Monday, November 12, 2012

Light-Based 'Remote Control' Developed for Proteins Inside Cells


ScienceDaily (Nov. 8, 2012) — Scientists at Stanford University have developed an intracellular remote control: a simple way to activate and track proteins, the busiest of cellular machines, using beams of light.

The new method, described in a paper to be published Nov. 9 in Science, will let researchers shine light on a specific cell region to quickly activate a protein in that area, producing an unusually fine-grained view of the location and timing of protein activity. In addition, the method may eventually enable physicians to direct the movement and activity of stem cells used to treat injury or illness in light-accessible body parts, such as the eye or skin. Stanford has filed a patent application for the work.

The method involves splicing two pieces of a specific fluorescent protein to other proteins of interest. The resulting hybrids -- called fluorescent, light-inducible proteins, or FLIPs -- have two interesting features: Not only are they turned on by light, but they also glow less brightly when activated, a change that provides an easy way to sense protein activity.

"It's sort of like having a garage door opener that also tells you if the garage door is open or closed," said Michael Lin, MD, PhD, an assistant professor of pediatrics and of bioengineering and the senior author of the paper. "I'm always driving out of my house, closing the garage door, and then wondering after I drive away if it's shut, so I have to drive back and check." If garage doors were like FLIPs, Lin would be spared his return trip, since these proteins not only turn on at the flip of a light switch, but also tell an observer that they're working. "One molecule can tell you where it is and what it's doing," said Lin.

The trick to the new method is that it uses pieces of a Velcro-like fluorescent protein called Dropna. In the dark, Dropna units adhere to each other and fluoresce. Under cyan-colored light, the units detach and begin to dim. Lin's team spliced a Dropna unit to each end of the proteins they wanted to study to make the FLIPs. In the dark, the Dropna units stuck together and physically blocked the active sites of the proteins under study. When cyan-colored light was shone on the proteins, the Dropna units fell apart, exposing the protein's active site so it could work. Under cyan light, the Dropna units also glowed less brightly, signaling that the FLIP was switched on.

It's easiest to build FLIPs from proteins that fold with both their head and tail ends near the active site, though the research team is now figuring out how to attach Dropna units to other parts of a protein, not just an end. With that modification, Lin anticipates that FLIPs could be created from most proteins that scientists want to investigate.

"For science geeks, this is very interesting in that it converges two exciting fields: biological sensing, which has been dominated by fluorescent proteins, and optogenetics, the use of light to investigate biology," Lin said.

In the past, scientists who specialized in biological sensing have tagged bits of the cellular machinery with fluorescent proteins to see where certain processes occurred in the cell. Separately, optogenetics experts -- using methods that originated at Stanford -- have figured out how to switch on specific neuron circuits with light. Lin's method combines advantageous features of both techniques, and is the first instance of optogenetics-type techniques being applied to individual proteins.

Outside the research lab, the method could be used to give directions to stem cells injected for therapeutic purposes. For instance, if the stem cells were engineered to contain FLIPs that control cell motility, a beam of light could then direct implanted stem cells to a particular location. Similarly, FLIPs and appropriately timed light beams could be used to control what a stem cell does when it reaches its destination.

"If you think about how we might want to use stem cells to regenerate tissues, we may need control over where cells go, when they proliferate and when they die," Lin said. At present, this application seems most likely for tissues at the body's surface, such as the eye and skin, because physicians would need to be able to deliver light to the treatment site.

http://www.sciencedaily.com/releases/2012/11/121108142744.htm?utm_source=feedburner&utm_medium=email&utm_campaign=Feed%3A+sciencedaily+%28ScienceDaily%3A+Latest+Science+News%29

Hunting Neuron Killers in Alzheimer's and Traumatic Brain Injury


ScienceDaily (Nov. 9, 2012) — Sanford-Burnham researchers discovered that the protein appoptosin prompts neurons to commit suicide in several neurological conditions -- giving them a new therapeutic target for Alzheimer's disease and traumatic brain injury.

Dying neurons lead to cognitive impairment and memory loss in patients with neurodegenerative disorders-conditions like Alzheimer's disease and traumatic brain injury. To better diagnose and treat these neurological conditions, scientists first need to better understand the underlying causes of neuronal death.

Enter Huaxi Xu, Ph.D., professor in Sanford-Burnham's Del E. Webb Neuroscience, Aging, and Stem Cell Research Center. He and his team have been studying the protein appoptosin and its role in neurodegenerative disorders for the past several years. Appoptosin levels in the brain skyrocket in conditions like Alzheimer's and stroke, and especially following traumatic brain injury.

Appoptosin is known for its role in helping the body make heme, the molecule that carries iron in our blood (think "hemoglobin," which makes blood red). But what does heme have to do with dying brain cells? As Xu and his group explain in a paper they published recently in the Journal of Neuroscience, excess heme leads to the overproduction of reactive oxygen species, which include cell-damaging free radicals and peroxides, and triggers apoptosis, the carefully regulated process of cellular suicide. This means that more appoptosin and more heme cause neurons to die.

Not only did Xu and his team unravel this whole appoptosin-heme-neurodegeneration mechanism, but when they inhibited appoptosin in laboratory cell cultures, they noticed that the cells didn't die. This finding suggests that appoptosin might make an interesting new therapeutic target for neurodegenerative disorders.

What's next? Xu and colleagues are now probing appoptosin's function in mouse models. They're also looking for new therapies that target the protein.

"Since the upregulation of appoptosin is important for cell death in diseases such as Alzheimer's, we're now searching for small molecules that modulate appoptosin expression or activity. We'll then determine whether these compounds may be potential drugs for Alzheimer's or other neurodegenerative diseases," Xu explains.
Putting a stop to runaway appoptosin won't be easy, though. That's because we still need the heme-building protein to operate at normal levels for our blood to carry iron. In a previous study, researchers found that a mutation in the gene that encodes appoptosin causes anemia. "Too much of anything is bad, but so is too little," Xu says.

New therapies that target neurodegenerative disorders and traumatic brain injury are sorely needed. According to the CDC, approximately 1.7 million people sustain a traumatic brain injury each year. It's an acute injury, but one that can also lead to long-term problems, causing epilepsy and increasing a person's risk for Alzheimer's and Parkinson's diseases. Not only has traumatic brain injury become a worrisome problem in youth and professional sports in recent years, the Department of Defense calls traumatic brain injury "one of the signature injuries of troops wounded in Afghanistan and Iraq."

http://www.sciencedaily.com/releases/2012/11/121109111509.htm?utm_source=feedburner&utm_medium=email&utm_campaign=Feed%3A+sciencedaily+%28ScienceDaily%3A+Latest+Science+News%29

Thursday, November 8, 2012

Reactions to Everyday Stressors Predict Future Health


ScienceDaily (Nov. 2, 2012
Contrary to popular perception, stressors don't cause health problems -- it's people's reactions to the stressors that determine whether they will suffer health consequences, according to researchers at Penn State.


"Our research shows that how you react to what happens in your life today predicts your chronic health conditions and 10 years in the future, independent of your current health and your future stress," said David Almeida, professor of human development and family studies.

 "For example, if you have a lot of work to do today and you are really grumpy because of it, then you are more likely to suffer negative health consequences 10 years from now than someone who also has a lot of work to do today, but doesn't let it bother her."

Using a subset of people who are participating in the MIDUS (Midlife in the United States) study, a national longitudinal study of health and well being that is funded by the National Institute on Aging, Almeida and his colleagues investigated the relationships among stressful events in daily life, people's reactions to those events and their health and well being 10 years later.

Specifically, the researchers surveyed by phone 2,000 individuals every night for eight consecutive nights regarding what had happened to them in the previous 24 hours. They asked the participants questions about their use of time, their moods, the physical health symptoms they had felt, their productivity and the stressful events they had experienced, such as being stuck in traffic, having an argument with somebody, or taking care of a sick child.

"Most social-science surveys are based on long retrospective accounts of your life in the past month or maybe the past week," Almeida said. "By asking people to focus just on the past 24 hours, we were able to capture a particular day in someone's life. Then, by studying consecutive days, we were able to see the ebb and flow of their daily experiences."

The researchers also collected saliva samples from the 2,000 individuals at four different times on four of those eight days. From the saliva, they were able to determine amounts of the stress hormone, cortisol. They then linked the information they collected to data from the larger MIDUS study, including the participants' demographic information, their chronic health conditions, their personalities and their social networks.

"We did this 10 years ago in 1995 and again in 2005," Almeida said. "By having longitudinal data, not only were we able to look at change in daily experiences over this time but how experiences that were occurring 10 years ago are related to health and well being now."

The team found that people who become upset by daily stressors and continue to dwell on them after they have passed were more likely to suffer from chronic health problems -- especially pain, such as that related to arthritis, and cardiovascular issues -- 10 years later.

"I like to think of people as being one of two types," Almeida said. "With Velcro people, when a stressor happens it sticks to them; they get really upset and, by the end of the day, they are still grumpy and fuming. With Teflon people, when stressors happen to them they slide right off. It's the Velcro people who end up suffering health consequences down the road."

The results appear online in the current issue of Annals of Behavioral Medicine
According to Almeida, certain types of people are more likely to experience stress in their lives. Younger people, for example, have more stress than older people; people with higher cognitive abilities have more stress than people with lower cognitive abilities; and people with higher levels of education have more stress than people with less education.

"What is interesting is how these people deal with their stress," said Almeida. "Our research shows that people age 65 and up tend to be more reactive to stress than younger people, likely because they aren't exposed to a lot of stress at this stage in their lives, and they are out of practice in dealing with it. Younger people are better at dealing with it because they cope with it so frequently. Likewise, our research shows that people with lower cognitive abilities and education levels are more reactive to stress than people with higher cognitive abilities and education levels, likely because they have less control over the stressors in their lives."

While stress may be a symptom that a person's life is filled with hardship, it could also simply mean that the person is engaged in a wide variety of activities and experiences.

"If this is the case, reducing exposure to stressors isn't the answer," said Almeida. "We just need to figure out how to manage them better."

The National Institutes of Health provided funding for this research. Other authors on the paper include Susan Charles of the University of California at Irvine, Jennifer Piazza of California State University at Fullerton, and Martin Sliwinski and Jacquie Mogle, both at Penn State.

Tuesday, August 21, 2012

Why are elderly duped?


Why are elderly duped? Area in brain where doubt arises changes with age


ScienceDaily (Aug. 16, 2012) — Everyone knows the adage: "If something sounds too good to be true, then it probably is." Why, then, do some people fall for scams and why are older folks especially prone to being duped?
An answer, it seems, is because a specific area of the brain has deteriorated or is damaged, according to researchers at the University of Iowa. By examining patients with various forms of brain damage, the researchers report they've pinpointed the precise location in the human brain, called the ventromedial prefrontal cortex, that controls belief and doubt, and which explains why some of us are more gullible than others.
"The current study provides the first direct evidence beyond anecdotal reports that damage to the vmPFC (ventromedial prefrontal cortex) increases credulity. Indeed, this specific deficit may explain why highly intelligent vmPFC patients can fall victim to seemingly obvious fraud schemes," the researchers wrote in the paper published in a special issue of the journalFrontiers in Neuroscience.
A study conducted for the National Institute of Justice in 2009 concluded that nearly 12 percent of Americans 60 and older had been exploited financially by a family member or a stranger. And, a report last year by insurer MetLife Inc. estimated the annual loss by victims of elder financial abuse at $2.9 billion.
The authors point out their research can explain why the elderly are vulnerable.
"In our theory, the more effortful process of disbelief (to items initially believed) is mediated by the vmPFC, which, in old age, tends to disproportionately lose structural integrity and associated functionality," they wrote. "Thus, we suggest that vulnerability to misleading information, outright deception and fraud in older adults is the specific result of a deficit in the doubt process that is mediated by the vmPFC."
The ventromedial prefrontal cortex is an oval-shaped lobe about the size of a softball lodged in the front of the human head, right above the eyes. It's part of a larger area known to scientists since the extraordinary case of Phineas Gage that controls a range of emotions and behaviors, from impulsivity to poor planning. But brain scientists have struggled to identify which regions of the prefrontal cortex govern specific emotions and behaviors, including the cognitive seesaw between belief and doubt.
The UI team drew from its Neurological Patient Registry, which was established in 1982 and has more than 500 active members with various forms of damage to one or more regions in the brain. From that pool, the researchers chose 18 patients with damage to the ventromedial prefrontal cortex and 21 patients with damage outside the prefrontal cortex. Those patients, along with people with no brain damage, were shown advertisements mimicking ones flagged as misleading by the Federal Trade Commission to test how much they believed or doubted the ads. The deception in the ads was subtle; for example, an ad for "Legacy Luggage" that trumpets the gear as "American Quality" turned on the consumer's ability to distinguish whether the luggage was manufactured in the United States versus inspected in the country.
Each participant was asked to gauge how much he or she believed the deceptive ad and how likely he or she would buy the item if it were available. The researchers found that the patients with damage to the ventromedial prefrontal cortex were roughly twice as likely to believe a given ad, even when given disclaimer information pointing out it was misleading. And, they were more likely to buy the item, regardless of whether misleading information had been corrected.
"Behaviorally, they fail the test to the greatest extent," says Natalie Denburg, assistant professor in neurology who devised the ad tests. "They believe the ads the most, and they demonstrate the highest purchase intention. Taken together, it makes them the most vulnerable to being deceived." She added the sample size is small and further studies are warranted.
Apart from being damaged, the ventromedial prefrontal cortex begins to deteriorate as people reach age 60 and older, although the onset and the pace of deterioration varies, says Daniel Tranel, neurology and psychology professor at the UI and corresponding author on the paper. He thinks the finding will enable doctors, caregivers, and relatives to be more understanding of decision making by the elderly.
"And maybe protective," Tranel adds. "Instead of saying, 'How would you do something silly and transparently stupid,' people may have a better appreciation of the fact that older people have lost the biological mechanism that allows them to see the disadvantageous nature of their decisions."
The finding corroborates an idea studied by the paper's first author, Erik Asp, who wondered why damage to the prefrontal cortex would impair the ability to doubt but not the initial belief as well. Asp created a model, which he called the False Tagging Theory, to separate the two notions and confirm that doubt is housed in the prefrontal cortex.
"This study is strong empirical evidence suggesting that the False Tagging Theory is correct," says Asp, who earned his doctorate in neuroscience from the UI in May and is now at the University of Chicago.
Kenneth Manzel, Bryan Koestner, and Catherine Cole from the UI are contributing authors on the paper. The National Institute on Aging and the National Institute of Neurological Disorders and Stroke funded the research.

Journal Reference:
  1. Erik Asp, Kenneth Manzel, Bryan Koestner, Catherine A. Cole, Natalie L. Denburg, Daniel Tranel. A Neuropsychological Test of Belief and Doubt: Damage to Ventromedial Prefrontal Cortex Increases Credulity for Misleading AdvertisingFrontiers in Neuroscience, 2012; 6 DOI: 10.3389/fnins.2012.00100



A GPS in your DNA


ScienceDaily (Aug. 16, 2012) — While your DNA is unique, it also tells the tale of your family line. It carries the genetic history of your ancestors down through the generations. Now, says a Tel Aviv University researcher, it's also possible to use it as a map to your family's past.
Prof. Eran Halperin of TAU's Blavatnik School of Computer Science and Department of Molecular Microbiology and Biotechnology, along with a group of researchers from University of California, Los Angeles, are giving new meaning to the term "genetic mapping." Using a probabilistic model of genetic traits for every coordinate on the globe, the researchers have developed a method for determining more precisely the geographical location of a person's ancestral origins.
The new method is able to pinpoint more specific locations for an individual's ancestors, for example placing an individual's father in Paris and mother in Barcelona. Previous methods would "split the difference" and place this origin inaccurately at a site between those two cities, such as Lyon.
Published in the journal Nature Genetics, this method has the potential to reveal the ancestry, origins, and migration patterns of many different human and animal populations. It could also be a new model for learning about the genome.

Points of origin
There are points in the human genome called SNPs that are manifested differently in each individual, explains Prof. Halperin. These points mutated sometime in the past and the mutation was then passed to a large part of the population in a particular geographic region. The probability of a person possessing these mutations today varies depending on the geographical location of those early ancestors.
"We wanted to ask, for example, about the probability of having the genetic mutation 'A' in a particular position on the genome based on geographical coordinates," he says. When you look at many of these positions together in a bigger picture, it's possible to group populations with the same mutation by point of origin.
To test their method, Prof. Halperin and his fellow researchers studied DNA samples from 1,157 people from across Europe. Using a probabilistic mathematical algorithm based on mutations in the genome, they were able to accurately determine their ancestral point or points of origin using only DNA data and the new mathematical model, unravelling genetic information to ascertain two separate points on the map for the mother and father. The researchers hope to extend this model to identify the origins of grandparents, great-grandparents, and so on.
The new method could provide information that has applications in population genetic studies -- to study a disease that impacts a particular group, for example. Researchers can track changes in different genomic traits across a map, such as the tendency for southern Europeans to have a mutation in a gene that causes lactose intolerance, a mutation missing from that gene in northern Europeans.

A closer look at migration
The researchers believe that their model could have also relevance for the animal kingdom, tracking the movement of animal populations. "In principle, you could figure out where the animals have migrated from, and as a result learn about habitat changes due to historical climate change or other factors," says Prof. Halperin.

B Cell Survival Holds Key to Chronic Graft Vs. Host Disease


B cell survival holds key to chronic graft vs. host disease


ScienceDaily (Aug. 16, 2012) — A team from UNC Lineberger Comprehensive Cancer Center, shows in the laboratory that B cells from patients with chronic GVHD are much more active than cells from patients without the disease.
In chronic Graft vs. Host Disease (GVHD), the differences between the donor bone marrow cells and the recipient's body often cause these immune cells to recognize the recipient's body tissues as foreign and the newly transplanted cells attack the transplant recipient's body. Symptoms can range from dry eyes and dry mouth, hair loss and skin rashes, vulnerability to infection, liver and lung and digestive tract disorders. For patients who received bone marrow or stem cells, it is estimated that 40-70 percent may experience chronic GVHD.
B cells, which produce proteins called antibodies, are one type of immune cell involved in GVHD. In a paper published online August 15 by the journal, Blood, a team from the University of North Carolina's Lineberger Comprehensive Cancer Center, shows in the laboratory that B cells from patients with chronic GVHD are much more active than cells from patients without the disease. The team also outlines the cell signaling pathways that contribute to this increased activity -- identifying a promising target for developing new therapies for the diseases.
Jessica Allen, PhD, the paper's first author, says "We found that B cells from patients with active chronic GVHD were in a heightened metabolic state and resist programmed cell death."
Senior author, Stefanie Sarantopoulos, MD, PhD, assistant professor in the division of hematology/oncology and the departments of microbiology and immunology at the UNC School of Medicine, adds, "Steroids are currently our only standard treatment for chonic GVHD and they are often not effective. This study adds to our previously published work because it implicates the TNF family member protein called BAFF in the 'revved up' B-cell signaling we found in our patients. We hope to develop targeted therapeutic agents, like anti-BAFF agents or small molecule inhibitors of serine/ threonine kinases, for treatment of our chronic GVHD patients."

[Other UNC Lineberger researchers on the team include Albert Baldwin, PhD, Jonathan Serody, MD, Kristy Richards, MD, PhD, Thomas C. Shea, MD, Don A. Gabriel, MD, PhD, James Coghill, MD, Paul Armistead, MD, PhD, Matthew Fore, BA, and Jenna Wooten, PhD.
Additional team members from the UNC Stem Cell Transplant team and UNC Lineberger include Philip Roehrs, MD, Amber Essenmacher, Robert Irons, Allison Deal, Andrew Sharf, and Todd Hoffert.
Jerome Ritz MD, Corey Cutler, MD MPH, and Nazmim S. Bhuiya, BA, MPH from the Dana-Farber Cancer Institute also contributed to the research.
The study could not have been performed without the gracious consent of transplant patients at UNC and Dana Farber who donated extra blood samples. The University Cancer Research Fund (UCRF) and the UNC Tissue Procurement Core Facility supported collection of de-identified patient samples for these laboratory studies.
The research was funded by the National Marrow Donor Program through the Be The Match Foundation and National Institutes of Health grants K08HL107756 and CA142106.
Leukemia and lymphoma patients who receive life-saving stem cell or bone marrow transplants often experience chronic side effects that significantly decrease quality of life, can last a lifetime, and ultimately affect their long-term survival.]

molecular trigger for wound-healing


Scientists find an important molecular trigger for wound-healing


ScienceDaily (Aug. 16, 2012) — Scientists at The Scripps Research Institute have made a breakthrough in understanding a class of cells that help wounds in skin and other epithelial tissues heal, uncovering a molecular mechanism that pushes the body into wound-repair mode.
The findings, which appear in an advance, online version of the Immunity on August 16, 2012, focus on cells known as γδ (gamma delta) T cells. The new study demonstrates a skin-cell receptor hooks up with a receptor on γδ T-cells to stimulate wound healing.
"This is a major activation pathway for γδ T cells, and it may be a key to treating slow-wound-healing conditions, such as we see in diabetes," said Scripps Research Professor Wendy L. Havran, senior author of the study. "Chronic non-healing wounds among diabetics and the elderly are an increasing clinical problem."

Rounding and Multiplying
Havran's laboratory specializes in the study of γδ T cells, and the team has produced many of the findings in this research field, including the discovery of these cells' major role in epithelial wound repair. Epithelial tissues are barrier tissues to the outside world, such as skin and the inner surfaces of the gut and lungs.
Normally, γδ T cells reside in these tissues and extend finger-like projections, called dendrites, that contact neighboring epithelial cells. When injury or infection occurs, the epithelial cells signal their damaged condition to the γδ T cells. In response, the T cells retract their dendrites, become round, start proliferating, and secrete growth-factor proteins that stimulate the production of new epithelial cells in the vicinity -- thus helping to repair the wound.
Researchers know of very few interactions between epithelial cells and γδ T cells that are involved in this process. Two, however, are known to be crucial. One of these is through the gd T cell receptor and the other was described in a 2010 Science paper, whose first author was Havran laboratory Senior Staff Scientist Deborah A. Witherden. But these two interactions don't fully explain the transformation that γδ T cells undergo in the vicinity of wounds. "We've wanted to learn more about the molecules that mediate this dramatic change," Havran said.

Signaling a Transformation
To do that, Witherden identified an antibody that could block keratinocytes' ability to activate γδ T cells in culture. She found that the antibody bound to a keratinocyte surface receptor called plexin B2. She also found that when lab mice have small skin wounds, their injured keratinocytes express more plexin B2 soon after the wounding occurs -- pointing to a role for plexin B2 in signaling skin-cell damage.
The next step was to find plexin B2's signaling partner on γδ T cells. "Plexin B2 is very similar to other plexin B family members, including plexin B1, which previously has been shown to bind the CD100 receptor on T cells," said Witherden. "So we thought that perhaps plexin B2 and CD100 can interact as well."
Further tests revealed that plexin B2 and CD100 do indeed bind tightly together; moreover, γδ T cells can't go fully into wound-repair mode when they lack CD100. Witherden found as well that skin wounds in mice take an extra day or two to heal when the mice don't have this receptor. "This is very similar to what we see in mice that lack γδ T cells altogether," she said.
Removing CD100 from other types of T cells had no effect on wound healing time, indicating that the absence of this receptor specifically on γδ T cells is the reason for the slower healing.
By stimulating CD100 with plexin B2 molecules or even with CD100-binding antibodies, the team showed that this receptor is the principal trigger for the dramatic appendage-retraction and rounding phenomenon seen in γδ T cells after nearby wounds. Without it, the T cells are largely unable to undergo this transformation. "This rounding process seems to be vital for these T cells to function normally in wound healing," said Witherden.
Potential Clinical Significance
In early follow-on work, the team has found evidence that this same plexin B2-CD100 interaction is also needed for the prompt activation of γδ T cells and wound healing in the lining of mouse intestines -- which suggests that this receptor helps govern wound healing in epithelial tissues generally.
The finding clearly is important for the basic scientific understanding of T cells and their functions. But it is likely to have medical significance, too. Non-healing wounds affect more than 4 million people in the United States and are the leading cause of amputations. These chronic wounds have a major impact on patient's lives and result in enormous health care costs. "If deficiencies in this γδ T cell activation pathway are even partly responsible, then we may be able to develop drugs to boost this pathway and treat conditions involving chronic non-healing wounds," said Havran.
The γδ T cell population appears to be involved not just in wound healing, but also in defending against other threats to epithelial tissues. "One of the future directions of our research will be to understand the roles of these molecules in gd T cell activation pathways in fighting infections and tumors," she added.
Journal Reference:
  1. Deborah A. Witherden, Megumi Watanabe, Olivia Garijo, Stephanie E. Rieder, Gor Sarkisyan, Shane J.F. Cronin, Petra Verdino, Ian A. Wilson, Atsushi Kumanogoh, Hitoshi Kikutani, Luc Teyton, Wolfgang H. Fischer, Wendy L. Havran. The CD100 Receptor Interacts with Its Plexin B2 Ligand to Regulate Epidermal γδ T Cell Function.Immunity, 2012; DOI: 10.1016/j.immuni.2012.05.026


increase in size of the human brain explained


Evolutionary increase in size of the human brain explained: Part of a protein linked to rapid change in cognitive ability


ScienceDaily (Aug. 16, 2012) — Researchers have found what they believe is the key to understanding why the human brain is larger and more complex than that of other animals.
The human brain, with its unequaled cognitive capacity, evolved rapidly and dramatically.
"We wanted to know why," says James Sikela, PhD, who headed the international research team that included researchers from the University of Colorado School of Medicine, Baylor College of Medicine and the National Institutes of Mental Health. "The size and cognitive capacity of the human brain sets us apart. But how did that happen?"
"This research indicates that what drove the evolutionary expansion of the human brain may well be a specific unit within a protein -- called a protein domain -- that is far more numerous in humans than other species."
The protein domain at issue is DUF1220. Humans have more than 270 copies of DUF1220 encoded in the genome, far more than other species. The closer a species is to humans, the more copies of DUF1220 show up. Chimpanzees have the next highest number, 125. Gorillas have 99, marmosets 30 and mice just one. "The one over-riding theme that we saw repeatedly was that the more copies of DUF1220 in the genome, the bigger the brain. And this held true whether we looked at different species or within the human population."
Sikela, a professor at the CU medical school, and his team also linked DUF1220 to brain disorders. They associated lower numbers of DUF1220 with microcephaly, when the brain is too small; larger numbers of the protein domain were associated with macrocephaly, when the brain is too large.
The findings were reported today in the online edition of The American Journal of Human Genetics. The researchers drew their conclusions by comparing genome sequences from humans and other animals as well as by looking at the DNA of individuals with microcephaly and macrocephaly and of people from a non-disease population.
"The take home message was that brain size may be to a large degree a matter of protein domain dosage," Sikela says. "This discovery opens many new doors. It provides new tools to diagnose diseases related to brain size. And more broadly, it points to a new way to study the human brain and its dramatic increase in size and ability over what, in evolutionary terms, is a short amount of time."

Journal Reference:
  1. Laura J. Dumas, Majesta S. O’Bleness, Jonathan M. Davis, C. Michael Dickens, Nathan Anderson, J.G. Keeney, Jay Jackson, Megan Sikela, Armin Raznahan, Jay Giedd, Judith Rapoport, Sandesh S.C. Nagamani, Ayelet Erez, Nicola Brunetti-Pierri, Rachel Sugalski, James R. Lupski, Tasha Fingerlin, Sau Wai Cheung, James M. Sikela. DUF1220-Domain Copy Number Implicated in Human Brain-Size Pathology and EvolutionThe American Journal of Human Genetics, 2012; DOI: 10.1016/j.ajhg.2012.07.016

Thursday, July 19, 2012

Stress fuels breast cancer metastasis to bone

Stress can promote breast cancer cell colonization of bone, Vanderbilt Center for Bone Biology investigators have discovered.

The studies, reported July 17 in PLoS Biology, demonstrate in mice that activation of the sympathetic nervous system – the "fight-or-flight" response to stress – primes the bone environment for breast cancer cell metastasis. The researchers were able to prevent breast cancer cell lesions in bone using propranolol, a cardiovascular medicine that inhibits sympathetic nervous system signals.
Metastasis – the spread of cancer cells to distant organs, including bone – is more likely to kill patients than a primary breast tumor, said Florent Elefteriou, Ph.D., director of the Vanderbilt Center for Bone Biology.

"Preventing metastasis is really the goal we want to achieve," he said.

Elefteriou and his colleagues knew from their previous studies that the sympathetic nervous system stimulated bone remodeling, and that it used some of the same signaling molecules that have been implicated in breast cancer metastasis to bone.

"We came to the hypothesis that sympathetic activation might remodel the bone environment and make it more favorable for cancer cells to metastasize there," Elefteriou said.

Evidence from the clinic supported this notion. Breast cancer patients who suffered from stress or depression following their primary treatment had shorter survival times. Both stress and depression activate the sympathetic nervous system.

To explore this possible link, the researchers studied cancer cell metastasis in mice. They followed fluorescently "tagged" human breast cancer cells that were injected into the mouse heart to model the stage of metastasis when breast cancer cells leave the primary site and move through the circulation.
They found that treating the mice with a drug that mimics sympathetic nervous system activation caused more cancer lesions in bone. Using physical restraint to stress the mice and activate the sympathetic nervous system also caused more cancer lesions in bone. Treating the restrained mice with propranolol, one of a family of blood pressure medicines called "beta-blockers," reduced the number of bone lesions.

The investigators demonstrated that sympathetic nervous system activation increases bone levels of a signaling molecule called RANKL, which is known to promote the formation of osteoclasts – bone cells that break down bone tissue. RANKL has also been implicated in cell migration, and Elefteriou and colleagues were able to show that breast cancer cell migration to the bone depends on RANKL.
The findings suggest that beta-blockers or drugs that interfere with RANKL signaling, such as denosumab, may be useful in preventing breast cancer cell metastasis to bone. Propranolol and other beta-blockers are inexpensive, well characterized, and safe in most patients. They may be a good choice for long-term treatment if future studies in patients with breast cancer confirm their ability to block cancer cell metastasis to bone, Elefteriou said.

"If something as simple as a beta blocker could prevent cancer metastasis to bone, this would impact the treatment of millions of patients worldwide," he said.

Efforts to reduce stress and depression in patients with cancer may have unappreciated benefits in terms of metastasis prevention, he added.

http://www.eurekalert.org/pub_releases/2012-07/vumc-sfb071212.php

Sunday, May 6, 2012

New Muscular Dystrophy Treatment Approach Developed Using Human Stem Cells

ScienceDaily (May 4, 2012) — Researchers from the University of Minnesota's Lillehei Heart Institute have effectively treated muscular dystrophy in mice using human stem cells derived from a new process that -- for the first time -- makes the production of human muscle cells from stem cells efficient and effective.

The research, published May 4 in Cell Stem Cell, outlines the strategy for the development of a rapidly dividing population of skeletal myogenic progenitor cells (muscle-forming cells) derived from induced pluripotent (iPS) cells. iPS cells have all of the potential of embryonic stem (ES) cells, but are derived by reprogramming skin cells. They can be patient-specific, which renders them unlikely to be rejected, and do not involve the destruction of embryos.

This is the first time that human stem cells have been shown to be effective in the treatment of muscular dystrophy.

According to U of M researchers -- who were also the first to use ES cells from mice to treat muscular dystrophy -- there has been a significant lag in translating studies using mouse stem cells into therapeutically relevant studies involving human stem cells. This lag has dramatically limited the development of cell therapies or clinical trials for human patients.

The latest research from the U of M provides the proof-of-principle for treating muscular dystrophy with human iPS cells, setting the stage for future human clinical trials.

"One of the biggest barriers to the development of cell-based therapies for neuromuscular disorders like muscular dystrophy has been obtaining sufficient muscle progenitor cells to produce a therapeutically effective response," said principal investigator Rita Perlingeiro, Ph.D., associate professor of medicine in the Medical School's Division of Cardiology. "Up until now, deriving engraftable skeletal muscle stem cells from human pluripotent stem cells hasn't been possible. Our results demonstrate that it is indeed possible and sets the stage for the development of a clinically meaningful treatment approach."

Upon transplantation into mice suffering from muscular dystrophy, human skeletal myogenic progenitor cells provided both extensive and long-term muscle regeneration which resulted in improved muscle function.

To achieve their results, U of M researchers genetically modified two well-characterized human iPS cell lines and an existing human ES cell line with the PAX7 gene. This allowed them to regulate levels of the Pax7 protein, which is essential for the regeneration of skeletal muscle tissue after damage. The researchers found this regulation could prompt naïve ES and iPS cells to differentiate into muscle-forming cells.

Up until this point, researchers had struggled to make muscle efficiently from ES and iPS cells. PAX7 -- induced at exactly the right time -- helped determine the fate of human ES and iPS cells, pushing them into becoming human muscle progenitor cells.

Once Dr. Perlingeiro's team was able to pinpoint the optimal timing of differentiation, the cells were well suited to the regrowth needed to treat conditions such as muscular dystrophy. In fact, Pax7-induced muscle progenitors were far more effective than human myoblasts at improving muscle function. Myoblasts, which are cell cultures derived from adult muscle biopsies, had previously been tested in clinical trials for muscular dystrophy, however the myoblasts did not persist after transplantation.

"Seeing long-term maintenance of these cells without major adverse side effects is exciting," said Perlingeiro. "Our research proves that these differentiated stem cells have real staying power in the fight against muscular dystrophy."

According to John Wagner, M.D., scientific director of clinical research at the University's Stem Cell Institute and blood and marrow transplant expert, "This research is a phenomenal breakthrough. Dr. Perlingeiro and her collaborators have overcome one of the most significant obstacles to moving stem cell therapies into the treatment of children with devastating and life threatening muscular dystrophies."

The U of M researchers say alternative methods of Pax7 induction will need to be investigated before this study can be turned into a human clinical trial. Their method of delivering the Pax7 protein involved genetic modification of cells with viruses and because viruses sometimes cause mutations, they add risk to a clinical trial. But the U of M researchers are committed to developing a safe and effective clinical protocol, and are actively testing alternate methods of delivering Pax7.

Radbod Darabi, Robert W. Arpke, Stefan Irion, John T. Dimos, Marica Grskovic, Michael Kyba, Rita C.R. Perlingeiro. Human ES- and iPS-Derived Myogenic Progenitors Restore DYSTROPHIN and Improve Contractility upon Transplantation in Dystrophic Mice. Cell Stem Cell, 2012; 10 (5): 610 DOI: 10.1016/j.stem.2012.02.015

http://www.sciencedaily.com/releases/2012/05/120504110554.htm?utm_source=feedburner&utm_medium=email&utm_campaign=Feed%3A+sciencedaily+%28ScienceDaily%3A+Latest+Science+News%29

Wednesday, May 2, 2012

Telomeres in Disease

The ends of linear chromosomes have attracted serious scientific study—and Nobel Prizes—since the early 20th century. Called telomeres, these ends serve to protect the coding DNA of the genome. When a cell’s telomeres shorten to critical lengths, the cell senesces. Thus, telomeres dictate a cell’s life span—unless something goes wrong. Work over the past several decades has revealed an active, though limited, mechanism for the normal enzymatic repair of telomere loss in certain proliferative cells. Telomere lengthening in cancer cells, however, confers an abnormal proliferative ability.

In addition to cancer, telomeres have been found to be involved in numerous other diseases, including liver dysfunction and aplastic anemia, a condition in which the bone marrow does not produce a sufficient supply of new blood cells. Inadequate telomere repair and accelerated telomere attrition can be molecular causes of these diseases, and targeting these processes may lead to the development of novel therapies.

Chromosome tails

Telomeres consist of hexameric nucleotide sequences (TTAGGG in humans) that are repeated hundreds to thousands of times at each extremity of each chromosome. Telomeric DNA is coated by a group of proteins, collectively termed shelterin, which serves to protect telomere structure. Because DNA can only be synthesized in one direction, the RNA primers at the chromosome’s ends cannot be filled in, and thus a small amount of DNA is lost with every cell division—a loss that occurs in the telomeres. During normal aging of an animal or in cell culture, cells divide and telomeres shorten. As telomeric sequences do not contain genes, no important genetic information undergoes erosion during DNA replication. When telomeres become critically short, the cell becomes senescent—it ceases to divide—or undergoes apoptosis—it dies.

Telomere attrition explains the “Hayflick limit,” the number of divisions a cell is capable of undergoing in tissue culture before the cell stops dividing. Telomere length is therefore a type of “mitotic clock,” a measure of a cell’s proliferative history. Under circumstances in which cell proliferation continues despite critically short telomeres (usually about a few hundred hexanucleotide repeats), the telomere’s protective function is lost. Subtelomeric genetic information can be lost, and more importantly, recombination between chromosomes occurs, leading to chromosome instability, aneuploidy, and possible transformation to a cancer phenotype.

Some proliferative cells can elongate telomeres enzymatically through the telomerase complex. Telomerase (TERT) is a reverse transcriptase that employs a small RNA molecule (TERC) as a template to extend telomeres in cells. In this way, telomerase counterbalances the effects of cell division and cellular genetic “aging,” preventing senescence, apoptosis, and genetic instability. Telomerase-dependent telomere repair occurs naturally in some cells, such as embryonic and adult stem cells and some cells of the immune system—cell types that divide regularly to support development, maintain tissues, and combat infections, respectively.

Telomere maintenance is also possible by other mechanisms, such as the alternative pathway (ALT), which uses recombination between chromosomes to maintain telomere length. In ALT, telomeres are not newly elongated, but rather transferred from one chromosome to another, resulting in some daughter cells whose chromosomes have shorter telomeres and others with longer telomeres. The details of ALT’s components and regulation, however, are not well understood.

Telomere shortening and cancer

Most cells in which telomeres reach critically short lengths either die or enter senescence. In those few that survive, perhaps due to inadequate monitoring by p53 and related DNA damage-response safeguards, telomere repair would be subject to powerful selective pressure. Indeed, in most malignancies, telomerase gene upregulation or activation of the ALT pathway is thought to be necessary for the establishment of cellular immortality. Telomerase is so frequently increased in tumors and in cancer cell lines as to be considered an appropriate therapeutic target. Currently there are several clinical trials using telomerase inhibitors to treat a variety of cancers, but results have yet to be reported. Telomere shortening would also generate the equivalent of a “mutator phenotype” by increasing spontaneous chromosomal aberrations—from numerical changes to structural abnormalities—and would therefore increase the pool of aberrant cells upon which selection would act.

There are many sources of evidence suggesting that telomere attrition is associated with and likely etiologic of cancer. Patients with dyskeratosis congenita, a rare inherited bone marrow failure disease characterized by telomerase dysfunction, have a 1,000-fold increase in risk of tongue cancer and about a 100 fold increase in risk of acute myeloid leukemia. In aplastic anemia, patients with the shortest telomeres (absent mutations) are 4- to 5-fold more likely to have their disease undergo malignant transformation to myelodysplasia and leukemia. Telomere-free ends of chromosomes and aneuploidy may be apparent in cultured bone marrow years before progression to leukemia. Furthermore, some acute myeloid leukemia patients without prior bone marrow failure have inherited mutations in TERT and TERC.

Similarly, short leukocyte telomeres predict the development of cancer in patients with Barrett’s esophagitis, a condition in which the lining of the esophagus is damaged by stomach acid, or ulcerative colitis, a type of inflammatory bowel disease. In these diseases, the mechanism is less clear. Short telomeres in blood leukocytes may reflect the telomere length of the affected organ. Alternatively, they may be a biomarker of exposure to reactive oxygen species produced as a result of a chronic inflammatory process, which can both damage telomeres and cause cancer. Evidence for the latter hypothesis comes from the observation that cells cultured in room air have excessive telomere shortening in comparison to cells cultured at low oxygen tension.

More generally, genome-wide analyses have identified single nucleotide polymorphisms in TERT as risk factors in many cancers. In a recent report, short leukocyte telomeres were associated with increased risk of all cancers and of cancer fatalities in a large population followed over a decade. Circumstantially, telomere attrition is an accompaniment of aging, itself a major risk factor for cancer. Furthermore, secondary malignancies often occur after chemotherapy and radiation, which would be anticipated to cause marrow stress and telomere shortening. More direct data come from animal experiments. In a knockout mouse model, animals with reduced telomerase activity combined with diminished p53 surveillance of DNA damage developed a variety of epidermal cancers unusual in the rodent but typical of humans.

Other telomere diseases

In addition to cancer, other diseases have been linked to telomeres. Hematopoietic stem cells express telomerase in response to the enormous daily demand for red blood cells, white blood cells, and platelets. Thus while overexpression of telomerase in other tissues can cause malignant growth, faulty telomere repair in blood stem cells can also result in severe diseases caused by stem cell failure.

One such “telomere disease” is dyskeratosis congenita, an X-linked bone marrow disorder characterized by symptoms such as abnormal nails, a pigmented, net-like rash, a white patch or plaque in the mouth, and aplastic anemia. The disease usually presents in the first decade of life. The liver and lungs can also be affected, as is often observed after a hematopoietic stem-cell transplant is performed to correct the bone marrow disease. The reasons for liver and lung involvement are not clear, but the chemotherapy used for transplant conditioning and the new inflammatory cells in the transplanted bone marrow may accelerate the injury in these organs.

Patients suffering from dyskeratosis congenita inherit a mutation in a gene named DKC1, identified by Inderjeet Dokal at the Hammersmith Hospital in London, who performed linkage studies of large pedigrees. DKC1 encodes dyskerin, a protein that binds to the RNA component of the telomerase complex and stabilizes it. Later, heterozygous mutations in TERC were also found in some families with dyskeratosis congenita. The severe phenotype of X-linked dyskeratosis congenita is likely due to the loss of functional DKC1 and markedly reduced telomerase function, which results in defective telomere repair and leads to accelerated telomere attrition, causing cell senescence and organ failure.

Whereas dyskeratosis congenita caused by mutations in the DKC1 gene usually presents during infancy, mutations in TERC and in the enzymatic component encoded by TERT appear to have impacts later in life. The first TERT mutations found in humans were in adult patients with acquired aplastic anemia who lacked physical anomalies and did not have a family history of telomere-related disease. Penetrance of the phenotypes of TERT and TERC mutations is highly variable among and within families, as reflected by the severity, time of onset, and organs involved. Within pedigrees, members with the identical mutation may have minimal or no clinical manifestations, develop aplastic anemia late in life, or suffer pulmonary fibrosis (scarring of the lungs) or hepatic cirrhosis (scarring of liver tissue). Different organ systems may be affected in different family members at different times, and occasional patients have disease in marrow, lung, and liver. How faulty telomere repair leads to such diseases is not fully understood.

Even with the uncertainties that remain, the association of telomerase mutations with disease in such disparate organs systems has important practical consequences for patients and their physicians. In the family history, the presence of even mild blood count abnormalities, pulmonary fibrosis, and hepatic cirrhosis, as well as acute myeloid leukemia, are important clues for the diagnosis of a telomeropathy. Involvement of multiple medical subspecialties can be confusing; some patients have even made their own diagnoses after Internet searches. Telomere length in leukocytes can be measured commercially and is a reliable marker of these diseases when severely reduced. Sequencing TERT and TERC can also be diagnostic. The appropriate finding of a telomerase deficit has consequences for prognosis, treatment, and genetic counseling. But while the diagnosis of telomeropathies can be straightforward, there may be complications. Some typical families lack known mutations, and telomere length may be normal even in the presence of etiologic nucleotide substitutions. Furthermore, rare mutations in shelterin genes coding for the proteins that protect telomere structure can produce severe dyskeratosis but do not alter telomerase repair capacity. And regulatory regions of genes, not routinely screened, may be responsible in some cases.

Telomeres and aging

Telomeres shorten as we age. By analogy to the cellular mitotic clock, telomeres have been postulated as a marker of “genetic age,” and telomere length has been marketed as a simple predictor of longevity. Assays of telomere length have been bundled with recommendations for lifestyle modification and for drug therapy, neither based on appropriate clinical studies. Simple but appealing arguments relating telomeres and aging are currently controversial, likely simplistic, and potentially harmful. Telomere length does indeed reflect a cell’s past proliferative history and future propensity for apoptosis, senescence, and transformation. Cellular aging, however, is not equivalent to organ or organismal aging.

There are several considerations in relating telomere biology to aging. First, physiologically there is overlap between the shortest telomere length of young children and the longest telomeres of the elderly. Most telomere shortening occurs early in life, in association with growth, and when the rate of disease in general is low. The paradigmatic telomere syndrome of dyskeratosis congenita is not at all typical of progerias, inherited syndromes in which patients appear old and suffer diseases of aging such as premature atherosclerosis or dementia. Furthermore, the organ damage of dyskeratosis congenita is not very similar to normal aging of marrow, lungs, and liver. The marrow becomes mildly hypocellular in older individuals, but stem cell numbers may actually increase, and blood counts remain stable; and neither the liver nor lungs normally become fibrotic with advanced age, as they often do in dyskeratosis congenita patients. Although in adults, relatively short leukocyte telomeres have been associated with cardiovascular events—a common morbidity of the aging population—the clinical correlations have not been consistent, and may be related to overall reactive oxygen species exposure.

Studies in humans have attempted to relate telomere length to life span. In the provocative initial publication from the University of Utah in 2003, individuals around 60 years of age who had the longest telomeres lived longer than did subjects with the shortest telomeres, but the main cause of death in the latter group was, inexplicably, infectious disease; the persons with shorter telomeres did not have a higher rate of cancer deaths. Moreover, these findings have not been confirmed in other studies of older subjects. In another study evaluating a different population, telomere length failed to predict survival, but interestingly it correlated with years of healthy life. In a Danish study of people aged 73 to 101 years, telomeres correlated with life expectancy in a simple univariate analysis, but only before the researchers corrected for age, suggesting that the correlation was driven simply by the fact that younger subjects had longer telomeres. And a Dutch study of 78-year-old men found that while telomere lengths eroded with age, they failed to correlate with mortality. These discrepancies may have several causes. In some analyses, telomere lengths may have been studied as a surrogate marker of age. In addition, retrospective studies may identify “positive” associations that are random and cannot be reproduced in follow-up investigations.

The telomere hypothesis of aging also has been modeled in mice. For instance, in a murine model of telomerase deficiency and accelerated telomere attrition, researchers found that certain intracellular pathways involved in mitochondrial function and glucose metabolism were deregulated, a common occurrence in aging individuals, ultimately causing heart muscle disease. Interestingly, telomerase reactivation in these mice restored glucose production and heart function. However, these abnormalities observed in telomerase-deficient animals were not those typical of humans with very short telomeres; patients with telomeropathies usually do not suffer from heart disease. Indeed, the translation of mouse experiments on telomeres to human physiology and disease should be done with caution. Mice are not the ideal model for telomere attrition and its effects on aging as murine telomeres are 5 to 10 times longer than human telomeres, in spite of mice having a much shorter life span. When telomerase is knocked out in mice, they live a healthy life for several generations, and even late-generation animals with very short telomeres do not display the clinical phenotypes characteristic of human telomeropathies. Telomerase-deficient mice also do not have a higher incidence of cancer, which happens only if the p53 gene also is modulated, in contrast to humans with telomerase deficiency, who are at very high risk of developing cancer.

Implications for medicine

Telomeres and their repair are important in the growing field of regenerative medicine. Dolly the sheep had chromosomes with shorter telomeres probably because she was cloned from an adult mammary gland cell. This may have contributed to Dolly’s illnesses, especially her progressive lung disease. Embryonic stem cells, however, express telomerase and are able to maintain their telomere lengths despite numerous cell divisions. More recently, reprogramming mature adult skin cells to the pluripotent state has been achieved with introduction of just a few defined nuclear factors. During the process of reverting cells to a more immature and pluripotent state, the reprogrammed cells’ telomeres are highly elongated. In the first steps of reprogramming and likely in the early stages of embryogenesis, cells can elongate, and thus “rejuvenate,” their telomeres. Since telomere shortening limits cell proliferation, mechanisms that can elongate telomeres are highly desirable for effective regenerative medicine.

Telomerase expression is tightly regulated in the cell; just a few copies of the complex are present in the cell nucleus, and they exert their function during certain specific periods of the cell cycle. The mechanisms that modulate telomerase gene expression, resultant enzymatic activity, and telomere elongation are the focus of intensive research. MYC, a proto-oncogene that regulates the expression of many genes and cell pluripotency, activates telomerase expression. Sex hormones also activate telomerase expression in reproductive and nonreproductive organs, such as the bone marrow. The promoter region of the telomerase gene contains regulatory sequences that are modulated by estrogen; cells exposed to estrogens (or androgens converted into estrogens) upregulate telomerase expression. In retrospect, the clinical response of improved blood cell counts in patients with aplastic anemia, especially children with inherited marrow failure, to androgens may be attributable to this mechanism. However, whether higher blood levels of sex hormones or exposure to exogenous sex hormones causes telomere elongation is still unknown.

Conclusions

Telomeres and telomere repair are basic molecular processes in cells possessing linear DNA chromosomes. Accelerated telomere attrition due to genetic defects in telomerase and in the shelterin protein genes is etiologic in several human diseases not previously considered related in the clinic. These include aplastic anemia, pulmonary fibrosis, and hepatic cirrhosis. The telomeropathies, especially in their milder and more chronic forms, may not be rare and almost certainly are often unrecognized by physicians. The importance of telomere repair in tissues under regenerative stress is of special interest, particularly in the reactive responses of fibrogenesis in the liver and the lungs. The maintenance of adequate telomere lengths also may be important in embryonic and adult stem cells to enable proliferation while preventing chromosome instability, thus avoiding potential malignant transformation. Also of interest is the connection linking telomere attrition and chronic inflammation to cancer and other diseases. (See “An Aspirin for Your Cancer,” The Scientist, April 2011.)

There is still much to be learned about how telomerase gene mutations cause disease, why they only affect certain organs, and how telomeres can be targeted for therapies. Both the genetic regulation of telomerase expression and the effect of an organism’s environment on telomere attrition are poorly understood. Drugs or hormones that might modulate telomerase expression and maintain or elongate telomeres would be appealing in the treatment of the telomeropathies and in conditions in which telomere attrition has known medical consequences. Whether telomere shortening mediates human aging—and conversely, whether telomere elongation may reverse aging or prevent age-related diseases—are still controversial issues.

Rodrigo Calado is an Associate Professor of Medicine at the University of São Paulo. Neal Young is the Chief of the Hematology Branch at the US National Institutes of Health.

This article is adapted from a review in F1000 Medicine Reports, DOI:10.3410/M4-8 (open access).

http://the-scientist.com/2012/05/01/telomeres-in-disease/


Tuesday, May 1, 2012

Crystal Structure of Human Argonaute2

Scripps Research Institute Scientists Find the Structure of a Key Gene Silencer Protein
The Structure reveals potential therapeutic targets in area with untapped potential

LA JOLLA, CA, April 26, 2012:-
Scientists at The Scripps Research Institute have determined the three-dimensional atomic structure of a human protein that is centrally involved in regulating the activities of cells. Knowing the precise structure of this protein paves the way for scientists to understand a process known as RNA-Silencing and to harness it to treat diseases.

"Biologists have known about RNA-Silencing for only a decade or so, but it's already clear that there's an enormous untapped potential here for new therapies," said Ian MacRae, an assistant professor at Scripps Research and senior author of the new report.

The new report, which appeared on April 26,2012 in the Journal Science's advance online publication, Science Express, focuses on Argonaute2. This protein can effectively silence a gene by intercepting and slicing the gene's RNA transcripts before they are translated into working proteins.


Interception and Destruction of Messages

When a gene that code for a protein is active in a cell, its information is transcribed from DNA form into lengths of nucleic acid called messenger RNA. If all goes well, these coded mRNA signals make their way to the cell's protein factories, which use them as templates to synthesize new proteins. RNA-silencing, also called RNA-Interference is the interception and destruction of these messages and as such, is a powerful and specific regulator of cell activity, as well as a strong defender against viral genes.

The silencing process requires not only an Argonaute protein but also a small length of guide RNA, known as a short-interfering RNA or microRNA. The guide RNA fits into a slot on Argonaute and serves as a target recognition device. Like a coded strip of Velero(TM), it latches onto a specific mRNA target whose sequence is the chemical mirror image of its own- thus bringing Argonaute into contact with its doomed prey.

Argonaute2 is not the only type of human Argonaute protein, but it seems to be the only one capable fo destroying target RNA directly. "If the guide RNA is completely complementary to the target RNA, Argonaute2 will cleave the mRNA, and that will elicit the degradation of the fragments and the loss of the genetic message," said Nicole Schirle, the graduate student in MacRae's laboratory who was lead author of the paper.

Aimed at disease-causing genes or even a cell's own overactive guide RNAs, RNA-silencing could be a powerful therapeutic weapon. In principle, one needs only to inject target-specific guide RNAs, and these will link up with Argonaute protein in cells to find and destroy the target RNAs. Scientists have managed to do this successfully with relatively accessible target cells, such as in the eye. But they have found it difficult to develop guide RNAs that can get from the bloodstream into distant tissues and still function.

"You have to modify the guide RNA, in some way to get it through the blood and into cells, but as soon as you start modifying it, you disrupt its ability to interact with Argonaute,"said MacRae. Knowing the precise structure of Argonaute should enable researchers to clear this hurdle by designing better guide RNA.

More Points for Manipulation

Previous structural studies have focused mostly on Argonaute protein from bacteria and other lower organisms, which have key differences from their human counterparts. Schirle was able to produce the comparatively large and complex human Argonaute2 and to manipulate it into forming crystals for X-ray crystallography analysis a feat that structural biologists have wanted to achieve for much of the past decade. "It was just excellent and diligent crystallography on her part," said MacRae.

The team's analysis of Argonaute2's structure revealed that it has the same basic set of working parts as bacterial Argonaute proteins, except that they are arranged somewhat differently. Also, key parts of Argonaute2 have extra loops and other structures, not seen on bacterial versions, which may play roles in binding to guide RNA. Finally, Argonatue2 has what appear to be binding sites for additional co-factor proteins that are thought to perform other destructive operations on the target mRNA.

"Basically, this Argonaute protein is more sophisticated than its bacterial cousins; it has more bells and whistles, which give us more points for manipulation. With this structure solved, we no longer need to use the prokaryotic structures to guess at what human Argonaute protein look like," MacRae said.

He and Schirle and others in the lab now are analyzing the functions of Argonaute2's substructures, as well as looking for ways to design better therapeutic guide RNAs.

"Now with the structural data, we can see what synthetic guide RNAs will work with Argonaute and what won't, " MacRae said, "we might even be able to make guide RNAs that can outcompete natural ones."

The research that led to Schirle and MacRae's new paper, "The Crystal Structure of Human Argonaute2," was funded by the National Institute of General Medical Sciences, part of the National Institute of Health.

press@scripps.edu