation affected the organization of these cells, says Bush. His group has filmed cells as they rush about to self-organize when theyre mixed together. In those films, he explains, you can see that the mutated cells, which are dyed red, segregate from the normal cells, which are green they are like oil and water. In other words, the mutated cells completely disrupt the behavior of all the cells. By contrast, in the films of cells without the mutation, all the cells circulate easily among one another, like fish in an aquarium. This understanding has allowed Bush to begin to think about a drug that blocks this separation. He has several promising candidates that his team will test in pregnant mice. Right now, he says, there isnt a single drug that we can use for any kind of structural birth defects. If we could show that a medication blocks the effects of this mutation, it would serve as proof of principle that something besides surgery can be done. But we would have to know that it was safe for mother and child and that we could catch it early enough.

Jason Pomerantz, MD, a plastic surgeon, falls into the regeneration camp. His clinical work is typified by a recent eight-hour operationon a 17-year-old boy with Crouzon syndrome, a severely disfiguring condition affecting every organ in the craniofacial structure muscle, bone, and skin. My patient is excited for the outcome, but not about the process, says Pomerantz, surgical director of the UCSF Craniofacial Center. For three months, the patient will wear a large metal frame on his head with wires that will pull the bones in his face forward. Prior to the surgery, the boys face was nearly concave, collapsed inward at the nose.

Yet bone is not all Pomerantz needs to work with to restructure a face. The subtle bends, creases, and curves of expressionthat make a face ones own are the work of tiny muscles.Right now we can move a big muscle say, from the thighto the face so that people can smile, he says. But we cant reconstruct the fine ones that enable people to move their eyebrows up or move the eyeballs around. That requires little muscles. This is where we can make headway with stem cell biology.

We have actually made a humanized organ in an animal, he continues, pointing to a picture of a mouse on his wall. Pomerantz is now considering incubating small human muscles in animals for use in his patients faces. In a recent project, he inserted stem cells from human muscles into a mouse whose own muscle stem cells had been incapacitated. He then perturbed the muscle to stimulate regeneration. As the muscle healed, the cells created new muscle tissue, which the mouses nerves innervated to make a functioning muscle. Its exactly the size of the muscles Pomerantz needs for full articulation of expression and function in a human face or hand.

Muscles are part of a vast and intricate system strewn throughoutthe body. Teeth, on the other hand, are islands unto themselves. Teeth intrigue me from a regeneration perspective, says OphirKlein, MD, PhD, chair of the Division of Craniofacial Anomalies, the Hillblom Professor of Craniofacial Anomalies, the Epstein Professor of Human Genetics, and a resident alumnus. They are discrete organs all the parts are there. More intriguing still is the fact that many rodents have the ability to grow their front teeth continuously. Elephants and walruses also have ever-growing tusks, and even some primates lemurs can regrow their teeth.

In studying species that can [regrow teeth], we hope to unlock the regenerative potential in our own cells.

Ophir Klein, MD, PhD

Chair of the Division of Craniofacial Anomalies

A tooth can be regenerated in parts. Stem cells can be used to grow the root, and then a crown can be added to complete the tooth. To generate a whole organ at once, Kleins colleagues are planning to partner with bioengineers who can produce a biocompatible material that could serve as a framing device to jump-start the creation of dentin, one of the hard components of a tooth. If they start with the right cells, then the scaffolding will give the cellsthe shape information they need to create the right design. But even that isnt Kleins endgame. In my lab, were interested in figuring out why humans cant regrow teeth, he says. In studying species that can, we hope to unlock the regenerative potential in our own cells that might be turned off.

Kleins work to generate teeth is inspired by his patients with ectodermal dysplasia, a congenital disorder characterized by lack of sweat glands, hair, or teeth. Being able to generate the roots of teeth would be remarkable for these patients, since the rest can be done with a crown. Right now, they must be fitted with dentures.

Klein is also taking another tack to help these patients. We completed a clinical trial of a drug that basically goosed up the development of the organs when they werent forming properly, he says. The drug a protein developed by Swiss collaborators of Kleins, based on studies of embryonic mice, who develop theseorgans in early- to mid-gestation was given to infants with the disorder right after birth. The trial was unsuccessful. Now, scientists in Germany are running a trial of the same drug, giving it instead to mothers carrying babies with this genetic disorder. The scientists will try to gauge what the best timing is for delivering the drug.

Whats great about this drug is that it doesnt seem to have any effects on any other organs besides teeth,hair, and sweat glands, says Klein. Drugs for other conditions are far riskier, because they affect pathways that are important inthe development of many organs.

Sarah Knox, PhD, an assistant professor of cell and tissue biology, is using stem cells to figure out howto regenerate salivary glands compromised by radiation treatments for head and neckcancers or by craniofacial deformities. Her focus is on how the environment contributes to the activation and maintenance ofthe gland. The salivary gland, like all organs, is continuously replenishing the supply ofcells and tissues it needs to function. Knoxs research shows that the gland takesdirectional cues from nearby nerve cells not only to remain functional, but also to continuously replace itself. Her organoids are made of cells from a patient and nerve cells (ganglia) from a fetal mouse. We are trying to explore the relationship between the stem cells and the nerves, she says. How do the nerves know the tissue is there? How do the nerves provide instruction and feedback? Individual cells die off and new cells have to replace them. Organoids are giving us insight as to where those new cells are coming from and how we keep repopulating [them] all our lives.

As head of the UCSF Program in Craniofacial Biology which is based in the School of Dentistry and the Division of Genetics in the School of Medicine Klein stands at one of sciences most compelling crossroads: regenerative medicine and genetics. Far in the future, both fields have potential that seem like science fiction today. We live in a world where people die waiting for organ transplants. What if we could pull these organoids from their petri dish and supply them with the fuel they need to become full-blown organs? Such a feat would necessitate either a host embryo perhaps from apig, because pigs have organs the size of human organs or some other biological foundation. Some scientists are hoping to jump-start organ development with scaffolding, or cells engineered to speed the developmental process. Others are zeroing in on the genome, particularly in kids with craniofacial anomalies caused by just one mutation, like craniofrontonasal syndrome; for example, a tool called CRISPR could allow scientists to splice that gene out and replace it with a normal gene. But the tool has yet to be used in humans, let alone a human fetus.

Ethical questions pepper either route. At their best, stem cells regenerate tissues; at their worst, they go rogue and grow into a tumor. Yet with gene editing tools like CRISPR, you literally have the potential to change the species, says Klein. And in both scenarios, the cells can act with unforeseen off-target effects. Klein and his colleagues are in continual discussion about the repercussions of their work with the director of UCSF Bioethics, Barbara Koenig, RN, PhD 88. Gene therapy is an example of an exciting new treatment that cured one serious pediatric illness severe combined immunodeficiency syndrome (SCID) but the genes unwittingly led to the development of leukemia, explains Koenig. Genetic and stem cell interventions must be painstakingly studied before application. And, once they are ready, who will regulate them? Thereare many questions yet to be answered. The challenges are most extreme when we talk about modifying an egg or sperm cell, where the changes are passed on to the next generation.

So Klein and his colleagues proceed with caution, curiosity, and awe. The next decade will be an incredibly exciting time, says Klein. With continual advances in human genetics and developmental and cell biology, we hope to be able to make drugs and use genetic tools to appreciably change the lives of our patients.

Bone grows like a runaway train in Edward Hsiaos patients with fibrodysplasia ossificans progressiva (FOP). The slightest bump or injury can set off a spurt of bone growth that can fuse their vertebrae, lock their joints, or even freeze up their rib cages, leaving them unable to breathe.

No one, to date, has successfully engineered bone. Hsiao, MD, PhD, is hoping to spark the process with the help of a 3-D printer from Organovo, a firm that specializes in bioprinting technology. From iPSCs, he can make many of the essential ingredients of bone, including mesenchymal stem cells, endothelial cells, and macrophages. We are putting cells into the equivalent of an ink. Then we will print the structures with the ink, let the ink dissolve, and leave the cells, explains Hsiao. The hope is that the cells can then recapitulate the normal developmental process.

If the approach is successful, Hsiao hopes to use the resulting models to test drugs and other treatments to halt or prevent bone deformities. Down the line, his progress also stands to transform bone and joint replacements. Through his work with FOP, hes uncovered one mechanism that drives rapid bone growth. In these patients, we know that mature bone formation can happen in as quickly as two weeks, so it is possible to grow bone in an adult. We need to understand how to modulate that, says Hsiao. Someday, my dream would be to be able to identify the cells we need, give someone a drug that induces the right genes and recruits the right cells to the correct site, and have the cells rebuild the joint from scratch.

More here:
Sowing Stem Cells: Lab-Grown Organoids Hold Promise for Patient Treatments - UCSF News Services