Fighting antibiotic resistance with 'molecular drill bits'

PUBLIC RELEASE DATE:

17-Mar-2014

Contact: Michael Bernstein 214-853-8005 (Dallas Press Center, March 14-19) 202-872-6042 m_bernstein@acs.org

Contact: Katie Cottingham, Ph.D. 214-853-8005 (Dallas Press Center, March 14-19) 301-775-8455 k_cottingham@acs.org

American Chemical Society

DALLAS, March 17, 2014 In response to drug-resistant "superbugs" that send millions of people to hospitals around the world, scientists are building tiny, "molecular drill bits" that kill bacteria by bursting through their protective cell walls. They presented some of the latest developments on these drill bits, better known to scientists as antimicrobial peptides (AMPs), at the 247th National Meeting & Exposition of the American Chemical Society (ACS), the world's largest scientific society.

The meeting, which features more than 10,000 scientific reports across disciplines from energy to medicine, continues here through Thursday.

One of the researchers in the search for new ways to beat pathogenic bacteria is Georges Belfort, Ph.D. He and his team have been searching for a new therapy against the bacteria that cause tuberculosis (TB). It's a well-known, treatable disease, but resistant strains are cropping up. The World Health Organization estimates that about 170,000 people died from multidrug-resistant TB in 2012.

"If the bacteria build resistance to all current treatments, you're dead in the water," said Belfort, who is at Rensselaer Polytechnic Institute.

To avoid this dire scenario, scientists are developing creative ways to battle the disease. In ongoing research, Belfort's group together with his wife, Marlene Belfort, and her group at the University at Albany are trying to dismantle bacteria from within. They also decided to attack it from the outside.

Read more:

Fighting antibiotic resistance with 'molecular drill bits'

Penn Medicine Team to Examine Cognitive Impact of Space Flight as Part of NASA's Unprecedented Twin Astronaut Study

PHILADELPHIA A team of researchers from the Perelman School of Medicine at the University of Pennsylvania will take part in a first-of-its-kind investigation by the National Aeronautics and Space Administration (NASA) into the molecular, physiological and psychological effects of spaceflight on the human body by comparing identical twins. The unique opportunity is made possible by NASA's decision to fly veteran astronaut Scott Kelly aboard the International Space Station for one year, beginning March 2015, while his identical twin brother, retired astronaut Mark Kelly, remains on Earth. The research is part of NASAs continuous effort to reduce the health impacts of human space exploration.

Penns research team includes principal investigator Mathias Basner, MD, PhD, MSc, assistant professor of Sleep and Chronobiology, Department of Psychiatry, David F. Dinges, PhD, professor and chief, Division of Sleep and Chronobiology, Department of Psychiatry, and Ruben C. Gur, PhD, professor of Psychology, Director of Neuropsychology, the Brain Behavior Laboratory, and the Center for Neuroimaging in Psychiatry.

Their research will focus on psychosocial and neurobehavioral differences between the Kelly brothers including attention, spatial orientation, emotion recognition, and risk decision making, as a result of the spaceflight environment, which includes confinement, weightlessness, stress, and space radiation. "This is a unique opportunity to substantially increase our knowledge of the effects of prolonged exposure to the space flight environment on human physiology and cognition which will help us to better plan for a human mission to Mars, Basner said.

Using twins for the study will allow researchers to examine more subtle changes caused by spaceflight than previously understood. Since the twins have essentially almost identical DNA and DNA controls the biomolecular workings of the body any difference are likely due to spaceflight and not because the two subjects are genetically distinct.

In addition to the work by the Penn team, NASA's Human Research Program (HRP) will fund nine other studies of the Kelly brothers designed to better understand the effects of microgravity on the human body at the molecular level. These studies will look at the way genes in the cells are turned on and off as a result of spaceflight; and how stressors like radiation, confinement and microgravity prompt changes in the proteins and metabolites gathered in biological samples like blood, saliva, urine and stool. The National Space Biomedical Research Institute is partnering with HRP to provide genetic counseling and assisting in the management of the research.

In order to launch the new twin study, scientific and technical experts from academia and government reviewed 40 proposals submitted in response to NASAs research announcement "Human Exploration Research Opportunities - Differential Effects on Homozygous Twin Astronauts Associated with Differences in Exposure to Spaceflight Factors." The 10 selected proposals, which are from 10 institutions in seven states, will receive a combined $1.5 million during a three-year period.

For more information on the study, please visit the NASA website.

Original post:

Penn Medicine Team to Examine Cognitive Impact of Space Flight as Part of NASA's Unprecedented Twin Astronaut Study

Protein Key to Cell Motility Has Implications for Stopping Cancer Metastasis

Contact Information

Available for logged-in reporters only

Newswise PHILADELPHIA - Cell movement is the basic recipe of life, and all cells have the capacity to move, says Roberto Dominguez, PhD, professor of Physiology at the Perelman School of Medicine, University of Pennsylvania. Motility albeit on a cellular spatial scale -- is necessary for wound healing, clotting, fetal development, nerve connections, and the immune response, among other functions. On the other hand, cell movement can be deleterious when cancer cells break away from tumors and migrate to set up shop in other tissues during cancer metastasis.

The Dominguez team, with postdoctoral fellow David Kast, PhD, and colleagues, report online ahead of print in Nature Structural & Molecular Biology how a key cell-movement protein called IRSp53 is regulated in a resting and active state, and what this means for cancer-cell metastasis.

We characterized how IRSp53 connects to the cell-motility machinery, says Kast. It does this by starting the formation of cell filopodia - extensions that form when a cell needs to move.

Cells move like an inchworm, explains Dominguez. Filopodia are at the leading edge of moving cells. The trailing end of the cell follows the move forward through contraction of actin and myosin in the cytoskeleton, much like muscle contraction. A cell pushes out the leading edge of its membrane, and sticks it down on whatever it is moving across, namely other cells, and then moves the cell body along, unsticking the back end. This sets the cell up for its next move.

IRSp53 contains a region called a BAR domain that binds to and shapes cell membranes. Other parts of the protein connect it to the cytoskeleton (internal bits that give a cell structure and shape). Together, through the binding of cell membranes and other proteins IRSp53 regulates cell movement. The team found that in the resting state, human IRSp53 adopts a closed shape that prevents it from interacting with the membrane and the cytoskeleton. However, the binding of a signaling protein, called Cdc42, opens IRSp53, setting in motion the recruitment of a complex cellular machinery needed for motility.

One of the cytoskeleton components IRSp53 connects to is the tumor-promoting protein Eps8. IRSp53 is synergistically activated by the combined action of Cdc42 and binding of Eps8, which is upregulated in metastatic cancers.

Co-authors Tatyana Svitkina and Changsong Yang from the Penn Department of Biology, brought their expertise with living cells to the study. By introducing normal and mutant proteins into cells they could see how these proteins induced filopodia to form. The team found that mutations in critical regions of IRSp53 can either lead to enhanced or reduced filopodia formation and, as a consequence, cell motility. This finding shows how all these different proteins converge on IRSp53 to execute precise cellular functions, and that when one factor is disrupted, other proteins are affected down the activity pathway, says Dominguez.

The teams next steps will be to screen libraries of small molecule inhibitors that interfere with the IRSp53-Eps8 interaction, to figure out how to stop unwanted cell movement before it gets too far.

View post:

Protein Key to Cell Motility Has Implications for Stopping Cancer Metastasis

Challenges in Bioethics and Policy Development at the Frontiers of Science and Medicine – Video


Challenges in Bioethics and Policy Development at the Frontiers of Science and Medicine
Visit: http://seminars.uctv.tv/) Larry Goldstein,UCSD professor of cellular and molecular medicine, a Howard Hughes Medical Institute investigator and a lea...

By: UCTVSeminars

More here:

Challenges in Bioethics and Policy Development at the Frontiers of Science and Medicine - Video

Molecular Subtyping of Breast Cancer Can Better Identify Women at High Risk of Disease Recurrence

Contact Information

Available for logged-in reporters only

Newswise A method called molecular subtyping can help doctors better determine which of their breast cancer patients are at high risk of getting breast cancer again, a new study led by the University of South Florida reports. This sophisticated genetic profiling of an individuals specific tumor offers an additional resource to help identify patients who would most benefit from chemotherapy and those who would not.

The findings by researchers from USF and other institutions were presented in a scientific poster at the Miami Breast Cancer Conference, held March 6-9 in Miami Beach, Fla.

The most important takeaway for our colleagues in breast cancer diagnosis and treatment is the potential value of molecular subtyping to personalize and improve each womans treatment, said principal investigator Charles E. Cox, MD, McCann Foundation Endowed Professor of Breast Surgery, USF Health Morsani College of Medicine.

Molecular subtyping is a way of classifying breast cancer tumors into one of four genetically-distinct categories, or subtypes: Luminal A, Luminal B, Basal (a subset of triple negative), and HER2-type. Each subtype responds differently to different kinds of treatments, and some subtypes indicate a higher risk of disease recurrence.

Our data showed that a substantial number of breast cancer patients -- classified as low risk by one particular genomic test -- turn out to be at high risk of recurrence once we determined their subtype, Dr. Cox said. These are mostly Luminal B patients, and their physicians might not fully understand their patients situation unless they do subtyping.

The USF study examined why different genomic tests for breast cancer sometimes provide contradictory information about risk of recurrence. The key findings involved the 70-gene MammaPrint test; the 21-gene Oncotype DX test, which is an earlier commercially available test; and Mammostrat, a gene profiling test performed on slides of the breast tumor by a pathologist. The tests have generally been assumed to provide equivalent information about recurrence risk, but that is proving not to be the case.

Researchers examined tumor samples from a total of 148 patients. The greatest discordance (lack of agreement) about risk of disease recurrence occurred in a group of 51 patients. Of those 51, all were stratified by MammaPrint as high risk of recurrence, while Oncotype classified 18 of them (35 percent) as low risk.

BluePrint, an 80-gene test to identify a tumors molecular subtype, was also used for those stratified by MammaPrint. This process revealed that the 51 patients were Luminal B, a molecular subtype with a high risk of recurrence.

More here:

Molecular Subtyping of Breast Cancer Can Better Identify Women at High Risk of Disease Recurrence

Short people may be short on brains, study says

A study by Edinburgh University's Institute of Genetics and Molecular Medicine sees a "small" link between height and intelligence.

The long and short of intelligence?

I am not one to cast aspersions -- either up or down.

I had never thought that size meant anything, nor that height made any difference to people, other than altering their visual perspective.

Recently, though, an Oxford University study intimated that short people might be low on self-esteem. They tend, allegedly, not to have high opinions of themselves. Worse, there are many social problems that many short people feel the need to overcome. (See video below.)

Now along trots another British study that suggests the vertically challenged might be short on intellect. As Britain's Sunday Times (behind a tall paywall) reports, short people may tend to have smaller IQs.

Researchers at Edinburgh University's Institute of Genetics and Molecular Medicine analyzed the DNA of more than 6,800 people, none of whom was related to any other, to "estimate the genetic correlation between height and general intelligence." The end goal of the study, the first of its kind, was to explore whether this genetic link may determine other health outcomes.

The Institute's Riccardo Marioni told the Sunday Times: "What we found was a small association between height and intelligence such that people who are taller tend to be smarter."

You have to admire his use of the word "small." There is nothing more responsible than an academic who refuses to be alarmist.

In addition to describing the relationship as "small," though, Marioni let it slip that it was "statistically significant." He also said that the research indicated that 70 percent of short people's smaller IQs could be attributed to genetic factors.

Read the original:

Short people may be short on brains, study says

Mount Sinai study points to new biological mechanisms, treatment paradigm for kidney disease

PUBLIC RELEASE DATE:

3-Mar-2014

Contact: Sid Dinsay laura.newman@mountsinai.org 212-241-9200 The Mount Sinai Hospital / Mount Sinai School of Medicine

New York, NY Prevention and reversal of chronic kidney disease is an urgent public health need. The disease affects 1 in 10 Americans, is debilitating and deadly, and existing drugs, at best, offer only mild delay in progression to end-stage kidney failure. New research led by Icahn School of Medicine at Mount Sinai investigators has uncovered abnormal molecular signaling pathways from disease initiation to irreversible kidney damage, kidney failure, and death. Results from their preclinical and human research are published online March 3 in the Journal of Clinical Investigation.

"Our group is the first to show that endothelial mitochondrial oxidative stress [damage to blood vessel lining that affects the energy-producing part of the cell caused by oxidative stress] regulates the passage of proteins from blood to urine and filtration of waste products in the kidney," said Erwin Bottinger, MD, Director of the Charles Bronfman Institute for Personalized Medicine, and the study's senior author. Specifically, the researchers found albuminuria (protein in the urine) and depletion of the cells that form the kidney's glomerular filtration barrier. "These findings were unexpected and open the door for developing new therapeutic targets," Dr. Bottinger added.

In the preclinical part of the research, investigators used a mouse model to induce scarring in the filtration part of the kidney, or glomeruli. This allowed progressive amounts of protein to pass into the urine and interfered with the clearance of waste products by the kidney. Essentially, the researchers were examining how different signaling mechanism and cellular interactions work, and how when they are disturbed, they promote chronic kidney disease.

Initially, key cells of the glomerular filtration barrier, also called podocytes, cause alterations in endothelin-1, a vasoconstrictor, activating the endothelin receptor A. The activated endothelin receptor A triggered disturbances manifested as endothelial mitochondrial oxidative stress.

The research team was able to confirm that this worked the same way in humans. They studied kidney biopsies, comparing ten biopsies with glomerular sclerosis with six controls. Like in the animal models, the researchers confirmed activated endothelin receptor A and endothelial mitochondrial dysfunction in human glomerular sclerosis biopsies, but not in controls.

"These processes were absolutely essential in causing protein in the urine [or albuminuria], injured podocytes (tiny ball-shaped structures that constrict the blood vessels in the filtering part of the kidney), and cause scarring, all of which can ultimately lead to long-term, irreversible kidney disease. "This is called crosstalk and it is poorly understood," said Ilse S. Daehn, PhD, the study's lead researcher, and Assistant Professor of Medicine in the Division of Nephrology, at the Icahn School of Medicine at Mount Sinai. "We hope that these novel crosstalk findings lead to new therapies that help reverse or arrest chronic kidney disease, which affect millions of Americans," added Dr. Daehn.

Antioxidants that target the mitochondria and endothelin antagonists would alter the paradigm for preventing cell depletion and scarring of the filtration part of the kidney. "There is a pressing unmet medical need to prevent or reverse chronic kidney disease," Dr. Bottinger stressed. "The renin angiotensin inhibitors and angiotensin receptor blockers that are now widely used have not been proven effective in preventing end stage kidney failure. We need more effective drugs to treat the millions of Americans suffering from chronic kidney disease with the goal to eliminate its progression to end- stage kidney failure and with it the need for chronic dialysis and kidney transplantation."

See the article here:

Mount Sinai study points to new biological mechanisms, treatment paradigm for kidney disease

Molecular Diagnostics Market Dominated by Top Five Companies: Kalorama Information Report

New York, NY (PRWEB) March 03, 2014

Just five companies own the majority of the molecular diagnostics market as of this month, according to Kalorama Information. The healthcare market research publisher estimated Roche, Hologic, Qiagen, Becton Dickinson and Abbott alone earned 60% of revenues in the 5.5 billion molecular diagnostics market in 2013. The finding was made in Kaloramas Molecular Diagnostics Six Month Update, the latest look at this fast-growing IVD test segment.

Kalorama thinks top-tier companies will continue to dominate due to their global reach and multi-segment participation. Further, as IVD markets in N. America and Europe offer little growth opportunities for the vast number of tests, the top tier has invested in molecular and personalized medicine testing and also has reinforced international connections.

The report notes that while the names of the players dominating molecular testing have not changed extensively in recent years, their revenue growth rates have. High growth has shifted to innovators, while leaders are seeing flatter revenues.

Roche holds the number one market position but growth is slowing, said Shara Rosen, author of the report. Roche Diagnostics once earned close to double digit revenue growth in its molecular test business. This has slowed in the last two years as the company is conducting a major reorganization and reported that it will phase out its 454 Life Science unit by 2016.

The report said that IVD veterans Becton Dickinson, Gen-Probe (now part of Hologic), Abbott Diagnostics and Siemens Healthcare hold their own in a highly competitive environment, while newcomers Meridian Biosciences and GenMark have made gains with user-friendly infectious disease tests. Meanwhile Pharma specialist Novartis has bailed out of the blood screening business to concentrate on other business units.

The report says that molecular diagnostics is becoming a go-to resource in clinical practice and is the faster-growing part of the diagnostics market at the present time. Tests are being developed constantly, and changes in In light of the amount of activity in test development and regulatory and reimbursement challenges in 2013, Kalorama Information presents this updated review of molecular diagnostics with an emphasis on the last six months of 2013: July December. This report highlights trends and developments that portent the evolution of molecular tests and technologies. The ensemble of consumer demand, technological advances and market forces create a dynamic, energetic and fast-moving environment for new tests and companies.

Market analysis in the Molecular Diagnostics Six Month Update covers world markets for in vitro diagnostics, with a bias towards the developed areas of the globe -- North America and Western Europe. Segment markets in molecular diagnostics are broken out, and trends and partnerships are given extensive discussion. The Molecular Diagnostics Six Month Update is available at http://www.kaloramainformation.com/Molecular-Diagnostics-Month-7969379/.

About Kalorama Information Kalorama Information, a division of MarketResearch.com, supplies the latest in independent medical market research in diagnostics, biotech, pharmaceuticals, medical devices and healthcare; as well as a full range of custom research services. We routinely assist the media with healthcare topics. Follow us on Twitter, LinkedIn and our blog.

Originally posted here:

Molecular Diagnostics Market Dominated by Top Five Companies: Kalorama Information Report

In first moments of infection, a division and a decision

PUBLIC RELEASE DATE:

2-Mar-2014

Contact: Scott LaFee slafee@ucsd.edu 619-543-6163 University of California - San Diego

Using technologies and computational modeling that trace the destiny of single cells, researchers at the University of California, San Diego School of Medicine describe for the first time the earliest stages of fate determination among white blood cells called T lymphocytes, providing new insights that may help drug developers create more effective, longer-lasting vaccines against microbial pathogens or cancer.

The findings are published in the March 2, 2014 online issue of Nature Immunology.

Nave T lymphocytes patrol the front lines of the human bodys defense against infection, circulating in blood and tissues, searching for invasive microbes and other foreign antigens. Theyre called nave because they have not yet encountered an invader. When they do, these T cells activate and divide, giving rise to two types of daughter cells: effector lymphocytes responsible for immediate host defense and memory lymphocytes that provide long-term protection from similar infections.

Researchers have been trying for a very long time to understand when and how T lymphocytes give rise to effector and memory cells during an infection, said John T. Chang, MD, assistant professor in the Department of Medicine and the studys co-principal investigator, along with Gene W. Yeo, PhD, assistant professor in the Department of Cellular and Molecular Medicine and Institute for Genomic Medicine.

However, all studies up to this point were based on analyses on bulk populations of cells, making it impossible to understand fate decisions made by individual cells. First authors Janilyn Arsenio, a postdoctoral fellow in the Chang lab and Boyko Kakaradov, a graduate student in the Yeo lab and UCSD Bioinformatics graduate program said that they took advantage of recent technological advances in single-cell gene expression profiling and cutting-edge machine-learning algorithms to address this question on a level of detail that was not previously possible.

Chang, Yeo and colleagues discovered that the decision by an individual T cell to produce effector and memory cells is made almost at the moment of infection. The mother lymphocyte seems to divide into two daughter cells that are already different from birth, said Chang, with one becoming an effector cell while its sister becomes a memory cell.

Chang noted that the primary purpose of vaccines is to produce strong and durable immune protection, which depends heavily upon generation of memory lymphocytes. Our work suggests that the way T lymphocytes divide early during a microbial infection might be critical to whether or not they give rise to long-lived memory cells. Strategies that improve this process could potentially enhance durable immunity and help us to design more effective vaccines.

View post:

In first moments of infection, a division and a decision

The data cure: The changing science of biology and its impact on your health care

Molecular biologist and science policy leader Professor Keith Yamamoto discusses the current revolution in biological sciences and the emerging field of precision medicine.

SHANE HUNTINGTON Im Dr Shane Huntington, thanks for joining us. Scientists have long laboured to understand life and its complex processes. Their work through the centuries has brought us all enormous benefit, from the development of drugs to treat once incurable diseases, to an increasingly sophisticated understanding of the ecological impact of human activities on the planet. As we expand our investigations into data rich fields like genomics and personalised healthcare, biology is becoming a field populated not just by biologists but also by mathematicians, physicists and statisticians. Biology as a result is changing. Naturally these changes bring the promise of better healthcare standards for people with a cancer or other life threatening conditions, as well as preventative measures to keep people well in the first place. But are we really prepared for this revolution in biology? Do we need to adjust their educational models to make sure we equip health researchers with the right skills? How will this affect the type of healthcare systems we'll have in the coming decades? To answer these questions and discuss the broader implications of a new view of biology, we are joined on Up Close by molecular biologist Professor Keith Yamamoto, Vice Chancellor for Research, Executive Vice Dean of the School of Medicine and Professor of Cellular and Molecular Pharmacology at the University of California, San Francisco. Keith is in Melbourne as a guest of the ICT For Life Sciences forum. Welcome to Up Close Keith.

KEITH YAMAMOTO Thank you, it's nice to be here.

SHANE HUNTINGTON The field of biology has changed dramatically since you began your research career in the 1970s. Can you give us an idea of the main differences in how biology is practised now compared to back in those days?

KEITH YAMAMOTO We're at a very interesting time in biology and in science in general. Biological research at the time that I started in the 1970s as you said, was very much a descriptive field. We'd look through microscopes and examined cells, took pictures of them, did experiments in biochemistry where we would break down specific cellular components and look for the presence and absence of a signal. That descriptive period was a wonderful one. Ones where we gained immeasurably in our understanding of the components that are players in biological processes and understanding the framework - an outline - of the way that those biological processes work. Today things have changed a lot because we have realised that if we're going to move forward from collection of information, naming the players that are involved in the play to actually understanding those processes. Understanding them in ways that we can intercept them or modify them then we have to become a quantitative field. We have to understand things in numerical detail. And to do that biology needs to invite into the field - and it's doing this progressively successfully - scientists who practice their work in a different way. People who are doing physics and chemistry and math and computation and engineering, who'd bring a different way of thinking about problems, as well as working on them. So that's the transition; it's a remarkable one that we're just in the midst of right now.

SHANE HUNTINGTON Now I have to dig a little bit there when you refer to biology as a quantitative field in the current day and in the future, how do you define that relative to what it's done in the past? Certainly I think a lot of biologists would assume they were doing a quantitative version of a research.

KEITH YAMAMOTO Right. So we were able to infer biological processes and even the ways that those processes and even the ways that those processes worked with descriptive means, looking in a microscope to look at the change and the shape of a cell for example. Or the cells that a particular cell would choose to interact with; maybe even merge with and fuse with. Those kinds of descriptions carried us a long way in making theories about exactly how those processes worked. But they don't actually tell us how the processes work. So now the next step is to understand those processes using quantitative methods of engineering and chemistry and physics that will bring us the real numbers behind those observations. It's those numbers that turn out to give us the mechanistic detail to be able to carry forward. The real test of understanding something in a sense is being able to reproduce it yourself - by putting the pieces together and the steps together - the [imagines] working. So we're getting an outline of the players but don't know how to put them together well enough. We don't know whether when we put together a reaction in a descriptive mode whether when it looks like it's working whether it's working the same way that it works in the cell. But getting the numbers behind it all will tell us that. That level of understanding is crucial for doing some of the things that you talked about in your introductory statement where we have the chance to be able to understand them well enough to be able to intercept disease mechanisms and things of that sort.

SHANE HUNTINGTON This presumably will mean that we have to look at our education models - especially at university level - for training biologists. Is the current version adequate to deal with this new biology that you speak of? Or do we have to go back to the drawing board and start redescribing the way in which a biologist will go about their day?

KEITH YAMAMOTO I think we have to go back to the drawing board. But it's going back to the drawing board in I think exciting ways that are going to extend further back from the graduate period of training into undergraduate and even earlier and that is finding a common language for all of these different scientists to speak. The work has gone forward in ways that have taken us to more and more hyper-specialisation. So there are biologists who speak different languages and really can't communicate well with each other. You can imagine what happens when we begin to try to interact with engineers and physicists. So we're at a stage where finding that common language will have a huge payoff; it's going to be very exciting. And we can begin doing that early on. One of the things that we're doing in the University of California, San Francisco UCSF where I work is to begin bringing our first year graduate students together in teams in which the team members - four or five people - come with different backgrounds. Some have been training in physics, some have been training in molecular biology, some have been training in computer science. Bringing them together in teams and then having them to go through a series of so-called boot camp courses - very short intensive courses - intended to bring everyone up to a common level of literacy. And they see immediately the different languages, but somebody on the team understands the language and other people don't and they begin interacting with each other and teaching each other right away. You can see that that can be done any time, it doesn't have to wait until graduate school. So we think that that kind of model can actually get us to where we need to go, not only painlessly but in a way that's fun and interesting.

SHANE HUNTINGTON In that model you're not just talking about retraining the language skills of the biologists, but the other fields as well - the physicists. So it's a two way process isn't it?

See more here:

The data cure: The changing science of biology and its impact on your health care

Blocking Autophagy with Malaria Drug May Help Overcome Resistance to BRAF Drugs in Melanoma

Contact Information

Available for logged-in reporters only

Newswise PHILADELPHIA Half of melanoma patients with the BRAF mutation have a positive response to treatment with BRAF inhibitors, but nearly all of those patients develop resistance to the drugs and experience disease progression.

Now, a new preclinical study published online ahead of print in the from Penn Medicine researchers found that in many cases the root of the resistance may lie in a never-before-seen autophagy mechanism induced by the BRAF inhibitors vermurafenib and dabrafenib. Autophagy is a process by which cancer cells recycle essential building blocks to fuel further growth. Block this pathway with the antimalarial drug hydroxycholoroquine (HCQ), the authors found, and the BRAF inhibitors will be able to do their job better.

This study opens the door for combination therapy with BRAF inhibitors and autophagy inhibitors, which havent been explored deeply as a therapeutic option for patients whose tumors are resistant, said Ravi K. Amaravadi, MD, assistant professor of Medicine in the division of Hematology/Oncology at the Perelman School of Medicine and co-leader of the Cancer Therapeutics Program at Penn Medicines Abramson Cancer Center. Here, we show that the BRAF inhibitors induce autophagy as a way to escape cell death, which gives us clues on how to interfere with this mechanism of resistance and improve outcomes for these patients.

Based on these promising preclinical results, Dr. Amaravadi and his team have already launched a clinical trial for patients with advanced BRAF mutant melanoma to see how well-tolerated HCQ is with the BRAF inhibitor vemurafenib. So far, he said, we are seeing a benefit to patients and low toxicity.

BRAF inhibitors are a first line of treatment for melanoma patients who harbor the BRAF mutation, which is an abnormal change in a gene that causes some melanoma tumors to grow and spread more aggressively. While 50 percent of patients initially respond to that treatment, nearly 100 percent exhibit disease progression seven months after treatment, making it imperative to find a way to re-sensitize the tumor to treatment.

Autophagy has emerged as a key pathway that cancer cells use to survive in the face of assault by chemotherapy and radiation; however, autophagy as a potential druggable mechanism in patients who become resistant to BRAF inhibitors has not been investigated.

Using tumor biopsies from BRAF melanoma patients treated with either BRAF inhibitors or with combined BRAF and MEK inhibitors, a recently FDA-approved drug combination to fight the other mechanisms of resistance, the researchers found that tumors resistant to the BRAF inhibitors had increased levels of autophagy compared with baseline tumors. Moreover, the level of therapy-induced autophagy was correlated with lower response rates and shorter progression-free survival times.

The researchers also examined BRAF mutant melanoma cell lines, and found that BRAF inhibition induced autophagy by way of an endoplasmic reticulum (ER) stress response. The binding of a BRAF mutation to the ER stress gatekeeper GRP78 is a new and unexpected molecular interaction driving resistance, and establishes a new signaling axis that has multiple drug targets, Dr. Amaravadi said.

More here:

Blocking Autophagy with Malaria Drug May Help Overcome Resistance to BRAF Drugs in Melanoma

SIU SDM's Gillespie Named Associate Editor of Anaerobe

Edwardsville, Ill. (PRWEB) February 24, 2014

Anaerobe has selected Dr. Jane Gillespie, professor of microbiology at the Southern Illinois University School of Dental Medicine, as associate editor of Molecular Biology and Genetics. Anaerobe is the official journal of the Anaerobe Society of the Americas and the Japanese Association for Anaerobic Infection Research.

Anaerobe is the leading scientific journal focused on bacteria and other organisms that live in anaerobic conditions, i.e., in the absence of oxygen. The journal has high relevance to dentistry, since most dental infections occur in either anaerobic or low oxygen environments.

Anaerobe publishes articles, notes and case reports in these categories: Anaerobes in the Microbiome, Clostridium difficile, Clinical Microbiology, Pathogenesis and Toxins, and Molecular Biology and Genetics. Gillespie will serve as associate editor for Molecular Biology and Genetics.

A member of the editorial board since its founding in 1996, Gillespie is proud to be named associate editor.

The journal articles reporting original, peer-reviewed research gradually, and sometimes dramatically, impact a field of study, said Gillespie. Therefore, it is an honor to be considered worthy of this task which requires both expertise in the discipline and sound judgment in determining what new knowledge will be disseminated. It is also an opportunity to continue to learn and remain current in my field.

Gillespie earned a bachelor of science in microbiology from Texas Tech University, and a master's in microbiology and doctorate in microbial physiology from the University of New Mexico. She completed post-doctoral training in dental research at The University of Texas Health Science Center in San Antonio and The State University of New York at Buffalo. Gillespie has also served as a peer reviewer for the Journal of Endodontics, Journal of Dental Education and other microbiology journals.

She became a faculty member with the SIU School of Dental Medicine in 1997.

The SIU School of Dental Medicine students manage approximately 35,000 patient visits each year at its patient clinics in Alton and East St. Louis. In addition, students offer oral health treatment, screenings and education to more than 10,000 people annually through a wide variety of off-campus community outreach events. These opportunities provide students the training they need to graduate and become highly skilled dentists. The School of Dental Medicine is a vital oral health care provider for residents of southern and central Illinois, and the St. Louis metropolitan region.

See original here:

SIU SDM's Gillespie Named Associate Editor of Anaerobe

Molecular 'cocktail' transforms skin cells into beating heart cells

The power of regenerative medicine appears to have turned science fiction into scientific reality -- by allowing scientists to transform skin cells into cells that closely resemble beating heart cells. However, the methods required are complex, and the transformation is often incomplete. But now, scientists at the Gladstone Institutes have devised a new method that allows for the more efficient -- and, importantly, more complete -- reprogramming of skin cells into cells that are virtually indistinguishable from heart muscle cells. These findings, based on animal models and described in the latest issue of Cell Reports, offer new-found optimism in the hunt for a way to regenerate muscle lost in a heart attack.

Heart disease is the world's leading cause of death, but recent advances in science and medicine have improved the chances of surviving a heart attack. In the United States alone, nearly 1 million people have survived an attack, but are living with heart failure -- a chronic condition in which the heart, having lost muscle during the attack, does not beat at full capacity. So, scientists have begun to look toward cellular reprogramming as a way to regenerate this damaged heart muscle.

The reprogramming of skin cells into heart cells, an approach pioneered by Gladstone Investigator, Deepak Srivastava, MD, has required the insertion of several genetic factors to spur the reprogramming process. However, scientists have recognized potential problems with scaling this gene-based method into successful therapies. So some experts, including Gladstone Senior Investigator Sheng Ding, PhD, have taken a somewhat different approach.

"Scientists have previously shown that the insertion of between four and seven genetic factors can result in a skin cell being directly reprogrammed into a beating heart cell," explained Dr. Ding, the paper's senior author and a professor of pharmaceutical chemistry at UCSF, with which Gladstone is affiliated. "But in my lab, we set out to see if we could perform a similar transformation by eliminating -- or at least reducing -- the reliance on this type of genetic manipulation."

To that effect, the research team used skin cells extracted from adult mice to screen for chemical compounds, so-called 'small molecules,' that could replace the genetic factors. Dr. Ding and his research team have previously harnessed the power of small molecules to reprogram skin cells into neurons and, more recently, insulin-producing pancreas cells. They reasoned that a similar technique could be used to do the same with heart cells.

"After testing various combinations of small molecules, we narrowed down the list to a four-molecule 'cocktail,' which we called SPCF, that could guide the skin cells into becoming more like heart cells," said Gladstone Postdoctoral Scholar Haixia Wang, PhD, the paper's lead author. "These newly reprogramed cells exhibited some of the twitching and contracting normally seen in mature heart cells, but the transformation wasn't entirely complete."

So, Drs. Ding and Wang decided to add one genetic factor, called Oct4, to the small molecule cocktail. And by doing so, the research team was able to generate a completely reprogrammed beating heart cell.

"Once we added Oct4 to the mix, we observed clusters of contracting cells after a period of just 20 days," explained Dr. Ding. "Remarkably, additional analysis revealed that these cells showed the same patterns of gene activation and electric signaling patterns normally seen in the ventricles of the heart."

Dr. Ding and his team believe that these results may point to a more desirable method for reprogramming, as ventricular heart cells are the type of cells typically lost during a heart attack. These findings give the team newfound optimism that the research is well on its way towards an entirely pharmaceutical-based method to regrow heart muscle.

"The fact that the combination of Oct4 and small molecules appears to generate beating heart cells in an accelerated fashion is encouraging," said Joseph Wu, MD, PhD, Director of the Stanford Cardiovascular Institute, who was not involved in this study. "Future advances by Dr. Ding and others will likely focus on improving the efficiency of conversion as well as duplicating the data in adult human cells."

Read the original here:

Molecular 'cocktail' transforms skin cells into beating heart cells

A*STAR scientists discover protein's role in human memory and learning functions

PUBLIC RELEASE DATE:

18-Feb-2014

Contact: Tan Yun Yun tan_yun_yun@a-star.edu.sg 656-826-6273 Biomedical Sciences Institutes (BMSI)

1. Scientists at A*STAR's Institute of Molecular and Cell Biology (IMCB) have identified the precise role of the protein, SNX27, in the pathway leading to memory and learning impairment. The study broadens the understanding of the brain's memory function and could be used to explain defects in the cognitive development of those with Down's syndrome. The newly established knowledge could potentially facilitate exploration of strategies to improve memory and learning abilities in Down's syndrome.

2. Down's syndrome is a genetic condition characterized by the presence of an additional copy of chromosome 21. About one in eight hundred new-borns is diagnosed with Down's syndrome. It is a condition that leads to impairments in both cognitive ability and physical growth that range from mild to moderate developmental disabilities. Yet, there is still no treatment for it.

3. In an earlier study published in Nature Medicine, an international team of scientists discovered that the additional copy of chromosome 21 in Down's syndrome reduces the production of SNX27 in the brain and results in synaptic dysfunction. Synapse, a structure that permits nerve cells to pass chemical signals to each other, is known to have an important role in memory formation and its dysfunction could result in impairment. By re-introducing SNX27 into the brain, memory could be restored hence suggesting that SNX27 is an essential protein for memory and learning.

4. The protein's role in the pathway leading to memory impairment, however, remained unclear until scientists from IMCB utilised live-cell imaging techniques to elucidate the mechanism of memory impairment and illustrated how SNX27 attributes to synaptic dysfunction. The scientists observed that transmission of chemical signals between the nerve cells are facilitated by AMPA-receptors and the activity occurs on the brain cell surface. As SNX27 plays an important role in shuttling the AMPA-receptor to the brain cell surface, lower levels of SNX27 means fewer receptors are carried to the surface, which consequently interrupts the signal transmission and impairs memory. This discovery was reported in the 24 January, 2014 issue of the renowned scientific journal, Nature Communications.

5. The correlation established between SNX27 levels and memory could explain why individuals with Down's syndrome encounter memory and learning difficulties. Identifying the target and its role is a crucial first step to therapy having known the role of SNX27 in memory impairment, future research on Down's syndrome could focus on developing strategies which can effectively re-introduce the protein into the brain to restore memory and learning abilities.

6. Dr Loo Li Shen was an A*STAR scholar who is now working with Executive Director of IMCB, Prof Wanjin Hong, and is also an adjunct Assistant Professor at LKC medical school of NTU. The lead author of the Nature Communications paper and co-author of the Nature Medicine paper said, "Memory and the ability to learn capture the essence of life. Our research goes beyond the lab to make a difference by finding ways to grant these fundamental capabilities to those diagnosed with Down's syndrome. Our ultimate goal is to create a positive impact on the lives of these valuable children."

7. Prof Hong added, "In IMCB, we conduct research that would contribute to our understanding of mankind. This is a good example where the study is directed at a human condition - the Down's syndrome. The knowledge established from the findings could potentially translate into treatments for Down's syndrome or even become applicable to other similar human conditions."

The rest is here:

A*STAR scientists discover protein's role in human memory and learning functions

Molecular diagnosis helps patients avoid thyroid surgery

A molecular diagnosis gives doctors and patients better treatment options when suspicious lumps are found in the neck.

Genetic biopsy: A Veracyte technician tests 142 genes from patients with suspicious nodules in their thyroid glands.

Later this year, doctors in the U.S. will be able to use a gene test to guide thyroid cancer surgery. The test helps determine when patients harbor a particularly dangerous form of the disease, which can require surgeons to do a second operation on top of the initial diagnostic procedure. Knowing that a patient has this particular form of thyroid cancer could enable surgeons to instead do a single, more extensive surgery.

The company behind the test, Veracyte, already sells a unique genetic assay that helps doctors decide whether to perform surgery on thyroid cancer patients at all. Thyroids that are not cancerous are often removed, which means unnecessary surgery and lifelong hormone replacement therapy for some patients.

Both tests are part of a broader movement in recent years to bring genetic tests into medical care, with oncology leading the way. One test, from Myriad Genetics, looks for mutations linked to increased risk of cancer; others, such as one offered by Foundation Medicine, help doctors prescribe drugs tailored to a particular tumor (see "Foundation Medicine: Personalizing Cancer Drugs").

Veracyte's first test is the only one that rules out cancer. A lump, or nodule, is caused by growths of cells in the thyroid gland, which is located in the base of the neck. Most often these growths are not cancers. To figure out whether they are, doctors will first take a small needle to extract cells from the lump and then look at the cells under the microscope. And up to 30 percent of the time in U.S. clinics, that test is inconclusive. Because cancer can't be ruled out, typically the next step is to remove the thyroid. The gland normally produces important hormones that regulate metabolism and other body functions, so patients usually then have to take hormone replacement therapy for the rest of their lives.

Between 60 and 80 percent of the time, the nodule in the removed thyroid turns out to be benign. "You have unnecessarily put a patient through surgery," says Kishore Lakshman, director of a community thyroid care center in Fall River, Massachusetts. This puts patients at risk for complications such as infection, and creates dependence on hormone therapy. Since 2011, Lakshman has been using Veracyte's gene test to assess the risk of cancer in patients whose initial thyroid screen was inconclusive. "When I found out that there was a very efficient way of knowing the benign potential of a nodule without exposing a patient to surgery, I was quick to jump on it," says Lakshman.

Veracyte analyzed gene expression levels in hundreds of patients with thyroid nodules, some cancerous, some not, and identified 142 genes that can reliably separate benign from malignant samples. "Measuring every gene in the human genome, our scientific team was able to extract genomic information and interpret it with machine-learning algorithms taught to recognize patients with benign nodules," says Bonnie Anderson, CEO and cofounder of the South San Francisco-based company.

The performance of the test was evaluated and published in the New England Journal of Medicine in 2012. That trial showed that Veracyte's test can reclassify a nodule from indeterminate to benign 95 percent of the time.

Here is the original post:

Molecular diagnosis helps patients avoid thyroid surgery