header logo image


Page 20«..10..19202122..30..»

Archive for the ‘Molecular Genetics’ Category

Clemson geneticists’ collaborative research sheds light on ‘dark’ portion of genome – Clemson Newsstand

Monday, March 9th, 2020

CLEMSON, South Carolina Just as there is a mysterious dark matter that accounts for 85 percent of our universe, there is a dark portion of the human genome that has perplexed scientists for decades. A study published March 9, 2020, in Genome Research identifies new portions of the fruit fly genome that, until now, have been hidden in these dark, silent areas.

The collaborative paper titled Gene Expression Networks in the Drosophila Genetic Reference Panel is the culmination of years of research by Clemson University geneticists Trudy Mackay and Robert Anholt. Their groundbreaking findings could significantly advance sciences understanding of a number of genetic disorders.

Robert Anholt (left) and Trudy Mackay in their lab at the Clemson Center for Human Genetics.Image Credit: Robert Bradley, College of Science

The dark portion refers to the approximate 98 percent of the genome that doesnt appear to have any obvious function. Only 2 percent of the human genome codes for proteins, the building blocks of our bodies and the catalysts of the chemical reactions that allow us to thrive. Scientists have been puzzled by this notion since the 1970s when gene sequencing technologies were first developed, revealing the proportion of coding to noncoding regions of the genome.

Genes are traditionally thought to be transcribed into RNAs, which are subsequently translated into proteins, as dictated by the central dogma of molecular biology. However, the entire assemblage of RNA transcripts in the genome, called the transcriptome, contains RNA species that appear to have some other function, apart from coding for proteins. Some have proposed that noncoding regions might contain regulatory regions that control gene expression and the structure of chromosomes, yet these hypotheses were difficult to study in past years as diagnostic technology was developing.

Only in recent years, with the sequencing of the entire transcriptome complete, have we realized how many RNA species are actually present. So, that raises the whole new question: if they arent making the proteins the work horses of the cell then what are they doing? said Mackay, director of Clemson Universitys Center for Human Genetics (CHG), which is part of the College of Science.

For Mackay and Anholt, also of the CHG, these human genetics questions can be probed by studying the common fruit fly, Drosophila melanogaster. Because many genes are conserved between humans and fruit flies, findings revealed by analyzing the Drosophila genome can be extrapolated to human health and disease.

Mackay and Anholts former postdoctoral researchers, Logan Everett and Wen Huang, led the charge on this latest research, which identified more than 4,500 new transcripts in Drosophila that have never been uncovered before. Referred to by the researchers as novel transcribed regions, these 4,500 transcripts consist primarily of noncoding RNAs that appear to be involved in regulating networks of genes and that could contribute to genetic disorders.

Most disease-causing mutations are known to occur in the protein-coding portion of the genome, known as the exome, but when youre only sequencing the exome, you miss other disease-related factors in other parts of the genome, such as these long noncoding RNAs, said Anholt, Provosts Distinguished Professor of Genetics and Biochemistry at Clemson University. Now that the cost of whole genome sequencing has gone down considerably, and we have the capability of sequencing whole genomes rapidly, we can look at elements of the genome that have traditionally been considered unimportant, and we can identify among them potential disease-causing elements that have never been seen before.

By probing several hundred inbred Drosophila fly lines, each containing individuals that are virtually genetically identical, the researchers discovered that many of the novel long noncoding RNAs regulate genes in heterochromatin, a tightly packed form of DNA in the genome that is usually considered silent. Because heterochromatin is so condensed, it was thought to be inaccessible to the molecular machinery that transcribes DNA into RNA. Thus, any genes contained within heterochromatin are kept off, silent and unexpressed or are they?

What we think is that the repression of gene expression in heterochromatin is somewhat leaky, and that there is variation in how those genes are repressed, Mackay said. The network of RNAs weve discovered may have to do with actually regulating chromatin state.

These noncoding RNAs may play an important role in opening up such regions of the genome for expression of genes in a way that varies among different individuals depending on their genetic background, Anholt added.

Trudy Mackay and Robert Anholt address human genetics questions by studying the common fruit fly, Drosophila melanogaster, because many genes are conserved between humans and fruit flies, meaning research results can be extrapolated to human health and disease.Image Credit: College of Science

Another outcome of the study is the expression of jumping genes, known as transposons, that are pieces of DNA able to move around the genome. As transposons cut and paste into other genes, they may cause genome instability that leads to cancer, neurodegenerative disorders and other diseases. These transposons were also located in heterochromatin, but the identification of transcripts of these transposons shows that they are actually being expressed, despite residing in a usually silent portion of the genome. Identifying regulators of transposable elements, as the researchers found among these 4,500 novel transcribed regions, could prove useful in treating disorders that stem from transposon interference.

Overall, the study lends toward a greater understanding of gene regulatory networks that contribute to human health and disease.

These observations open up an entirely new area of biology that hasnt been explored and has unlimited potential for future follow-up, Anholt said.

The teams own follow-up studies are using CRISPR gene editing technology to uncover what happens when genes revealed by this study are altered or deleted from the Drosophila genome. If the expression of other genes is altered by knocking one out, important conclusions can be drawn about the role that deleted gene plays in development or progression of disease.

Everett, one of the lead authors on the Genome Research publication, is now a bioinformatics scientist at the U.S. Environmental Protection Agency. Wen Huang is an assistant professor in the Department of Animal Science at Michigan State University.

###

The study was supported by the National Institutes of Health under grant numbers R01-AA016560, R01-AG043490 and U01- DA041613. Additional support was provided by The Danish Council for Strategic Research. The researchers are wholly responsible for the content of this study, of which the funders had no input.

Originally posted here:
Clemson geneticists' collaborative research sheds light on 'dark' portion of genome - Clemson Newsstand

Read More...

Podcast: Bird poop, pus, and the Manhattan projectthe surprising origins of the genetic alphabet – Genetic Literacy Project

Monday, March 9th, 2020

On the latest episode of Genetics Unzipped, biologist Kat Arney explores the origins of the genetic alphabet: A, C, T and Gthe four letters that spell out all the genetic recipes encoded in DNA.

These letters are the initials of the four nucleotide chemicals that make up DNA: adenine, cytosine, thymine and guanine. They are ingrained in the scientific lexicon and burned into the brain of anyone whos ever worked with or even just learned about genes, genomes and DNA. Its a code thats as inseparable from genetics as the double helix itself.

But while many people know that the structure of DNA was figured out in the 1950s, far fewer people realize that the identities of these molecular letters were uncovered far earlier. In search of the origins of nucleotide names, Arney takes us from the bird poop boom of the 1840s through the heyday of atomic weapons research in the 1940s and beyond.

First we explore the guano mountains of Peru giant hills of solidified seabird excrement which were mined for fertilizer to feed a fast-developing world. Intrigued by the nourishing properties of guano, 19th-century scientists started to investigate its chemical components. This led to German chemist Julius Ungers discovery of guanine in 1846 the first nucleotide molecule to be identified.

DNA itself wasnt discovered until nearly twenty years later, when Johannes Friedrich Miescher isolated a strange sludgy substance from pus-soaked bandages obtained from a nearby clinic. He called it nuclein a name that still lingers in the formal chemical name for DNA, deoxyribonucleic acid. However, his discovery almost went ignored as his supervisor, Felix Hoppe-Seyler, did not believe that such an inexperienced researcher could make such an important finding.

Following up on Mieschers work, his colleague Albrecht Kossel identified cytosine, thymine and adenine as the other components of this mysterious nuclein, after purifying the chemicals from huge amounts of cow organs obtained from a nearby slaughterhouse.

The story of nucleotides doesnt end with the discovery of A, C, T and G. Although these four letters make up the genetic code of DNA, theres another base Uracil, or U that replaces thymine in RNA, a kind of molecular photocopy thats made when genes are read. And we also now know that DNA and RNA bases can be chemically altered to extend the genetic code in some very interesting ways. Finally, we hear how the discovery of the first modified RNA base, pesudouridine, came from a surprising source: the US atomic weapons program at Oak Ridge laboratory in Tennessee.

Full transcript, links and references available online atGeneticsUnzipped.com

Genetics Unzippedis the podcast from the UKGenetics Society,presented by award-winning science communicator and biologistKat Arneyand produced byFirst Create the Media.Follow Kat on Twitter@Kat_Arney,Genetics Unzipped@geneticsunzip,and the Genetics Society at@GenSocUK

Listen to Genetics Unzipped onApple Podcasts(iTunes)Google Play,Spotify,orwherever you get your podcasts

Follow this link:
Podcast: Bird poop, pus, and the Manhattan projectthe surprising origins of the genetic alphabet - Genetic Literacy Project

Read More...

NorthShore Moving Toward Greater Integration of Genomics Into Primary Care – GenomeWeb

Monday, March 9th, 2020

NEW YORK After assessing the impact genetic information can have on patient care within clinics and pilot programs, NorthShore University HealthSystem is working on expanding test access to primary care patients and integrating the results into their day-to-day care.

In 2020, "we're trying to focus on how to hardwire this so it's not just a bunch of pilots but really starting to be ingrained in practice," said Peter Hulick, medical director of NorthShore's Center for Personalized Medicine.

GenomeWeb Premium gives you: Full site access Interest-based email alerts Access to archives Never miss another important industry story.

Try GenomeWeb Premium now.

You may already have institutional access!

Check if I qualify.

Already a GenomeWeb or 360Dx Premium member?Login Now.

*Before your trial expires, well put together a custom quote with your long-term premium options.

Read more from the original source:
NorthShore Moving Toward Greater Integration of Genomics Into Primary Care - GenomeWeb

Read More...

The Biology Behind Your Love (or Hatred) of Coffee – Technology Networks

Monday, March 9th, 2020

Why do some people feel like they need three cups of coffee just to get through the day when others are happy with only one? Why do some people abstain entirely? New research suggests that our intake of coffee - the most popular beverage in America, above bottled water, sodas, tea, and beer - is affected by a positive feedback loop between genetics and the environment.

This phenomenon, known as "quantile-specific heritability," is also associated with cholesterol levels and body weight, and is thought to play a role in other human physiological and behavioral traits that defy simple explanation.

"It appears that environmental factors sort of set the groundwork in which your genes start to have an effect," said Paul Williams, a statistician at Lawrence Berkeley National Laboratory (Berkeley Lab). "So, if your surroundings predispose you to drinking more coffee - like your coworkers or spouse drink a lot, or you live in an area with a lot of cafes - then the genes you possess that predispose you to like coffee will have a bigger impact. These two effects are synergistic."

Williams' findings, published in the journalBehavioral Genetics, came from an analysis of 4,788 child-parent pairs and 2,380 siblings from the Framingham Study - a famous, ongoing study launched by the National Institutes of Health in 1948 to investigate how lifestyle and genetics affect rates of cardiovascular disease. Participants, who are all related to an original group from Framingham, Massachusetts, submit detailed information about diet, exercise, medication use, and medical history every three to five years. Data from the study have been used in thousands of investigations into many facets of human health.

Williams used a statistical approach called quantile regression to calculate what proportion of participants' coffee drinking could be explained by genetics - as the study follows families - and what must be influenced by external factors. Past research shows that the most significant environmental factors influencing coffee drinking are culture and geographic location, age, sex, and whether or not one smokes tobacco; with older male smokers of European ancestry drinking the most, overall.

The analysis indicated that between 36% and 58% of coffee intake is genetically determined (although the exact causative genes remain unknown). However, confirming Williams' hypothesis that coffee drinking is a quantile-specific trait, the correlation between a parent's coffee drinking and an offspring's coffee drinking got increasingly stronger for each offspring's coffee consumption quantile, or bracket (for example, zero cups per day, one to two cups, two to four cups, and five or more cups).

"When we started to decode the human genome, we thought we'd be able to read the DNA and understand how genes translate into behavior, medical conditions, and such. But that's not the way it's worked out," said Williams, who is a staff scientist in Berkeley Lab's Molecular Biophysics & Integrated Bioimaging (MBIB) Division. "For many traits, like coffee drinking, we know that they have a strong genetic component - we've known coffee drinking runs in families since the 1960s. But, when we actually start looking at the DNA itself, we usually find a very small percentage of the traits' variation can be attributed to genes alone."

The traditional assumption in genetic research has been that one's surroundings and lifestyle alter gene expression levels in consistent and measurable ways, ultimately creating the outward manifestation - called a phenotype - of a trait. Williams' statistics work shows that the situation is more complex, which helps explain the diversity of traits we see in the real world.

MBIB Division Director Paul Adams commented, "Paul's statistical studies complement the genomics research that Berkeley Lab bioscientists conduct to learn more about the relationship between genes and the environment."

Next, Williams plans to assess whether quantile-specific heritability plays a role in alcohol consumption and pulmonary function. "This is a whole new area of exploration that is just now opening up," he said. "I think it will change, in a very fundamental way, how we think genes influence a person's traits."

Reference:Paul T. Williams. (2020).Quantile-Specific Heritability may Account for GeneEnvironment Interactions Involving Coffee Consumption. Behavior Genetics.DOI: https://doi.org/10.1007/s10519-019-09989-0.

This article has been republished from the following materials. Note: material may have been edited for length and content. For further information, please contact the cited source.

Here is the original post:
The Biology Behind Your Love (or Hatred) of Coffee - Technology Networks

Read More...

A Conversation With a Harvard Geneticist on How to Live (Well) Past 100 – InsideHook

Monday, March 9th, 2020

In Parks and Rec, Rob Lowes Chris Traeger is a perennially positive, supplement-popping 45-year-old who glides through the rooms of Pawnee City Hall with golden retriever energy. He brings vegetable loaves to birthday parties, regularly runs 10 miles during his lunch breaks and touts just 2.8% body fat. In Season 2 of the show, Traeger reveals his lifes goal: to live to 150.

Scientists believe that the first human being to live to a 150 years has already been born I believe I am that human being. At first, it sounds like just another quotable line from a show thats famous for them. Traeger isnt to be taken seriously, after all. One of his other signature adages is simply Stop pooping. (On the exceedingly rare occasions that Traegers body fails him, he lands in a dark place.)

Believe it or not, though, Traegers right. At least one scientist has been predicting humankinds potential to live to 150 for the better part of a decade, a man whos furthered the notion of aging as disease since he arrived at MIT in the late 1990s. That would be Australian Dr. David Sinclair, a biology rockstar and former Time 100 honoree with an Order of Australia (Down Unders version of knighthood), and his own genetics lab at Harvard Medical School.

In September of last year, Dr. Sinclair released Lifespan: Why We Age and Why We Dont Have To. Its an explosive call to arms detailing Dr. Sinclairs core belief, which hes spent decades researching: most humans leave decades of high-quality life on the table simply because society doesnt afford aging the same attention and dollars it reserves for other health crises like cancer and heart disease. The book is one part memoir (Dr. Sinclair recalls the drawn-out final decades of his mother and grandmothers lives), one part crash-course in epigenetics (we hold far more in common with yeast cells than the common person knows) and one part sneak peek into the advancements being made in the worlds preeminent genetics labs (Dr. Sinclairs team has successfully cured blindness in mice).

Most refreshingly, though, Lifespan delights in giving answers. On top of the many science-fiction-esque wonders on display at Harvard Medical School each week (Dr. Sinclair is a pioneer of a practice called cellular programming, which effectively means resetting cells back to a younger age), the book includes functional day-to-day advice on how the layman or woman can activate survival processes in their epigenome, engaging specific sirtuin proteins (a class of protein that helps regulate cellular aging) to help foster greater longevity.

Basically, Sinclairs hypothesis is that eating a certain way, working out a certain way and exposure to a certain kind of temperature can make living past 100 a relative breeze. We recently caught up with Dr. Sinclair to discuss his book, intermittent fasting, Benjamin Button and more.

InsideHook: This book definitely doesnt mince concepts or words. Why was it important to you to write so boldly on aging as a disease?

Dr. David A. Sinclair: The world is in a stupor when it comes to aging. Theres a blind spot. I wrote the book to shake things up, and hopefully wake up those who dont think aging is important or worth working on. We focus as a society far too much on the end consequence of aging, playing whack-a-mole with these diseases that kill us. We ignore whats actually driving these diseases. The more we study aging, though, the more we realize that the diseases we treat are all manifestations of an underlying process. And its treatable.

Some of your peers in the field have said it isnt a good look to be so declarative in your predictions on aging. Have they changed their tune since the book was released?

I havent had any criticism from colleagues since the book came out. Either they havent read it, or theyre okay with my arguments. But also, the world is changing. What used to be considered crazy 10 years ago is no longer crazy. For example, scientists didnt used to say the phrase reversal of aging. But now, its a fact thats doable. Our field has proven that many aspects of aging are reversible, including blindness. Its also partly that I was ahead of the curve, and that things which were once forbidden are now in the realm of discussion and debate.

Im fascinated by the cellular reprogramming work your lab has done. In the book, you invoke F. Scott Fitzgeralds Benjamin Button story to describe how a 50-year-old could soon begin a routine that will have him/her feeling and looking 30 again. Are we actually close to seeing that sort of treatment in the developed world?

The first thing to say is we now understand that changes in your lifestyle can dramatically improve your age and physiology. We used to think that aging was just something that was in our genes, something that we couldnt modify. But very rapidly, within months of changing diet and exercise, you can reverse many aspects of aging. Its never too late, unless youre on your last legs. The fact that its that easy to slow down and reverse aspects of aging just with lifestyle changes totally fits with our understanding of molecular mechanisms. We should be able to slow aging even better with the reprogramming of cells. I see the work weve done as a proof of concept. While its true that Im working hard towards restoring eyesight in people whove lost their vision, its really just the beginning. This work is proof that its possible to restore the age of a complex tissue. In the same way that the Wright brothers werent building rockets to the moon, they could at least imagine that one day it would be possible. Weve shown that there is a backup copy of a youthful epigenome that we can turn on to reset the cell and get it to work again. If thats doable in the eye, it would be rather pessimistic to say we were just lucky to choose the right body part for this to work.

High-intensity training is one of the practices you cite as vital to this process. What about it encourages longevity genes?

Weve found that high-intensity training will induce the sirtuin defenses in the body, similar to what intermittent fasting does. When those genes come on, they defend the cell against diseases, and aging itself. When we dont engage those sirtuin genes, we dont reap the benefits. High-intensity training is particularly good at turning on the sirtuins, because it encourages a hypoxic response, which weve shown leads to the activation of these defense mechanisms. While walking is good, its not as good as doing high-intensity training.

Im glad you mentioned intermittent fasting, another practice you endorse. Are there any mistruths or misunderstandings in the way that popular media portrays it?

Based on recent results in animal studies, its not so much what you eat but when you eat. Of course, you cant eat a hamburger morning, noon and night, then fast the next day and expect to get the maximum benefits. That said, it seems to be more about just having a period of fasting in general. Theres one misconception that people need an optimal mix of protein, carbohydrates and fat, and that thats the most important thing to get right. Id say worry about that less, as long as youre getting nutrients and xenohormetic molecules, which are molecules produced by plants when theyre under stress. As long as youre doing those things, its far more important to skip meals.

One other thing: people claim that there is an optimal intermittent fasting protocol. The truth is, we dont know what the optimal is. Were still learning, and its individual. There are individual differences in all of us. There is a subset of people, myself included, who start producing glucose out of their livers early in the morning, at around 6 a.m. Which means, for me, to start eating breakfast around 7 a.m. makes no sense. Some people, though, have such low blood sugar in the morning that they can barely function. We also dont know the best method. Is it the 16/8 [hours, first on and then off of the fast]? Two days fasting out of every five? We really dont know yet. But we do know that if youre never hungry, if youre eating three meals a day and snacking in between, thats the worst thing you can do. It switches off your bodys defenses. Some fasting is better than none.

Do you eat meat?

I do, but its a gradient. Its mostly plants, then fish, rarely chicken, and almost never red meat.

From an aging perspective, do you recommend that people give up meat?

For the average person, focus on plants. Meat isnt going to kill you if you eat it once in a while, but the reason for the plant-based diet is we know where the hot spots are for longevity. We know what theyre eating. Its not a mystery. Theyre not carnivores. Theyre eating mostly plants, and a little bit of meat maybe, a bit of fish. Theyre consuming olive oil, avocados, red wine and other plants that have xenohormetic molecules. I dont think that thats a coincidence.

Theres been some coverage recently about the rise of wild swimming. In the UK, especially, people have started jumping into freezing cold water and claiming all sorts of health benefits. It reminded me of your points in the book about challenging the thermoneutral zone. Does one need to frequently experience extremely cold temperatures to reap benefits?

Cold baths, cryotherapy I was skeptical. I started out skeptical until proven otherwise. But theres some evidence that making brown fat is good. Adult humans can make brown fat as long as theyre not super old, and cold is a good way to do that. One of my favorite genes, the third of the seven sirtuin genes, boosts brown fat. All of these things that were talking about exercise, fasting, cold therapy, even a sauna its best to mix it up. You dont want to be constantly exercising, constantly hungry, or constantly at one temperature or another. You want to shock the body. Putting a few days of recovery in between makes a lot of sense. As for exposing yourself to cold, a little is still better than nothing. I do it once a week. But Im still trying to figure out when to do these ice baths. There was a study that an ice bath after a workout potentially lowers the benefit of the workout.

Lifespan devotes a ton of pages to metformin, the anti-diabetic medication thats been discovered to activate longevity genes. Are there adverse side effects from taking metformin? It seems a little too good to be true.

As far as drugs go, metformin is very safe. The World Health Organization declared it one of the essential medicines for humanity. One in 10,000 people have an adverse side reaction and have to stop taking it. The majority of complaints are attributed to a queasy stomach feeling until you get used to it. I actually dont mind, because it stops me from getting hungry. [Editors note: Dr. Sinclair takes metformin daily.] It doesnt give you anything like a greater risk of cancer or heart disease. The data actually suggests the opposite. The risk of getting old is pretty high, but the risk of taking metformin is pretty low, based on millions of people taking it.

Youre on the record saying the first person to live to 150 has been born. Would that person need to combine every single practice and innovation that you outline in this book in order to do so?

An important point of clarification: I dont think we have any technology today that would get us to 150. But if youre born today, you can be around until the mid-22nd century. Theres a lot thats going to happen between now and then. Were on a path of technological development. Once you see the trajectory and barriers are broken down, it gives me the license to say someone born today will live far longer than we can imagine. People born today will benefit from technologies that come about after were dead. The big breakthrough is being able to reprogram the body. If we can get that to work, wed be literally able to turn the clock back on cells. Weve done it once we managed to restore vision in mice but you might be able to reset cells twice. Or 100 times. Well just have to see.

Related: The Healthiest Blue Zone in Every State, Mapped

Subscribe herefor our free daily newsletter.

See the article here:
A Conversation With a Harvard Geneticist on How to Live (Well) Past 100 - InsideHook

Read More...

The complex biology behind your love (or hatred) of… – ScienceBlog.com

Monday, March 9th, 2020

Why do some people feellike they need three cups of coffee just to get through the day when others are happy with only one? Why do some people abstain entirely? New research suggests that our intake of coffee the most popular beverage in America, above bottled water, sodas, tea, and beer is affected by a positive feedback loop between genetics and the environment.

This phenomenon, known as quantile-specific heritability, is also associated with cholesterol levels and body weight, and is thought to play a role in other human physiological and behavioral traits that defy simple explanation.

It appears that environmental factors sort of set the groundwork in which your genes start to have an effect, said Paul Williams, a statistician at Lawrence Berkeley National Laboratory (Berkeley Lab). So, if your surroundings predispose you to drinking more coffee like your coworkers or spouse drink a lot, or you live in an area with a lot of cafes then the genes you possess that predispose you to like coffee will have a bigger impact. These two effects are synergistic.

Williams findings, published in thejournal Behavioral Genetics, came from an analysis of 4,788 childparent pairs and 2,380 siblings from the Framingham Study a famous, ongoing study launched by the National Institutes of Health in 1948 to investigate how lifestyle and genetics affect rates of cardiovascular disease. Participants, who are all related to an original group from Framingham, Massachusetts, submit detailed information about diet, exercise, medication use, and medical history every three to five years. Data from the study have been used in thousands of investigations into many facets of human health.

Paul T. Williams. (Credit: Roy Kaldschmidt/Berkeley Lab)

Williams used a statistical approach called quantile regression to calculate what proportion of participants coffee drinking could be explained by genetics as the study follows families and what must be influenced by external factors. Past research shows that the most significant environmental factors influencing coffee drinking are culture and geographic location, age, sex, and whether or not one smokes tobacco; with older male smokers of European ancestry drinking the most, overall.

The analysis indicated that between 36% and 58% of coffee intake is genetically determined (although the exact causative genes remain unknown). However, confirming Williams hypothesis that coffee drinking is a quantile-specific trait, the correlation between a parents coffee drinking and an offsprings coffee drinking got increasingly stronger for each offsprings coffee consumptionquantile, or bracket (for example, zero cups per day, one to two cups, two to four cups, and five or more cups).

When we started to decode the human genome, we thought wed be able to read the DNA and understand how genes translate into behavior, medical conditions, and such. But thats not the way its worked out, said Williams, who is a staff scientist in Berkeley Labs Molecular Biophysics & Integrated Bioimaging (MBIB) Division. For many traits, like coffee drinking, we know that they have a strong genetic component weve known coffee drinking runs in families since the 1960s. But, when we actually start looking at the DNA itself, we usually find a very small percentage of the traits variation can be attributed to genes alone.

The traditional assumption in genetic research has been that ones surroundings and lifestyle alter gene expression levels in consistent and measurable ways, ultimately creating the outward manifestation called a phenotype of a trait. Williams statistics work shows that the situation is more complex, which helps explain the diversity of traits we see in the real world.

MBIB Division Director Paul Adams commented, Pauls statistical studies complement the genomics research that Berkeley Lab bioscientists conduct to learn more about the relationship between genes and the environment.

Next, Williams plans to assess whether quantile-specific heritability plays a role in alcohol consumption and pulmonary function. This is a whole new area of exploration that is just now opening up, he said. I think it will change, in a very fundamental way, how we think genes influence a persons traits.

This research was funded by a grant from the National Institute of Environmental Health Sciences and a gift from HOKA ONE ONE. The Framingham Study Data were made available through the Biologic Specimen and Data Repository Information Coordinating Center of the National Heart, Lung, and Blood Institute.

See the original post here:
The complex biology behind your love (or hatred) of... - ScienceBlog.com

Read More...

Blue Shield of California Becomes First Health Plan in US to Cover Cost of Rapid Whole Genome Sequencing for Critically Ill Children – Hispanic PR…

Monday, March 9th, 2020

OAKLAND, Californiaand SAN DIEGO, March 9, 2020 /PRNewswire-HISPANIC PR WIRE/ Blue Shield of California has become the first health plan in the United States to cover rapid and ultra-rapid Whole Genome Sequencing to help critically ill babies and children in intensive care with unexplained medical conditions receive precision care.

Rady Childrens Institute for Genomic Medicine researchers have pioneered the fastest use of this advanced diagnostic technology to rapidly identify and decode the root causes of rare genetic disorders for some of the sickest infants and children hospitalized in intensive care across the country.

The Rady Childrens Institute team offers the quickest turnaround of genomic test results available nationwide, delivering a preliminary diagnosis in less than three days for medically urgent cases. The blood samples can be taken at any hospital and sent to Rady Childrens Institute for sequencing and analysis.

Our system is optimized to identify or rule out most genetic diseases in a single test, and provide the medical team at the bedside with child-specific, disease-specific information so they can make better, faster medical decisions, said Stephen Kingsmore, M.D., DSc, president and CEO of the Institute.

Whole genome sequencing scans a childs entire genetic makeup for thousands of anomalies from a blood sample. Rady Childrens specialists also provide consultation to the medical team caring for the patient to offer targeted guidance that can enable timely and precise personalized care.

We know that uncertainty and long testing wait times can create tremendous risks for children in intensive care, and anxiety for their families, all the while creating more challenges for physicians and specialists, said Terry Gilliland, M.D., executive vice president of Healthcare Quality and Affordability at Blue Shield of California. By providing our members with access to Rady Childrens Institute for Genomic Medicines pioneering work in rapid whole genome sequencing, were supporting them in what is often the most difficult time in their lives.

Blue Shield members with Individual and Family Plans or employer-sponsored health plans who have a critically ill child, up to age 18, hospitalized in neonatal or pediatric intensive care at any location with an undiagnosed condition may be eligible.

This is the latest example of Blue Shields leadership in making the newest evidence-based medical technologies and services available to its members.

The nonprofit health plan also was the first insurer to cover confirmatory testing for members who received a positive Ashkenazi Jewish BRCA finding from consumer genetic-testing companies such as 23andMe, as well as prostate gene expression assays for patients with low risk prostate cancer, helping them to avoid unnecessary radiation treatment and surgical intervention.

Without medical insurance coverage, access to rapid Whole Genome Sequencing is often not readily available for many hospitalized children who could potentially benefit from this service. Families in need of this care have often had to rely upon funding provided by private philanthropy and research grants to gain access to rapid Whole Genome Sequencing and associated precision care.

Genetic disease is a leading cause of infant death in the U.S. and Blue Shield is paving the way in providing coverage for this rapid, molecular diagnosis that can result in life-saving treatments, Dr. Kingsmore said.

Located on the campus of Rady Childrens Hospital-San Diego, the Institute houses a state-of-the-art genome sequencing lab and employs a multi-disciplinary team of experts who specialize in providing timely and accurate guidance to physicians caring for children with rare genetic disease.

About Blue Shield of CaliforniaBlue Shield of California strives to create a healthcare system worthy of our family and friends that is sustainably affordable. Blue Shield of California is a tax paying, nonprofit, independent member of the Blue Cross Blue Shield Association with over 4 million members, 6,800 employees and more than $20 billion in annual revenue. Founded in 1939 in San Francisco and now headquartered in Oakland, Blue Shield of California and its affiliates providehealth, dental, vision, Medicaid and Medicare healthcare service plans in California. The company has contributed more than $500 million to Blue Shield of California Foundation since 2002 to have an impact on California communities.

For more news about Blue Shield of California, please visitnews.blueshieldca.com. Or follow us on LinkedIn, Twitter, or Facebook.

About Rady Childrens Institute for Genomic MedicineThe Institute is leading the way in advancing precision healthcare for infants and children through genomic and systems medicine research. Discoveries at the Institute are enabling rapid diagnosis and targeted treatment of critically ill newborns and pediatric patients at Rady Childrens Hospital-San Diego and partner hospitals. The vision is to expand delivery of this life-saving technology to enable the practice of precision pediatric medicine at childrens hospitals across California, the nation and the world. RCIGM is a subsidiary of Rady Childrens Hospital and Health Center. Learn more at http://www.RadyGenomics.org. Follow us on Twitterand LinkedIn.

Photo https://mma.prnewswire.com/media/1120957/Rady_Childrens_Institute.jpg

Logo https://mma.prnewswire.com/media/810201/blue_jpg_Logo.jpg

SOURCE Blue Shield of California

Continued here:
Blue Shield of California Becomes First Health Plan in US to Cover Cost of Rapid Whole Genome Sequencing for Critically Ill Children - Hispanic PR...

Read More...

Molecular Genetics – an overview | ScienceDirect Topics

Monday, March 9th, 2020

Wayne W. Grody, Joshua L. Deignan, in Emery and Rimoin's Principles and Practice of Medical Genetics, 2013

This article is a revision of the previous edition article by Wayne W Grody, volume 1, pp 601626, 2007, Elsevier Ltd.

Molecular genetic testing has a unique range of indications, most of which are quite different from the uses of traditional clinical laboratory testing and even molecular biologic testing in other disease classes (e.g. infectious disease, cancer). The technical approaches as well as the psychosocial and ethical implications of molecular genetic tests may vary substantially depending on the reason for testing (e.g. diagnostic, carrier screening). Just as many of the applications are unique, so too the types of patient samples collected for molecular genetic testing may be different from those obtained for other types of clinical laboratory testing. In addition, the choice of technique will depend on the nature of the disease gene being studied (especially its size and mutational heterogeneity), the purpose of the test, and to some extent the condition of the specimen, and examples of specific conditions are discussed. Finally, high complexity laboratories performing molecular genetic testing need to be aware of the specific regulatory considerations involved.

Originally posted here:
Molecular Genetics - an overview | ScienceDirect Topics

Read More...

Ross Prize Awarded to Cold Spring Harbor Laboratory Professor – Yahoo Finance

Monday, March 9th, 2020

Dr. Adrian R. Krainer, pioneer in neuromuscular disease treatment to be honored at June 8 symposium in New York City

The Feinstein Institutes for Medical Research has selected Adrian R. Krainer, PhD, St. Giles Foundation Professor at Cold Spring Harbor Laboratory, as the eighth awardee of the Ross Prize in Molecular Medicine. The prize is awarded annually through the Feinstein Institutes peer-reviewed, open-access journal, Molecular Medicine, and includes a $50,000 award that will be presented to Dr. Krainer on June 8 at the New York Academy of Sciences (NYAS) in Manhattan.

This press release features multimedia. View the full release here: https://www.businesswire.com/news/home/20200302005570/en/

Dr. Adrian R. Krainer (Credit: Cold Spring Harbor Laboratory)

The Ross Prize is made possible by the generosity of Feinstein Institutes board members Robin and Jack Ross. It is awarded to scientists who have made a demonstrable impact in the understanding of human disease pathogenesis and/or treatment and who hold significant promise for making even greater contributions to the general field of molecular medicine. Dr. Krainer is being recognized for his pioneering work in introducing antisense therapy in clinical use, and for its successful application to spinal muscular atrophy.

Dr. Krainer studies the mechanisms of RNA splicing, ways in which they go awry in disease, and the means by which faulty splicing can be corrected. Dr. Krainers research is focused in part on genes associated with spinal muscular atrophy (SMA), a neuromuscular disease that has been the leading genetic cause of death in infants. He worked on antisense approaches to correct mis-splicing, and in collaboration with Ionis Pharmaceuticals and Biogen, developed the first treatment for pediatric and adult SMA.

"I am very grateful and honored to receive this years Ross Prize," said Dr. Krainer. "My trainees and I feel privileged that our research has helped SMA patients. In keeping with the intent of this generous award, we will redouble our efforts to explore new ways to address unmet medical needs."

After a brief award presentation, a symposium will be held during which Dr. Krainer will discuss his research along with Michelle Hastings, PhD, director at Rosalind Franklin University of Medicine and Science, Edward Kaye, MD, CEO of Stoke Therapeutics, and Timothy Yu, MD, PhD, attending physician and assistant professor at Boston Childrens Hospital, who will discuss their latest research.

"Dr. Krainers remarkable discoveries have revolutionized the treatment of a devastating, crippling pediatric illness. His inventions are already giving children the ability to crawl, walk, and live their lives," said Kevin J. Tracey, MD, president and CEO of the Feinstein Institutes and editor emeritus of Molecular Medicine.

Dr. Krainer and his lab have also worked to shed light on the role of splicing proteins in cancer, particularly breast cancer, and on fundamental mechanisms of splicing and its regulation.

Past recipients of the Ross Prize are: Daniel Kastner, MD, PhD, the National Institutes of Healths (NIH) National Human Genome Research Institute (NHGRI) scientific director; Huda Y. Zoghbi, MD, professor, Departments of Pediatrics, Molecular and Human Genetics, Neurology and Neuroscience at Baylor College of Medicine; Jeffrey V. Ravetch, MD, PhD, the Theresa and Eugene M. Lang Professor and head of the Leonard Wagner Laboratory of Molecular Genetics and Immunology at The Rockefeller University; Charles N. Serhan, PhD, DSc, director of the Center for Experimental Therapeutics and Reperfusion Injury at Brigham and Womens Hospital, the Simon Gelman Professor of Anaesthesia at Harvard Medical School and professor at Harvard School of Dental Medicine; Lewis C. Cantley, PhD, the Meyer Director of the Sandra and Edward Meyer Cancer Center at Weill Cornell Medical College and New York-Presbyterian Hospital; John J. OShea, MD, scientific director at the National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS); and Dan R. Littman, MD, PhD, the Helen L. and Martin S. Kimmel Professor of Molecular Immunology in the Skirball Institute of Biomolecular Medicine at New York University School of Medicine.

Story continues

To learn more about the Ross Prize celebration and symposium, and to register for the event, please visit http://www.nyas.org/RossPrize2020. If you would like to nominate a candidate for the 2021 Ross Prize, please make a submission here.

About the Feinstein Institutes

The Feinstein Institutes for Medical Research is the research arm of Northwell Health, the largest health care provider and private employer in New York State. Home to 50 research labs, 2,500 clinical research studies and 5,000 researchers and staff, the Feinstein Institutes raises the standard of medical innovation through its five institutes of behavioral science, bioelectronic medicine, cancer, health innovations and outcomes, and molecular medicine. We make breakthroughs in genetics, oncology, brain research, mental health, autoimmunity, and are the global scientific leader in bioelectronic medicine a new field of science that has the potential to revolutionize medicine. For more information about how we produce knowledge to cure disease, visit feinstein.northwell.edu.

About Molecular Medicine

Molecular Medicine sits at the forefront of its field, rapidly disseminating discovery in the genetic, molecular, and cellular basis of physiology and disease across a broad range of specialties. With over two decades of experience publishing to a multidisciplinary audience, and continually celebrating innovation through the Ross Prize in Molecular Medicine and Anthony Cerami Award in Translational Medicine, the journal strives towards the design of better molecular tools for disease diagnosis, treatment, and prevention. Molecular Medicine is published by BMC, part of Springer/Nature, in partnership with The Feinstein Institutes for Medical Research.

About the New York Academy of Sciences

The New York Academy of Sciences is an independent, not-for-profit organization that since 1817 has been committed to advancing science, technology, and society worldwide. With more than 20,000 members in 100 countries around the world, the Academy is creating a global community of science for the benefit of humanity. The Academy's core mission is to advance scientific knowledge, positively impact the major global challenges of society with science-based solutions, and increase the number of scientifically informed individuals in society at large. Please visit us online at http://www.nyas.org.

About Cold Spring Harbor Laboratory

Founded in 1890, Cold Spring Harbor Laboratory (CSHL) has shaped contemporary biomedical research and education with programs in cancer, neuroscience, plant biology and quantitative biology. CSHL has been a National Cancer Institute designated Cancer Center since 1987. Home to eight Nobel Prize winners, the private, not-for-profit Laboratory employs 1,100 people, including 600 scientists, students and technicians. The Meetings & Courses Program annually hosts more than 12,000 scientists. The Laboratorys education arm also includes an academic publishing house, a graduate school and the DNA Learning Center with programs for middle and high school students and teachers. For more information, visit http://www.cshl.edu

View source version on businesswire.com: https://www.businesswire.com/news/home/20200302005570/en/

Contacts

Matthew Libassi516-465-8325mlibassi@northwell.edu

Original post:
Ross Prize Awarded to Cold Spring Harbor Laboratory Professor - Yahoo Finance

Read More...

Society neglecting needs of people with rare diseases, AKU moot told – The News International

Monday, March 9th, 2020

Society neglecting needs of people with rare diseases, AKU moot told

Policymakers and stakeholders must pay greater attention to the challenges posed by rare diseases in Pakistan, said experts at a conference to mark Rare Disease Day 2020 at the Aga Khan University.

Worldwide, there are over 6,000 diseases classified as rare as they affect fewer than one in 2,000 people. In Pakistan, these diseases are not so rare because a major risk factor is prevalent: inter-family marriages which significantly raise the risk of their children suffering from genetic defects and disorders.

According to a press release issued by the AKU on Friday,

Dr Bushra Afroze, associate professor at the AKU and a clinical geneticist at the universitys teaching hospital, shared the story of Sadia Manzoor*, a girl from a small town in Sindh, to explain how the health care systems shortcomings were affecting those living with rare diseases.

Sadia was eight when the unusual symptoms started to appear. She began to lose her hair, fall over while walking, be inattentive in class and face difficulties in writing. As her problems appeared to be neurological, she was taken to a neurologist and erroneously diagnosed with a non-treatable disorder, preventing her from receiving timely treatment. When she started to experience additional symptoms such as the tendency to repeat words, she was thought to be crazy -- leading to more distress for her and her family.

Thankfully, her parents continued to feel that something was wrong and to search for help. They were finally referred to one of the countrys few genetics specialists in Karachi to learn that their daughter has remethylation defect in vitamin B12, a serious metabolic disorder. Sadia was prescribed disease-specific orphan drugs medication for a condition so rare that it is not feasible to mass produce that are very expensive. Such drugs have to be imported and are often in short supply, which in turn represents an emergency with potentially fatal consequences for Sadias family.

Sadia must have medication daily and will require continual monitoring and management of her condition. Her parents are very focused on helping her live as normal a life as possible with the result that Sadia is a happy child, attending school and enjoying her childhood.

Dr Afroze explained that providing quality care to children such as Sadia requires high-quality system-wide changes that can address several constraints: a lack of awareness; shortage of facilities, expertise and institutions as well as the geographic and economic inequities that people with rare diseases face. She quoted The Lancet Global Health Commission on High Quality Health Systems in the SDG Era 2018 report which stated that providing health services without guaranteeing a minimum level of quality was ineffective, wasteful and unethical.

Currently, there is a range of gaps in the arrangements to provide quality care for patients suffering from rare diseases. Narrowing these gaps will require collaboration between stakeholders across the spheres of research, healthcare, academia and government.

Quality is not a given, Dr Afroze said. Ensuring quality treatment for rare diseases will take vision, planning, investment, compassion, meticulous execution, and rigorous monitoring, from the national level to the smallest, remotest clinic.

As a first step, experts at the conference highlighted the importance of screening newborns for rare diseases. A simple blood test compulsory in China, Canada, the US, as well many countries in Asia and Europe can enable the prompt detection and treatment of such conditions. AKUs Professor Aysha Habib, chair of the conference, explained that tests that can screen for over 50 rare illnesses have been common in the developing world for over 50 years. In Pakistan less than one per cent of newborns are currently being screened for these diseases, since only a handful of private hospitals offer these services, and for a narrow range of just five rare diseases.

She added that more hospitals need to offer screening for rare diseases and called on the government to consider how such services could be scaled up through the public health insurance measures being introduced under the governments Ehsaas programme.

Speakers at the conference also spoke of the role of researchers in the field. At present, there are no national level studies or surveys on the prevalence of rare diseases. While there are a small number of patient registries for specific rare diseases, they exist in silos within hospitals. Data sharing between hospitals would not only enhance the accuracy of information (since a single patient travels to multiple doctors and could be recorded more than once) but would also help form a roster of patients that would enable treatment options to be explored through clinical trials. Efforts by the federal health ministry are also needed to support and streamline registries, speakers added.

Professor Habib noted that academics, clinicians and researchers need to create partnerships that would intensify the development of knowledge and skills in the field. She also highlighted the importance of patient advocacy groups in facilitating research and in fostering synergies between stakeholders.

Collaboration enables everyone to benefit from each others strengths, Professor Habib said. The challenge posed by rare diseases requires us to make the most of our existing resources while developing national and international partnerships that can meet the complex needs of those living with rare diseases that are currently being neglected.

The conference Reframe Rare in Pakistan: Breaking Silos and Bringing Synergies was preceded by two days of workshops that brought together genetic researchers, pathologists and child health specialists from public and private sector organisations across the country.

Other speakers at the event included Professor Shahid Mahmood Baig, head of Human Molecular Genetics at the National Institute for Biotechnology and Genetic Engineering and Professor Giancarlo La Marca, president of the Italian Society for Newborn Screening and Metabolic Diseases.

* The patients name has been changed to protect her identity.

View original post here:
Society neglecting needs of people with rare diseases, AKU moot told - The News International

Read More...

Solution for a scourge? University of Minnesota scientist is progressing with carp-killer tool – Minneapolis Star Tribune

Monday, March 9th, 2020

Sam Erickson followed his love of science to outer space one summer during an internship at NASA. He came away fascinated by seeing into deep space by interpreting interaction between matter and infrared radiation.

Now a full-fledged researcher at the University of Minnesotas College of Biological Sciences, the 25-year-old Alaska native is immersed in something far more earthly: killing carp. His fast-moving genetic engineering project is drawing attention from around the country as a potential tool to stop the spread of invasive carp.

I want to make a special fish, Erickson said in a recent interview at Gortner Laboratory in Falcon Heights.

In short, he plans to produce batches of male carp that would destroy the eggs of female carp during spawning season. The modified male fish would spray the eggs as if fertilizing them. But the seminal fluid thanks to DNA editing would instead cause the embryonic eggs to biologically self-destruct in a form of birth control that wouldnt affect other species nor create mutant carp in the wild.

His goal is to achieve the result in a controlled setting using common carp. From there, it will be up to federal regulators and fisheries biologists to decide whether to translate the technology to constrain reproduction of invasive carp in public waters.

What were developing is a tool, Erickson said. If we could make this work, it would be a total game-changer.

Supervised by University of Minnesota assistant professor Michael Smanski, Erickson recently received approval to accelerate his project by hiring a handful of undergraduate assistants. He also traveled last month to Springfield, Ill., to present his research plan to the 2020 Midwest Fish and Wildlife Conference.

Were pretty excited about where his project is at, said Nick Phelps, director of the Minnesota Aquatic Invasive Species Research Center at the U. Things are sure moving fast. Theres excitement and caution.

Ericksons research has received funding from Minnesotas Environment and Natural Resources Trust Fund. No breeding populations of invasive carp have been detected in Minnesota, but the Department of Natural Resources has confirmed several individual fish captures and the agency has worked to keep the voracious eaters from migrating upstream from the lower Mississippi River. Silver carp, bighead carp and other Asian carps pose a threat to rivers and lakes in the state because they would compete with native species for food and habitat.

Erickson views his birth control project as one possible piece in the universitys integrated Asian carp research approach to keep invasive carp out of state waters. Already the DNR has supported electric barriers and underwater sound and bubble deterrents at key migration points. Another Asian carp-control milestone was closing the Mississippi River lock at Upper St. Anthony Falls in Minneapolis in 2015.

Shooting star

Growing up in Anchorage, Erickson had never heard of Macalester College in St. Paul. But he visited the campus at the urging of a friend and felt like he fit in. He majored in chemistry and worked for a year at 3M in battery technology. But his interests tilted toward the natural world and how to better live in cooperation with nature, he said. Erickson met with Smanski about research opportunities at the university and was hired on the spot.

Smanski, one of the universitys top biological engineers, said carp is not an easy organism to work with and Erickson lacked experience in the field. But he hired the young researcher and assigned him to the carp birth control project because he seemed to have a rare blend of determination and intelligence.

I could tell right away when I was talking to him that he was like a shooting star, Smanski said. If you set a problem in front of him, he wont stop until he solves it Hes taken this farther than anyone else.

In two short years, Smanksi said, Erickson has mastered genetic engineering to the point that his research is starting to bear fruit.

With his new complement of research assistants, Erickson aims to clear his projects first major hurdle sometime this year. The challenge is to model his experiment in minnow-sized freshwater zebrafish. The full genetic code of zebrafish like common carp is already known.

Ericksons task is to make a small change to the DNA sequence of male zebrafish, kind of like inserting a DNA cassette into the fish, he said. During reproduction, the alteration will create lethal overexpression of genes in the embryonic eggs laid by females.

By analogy, Erickson said, the normal mating process is like a symphony with a single conductor turning on genes inside each embryo, Erickson said. But the DNA modification sends in a mess of conductors and the mixed signals destroy each embryo within 24 hours.

In the lab we have to make sure were causing the disruption with no off-target effects, he said. If we can do this in zebrafish, we hope to translate it. They are genetically similar to carp.

Ericksons upcoming experimentation with tank-dwelling live carp could be painfully slow because the fish only mate once a year. But hes working his way around that problem by altering lighting conditions and changing other stimuli in his lab to stagger when batches of fish are ready to reproduce.

The birth control process projected to be affordable for fisheries managers if it receives approval is already proven to work in yeast and insects. And Erickson said the same principles of molecular genetics have been used to create an altered, fast-growing version of Atlantic salmon approved for human consumption in the U.S.

Were not building a new carp from the bottom up but its kind of a whole new paradigm, so we have to get it done right, he said.

Read more here:
Solution for a scourge? University of Minnesota scientist is progressing with carp-killer tool - Minneapolis Star Tribune

Read More...

Connecting interferon, neuroinflammation and synapse loss in Alzheimer’s disease – Baylor College of Medicine News

Monday, March 9th, 2020

When immunologist Dr. Wei Cao joined Baylor College of Medicine three-and-a-half years ago, her first project was to investigate how inflammation contributes to Alzheimers disease.

Alzheimers is the most common cause of dementia among older adults. The current understanding is that, in addition to having beta-amyloid plaques and tau protein tangles, the brains of patients with this condition have a marked inflammatory response, and that this inflammation might be more of a problem than protein aggregation itself, said Cao, associate professor of molecular and human genetics and the Huffington Center on Aging at Baylor.

Inflammation in Alzheimers disease involves the activation of two types of cells in the brain: the resident immune cells called microglia, and astrocytes, star-shaped cells that support neuronal functions. In addition, there are elevated levels of cytokines, molecules that are produced by immune cells to promote inflammation. But the question remained, how does chronic inflammation in brains with Alzheimers disease lead to neuronal dysfunction and the consequent neurodegeneration and dementia?

Amyloid plaques in the brains of people with Alzheimers disease have a heterogeneous composition; for instance, some may also contain sugars, lipids or nucleic acids. Previously, Cao and her colleagues found that amyloid fibrils with nucleic acids, but not those without them, triggered immune cells in the blood to produce type 1 interferon (IFN). IFN is a potent cytokine produced when immune cells sense nuclei acids, such as those that come from viral particles, in their environment. IFN triggers a beneficial inflammatory response that is the first line of defense against viral infections.

While it is best known for its ability to induce an antiviral state in cells, IFN is also involved in immune modulation and tissue damage associated with infectious, autoimmune and other conditions. But, until now, IFN has not been explicitly implicated in Alzheimers disease, Cao said.

In this project, we focused on what was going on in brains with Alzheimers disease, said Dr. Ethan R. Roy, a graduate student in the Graduate Program in Translational Biology and Molecular Medicine at Baylor while he was working on this project. We began by investigating whether microglia from the brain were able to respond to the amyloid/nucleic acid combination by producing IFN.

Roy looked at multiple Alzheimers mouse models in the lab of Dr. Hui Zheng, professor of molecular and human genetics and director of the Huffington Center on Aging, who also is co-principal investigator of the study. Roy found that almost all the animals brains in these models had plaques containing nucleic acids. The composition of these plaques had not been well characterized before, Roy said.

Interestingly, Cao, Roy and their colleagues found that the same mouse brains that had amyloid plaques with nucleic acids also showed a molecular signature mimicking an antiviral IFN response. Further experiments revealed that nucleic acids in the plaques activated brain microglia, which produced IFN that in turn triggered a cascade of inflammatory reactions that led to the loss of synapses, the junctions between neurons through which they communicate. Synapse loss is a key part of neurodegeneration and can lead to memory loss and eventually dementia.

It is well known that synapse loss is directly mediated by the complement system, which is part of the immune system. It comprises a group of proteins that work together to clear microbes and damaged cells, but it also is involved in inflammation.

Although we knew that complement activation triggered synapse loss, what we discovered was the chain of events that led to this outcome. The chain of events points to IFN-mediated pathways controlling complement activation, Cao said.

We were very excited to find that blocking the IFN-triggered cascade of reactions significantly dampened microglia activation and reduced synapse loss in our mouse models, further supporting the leading role of IFN in this process, Roy said.

This study provides a major advance in the understanding of a process that leads to neuronal damage, by connecting IFN, complement and synapse loss: IFN controls the expression of multiple components of the complement cascade and mediates synapse elimination in a complement-dependent manner.

The researchers looked into human brains with Alzheimers disease to see if they presented with characteristics that were similar to those they had observed in mouse models of the condition.

We found that human brains with Alzheimers disease have profound activation of the IFN pathway, suggesting that mechanisms similar to the one we found in mice may be involved in neuronal destruction in people with the disease, Roy said. Further studies need to be conducted to evaluate this hypothesis.

This is important because it would lead to a better understanding of how the disease occurs and suggest novel therapies for this incurable disease.

The accumulation of amyloid plaques in human brains is known to poorly correlate with the severity or duration of dementia. There are people without signs of dementia who harbor significant amounts of both amyloid plaques and tau tangles in their brains, but remarkably lack the robust microglial activation and inflammatory response that is associated with loss of synapses and neurons. On the other hand, the brains of people with dementia linked to Alzheimers disease present with amyloid plaques, tau tangles and inflammation that is involved in neurodegeneration.

Our findings in mouse models suggest that it is plausible that plaques that accumulate in Alzheimers disease patients and those in non-demented individuals differ in their content of nucleic acids. It is thus of great interest to examine more closely the molecular constituents of amyloid plaques in the brains of cognitively resilient individuals and compared them to those of Alzheimers disease cases, Cao said.

This work also may provide new insights into the aging brain. Other work has shown that IFN also seems to participate in the normal aging process of the brain. Cao, Roy and their colleagues think it is also worthwhile to further explore the possibility of modulating IFN activity in aging populations.

Interested in all the details of this study? Find them in the Journal of Clinical Investigation.

Other contributors to this work include Baiping Wang, Ying-wooi Wan, Gabriel Chiu, Allysa Cole, Zhuoran Yin, Nicholas E. Propson, Yin Xu, Joanna L. Jankowsky, Zhandong Liu, Virginia M.-Y. Lee, John Q. Trojanowski, Stephen D. Ginsberg, Oleg Butovsky and Hui Zheng. The authors are affiliated with one or more of the following institutions: Baylor College of Medicine; Harvard Medical School; University of Pennsylvania School of Medicine; Nathan Kline Institute, N.Y., and New York University Langone Medical Center.

The study was funded by NIH grants AG05758, AG032051, AG020670, AG054111, NS092515, AG051812 and AG054672. Further support was provided by the Robert A. and Rene E. Belfer Family Foundation, BrightFocus ADR A20183775, Cure Alzheimers Fund and TBMM T32 training grant (ST32GM088129).

By Ana Mara Rodrguez, Ph.D.

Read the original:
Connecting interferon, neuroinflammation and synapse loss in Alzheimer's disease - Baylor College of Medicine News

Read More...

How SARS-CoV-2 Tests Work and Whats Next in COVID-19 Diagnostics – The Scientist

Monday, March 9th, 2020

The quick sequencing of the SARS-CoV-2 genome and distribution of the data early on in the COVID-19 outbreak has enabled the development of a variety of assays to diagnose patients based on snippets of the viruss genetic code. But as the number of potential cases increases, and concerns rise about the possibility of a global pandemic, the pressure is on to enable even faster, more-accessible testing.

Current testing methods are considered accurate, but governments have restricted testing to central health agencies or a few accredited laboratories, limiting the ability to rapidly diagnose new cases, says epidemiologist and immunologist Michael Mina, the director of the pathology laboratory and molecular diagnostics at Brigham and Womens Hospital in Boston. These circumstances are driving a commercial race to develop new COVID-19 tests that can be deployed within hospitals and clinics to provide diagnostic answers in short order.

Globally, nearly 89,000 cases have now been reportedmore than 80,000 of these in Chinaalong with more than 3,000 deaths. The virus has been found in 64 countries, six of those in just the past day.

The full genome of the novel coronavirus was published on January 10 of this year, just weeks after the disease was first identified in Wuhan, China. A week later, a group of researchers led by German scientists released the first diagnostic protocol for COVID-19 using swabbed samples from a patients nose and throat; this PCR-based protocol has since been selected by the World Health Organization (WHO).

Not all countries have adopted the WHOs recommended diagnostic, including the US.

The assay was initially developed from genetic similarities between SARS-CoV-2 and its close relative SARS, and later refined using the SARS-CoV-2 genome data to target viral genes unique to the newly discovered virus. In particular, the test detects the presence of SARS-CoV-2s E gene, which codes for the envelope that surrounds the viral shell, and the gene for the enzyme RNA-dependent RNA polymerase.

Yvonne Doyle, the medical director and the director of health protection for Public Health England, tells The Scientist in an email that once a sample is received by a laboratory, it takes 2448 hours to get a result. Commenting on the tests accuracy, she says all the positive results to date in the United Kingdom, a total of 36 so far, have been confirmed with whole genome sequencing of the virus isolated from patient samples, and the analytical sensitivity of the tests in use is very high.

This approach also underpins COVID-19 laboratory testing in Australia, where 27 cases have so far been diagnosed, says medical virologist Dominic Dwyer, the director of public health pathology for NSW Health Pathology at Westmead Hospital in Sydney. We decided in the end to have a screening approach using the WHO primers that target the so-called E gene of the coronavirus, he says. If a screening test is positive, we then do some confirmatory testing which selects other targets of the virus genome.

The laboratory at Westmead Hospital also does a complete sequencing of every virus sample to look for possible new strains of SARS-CoV-2 and has shared some of those sequences in the international Global Initiative on Sharing All Influenza Data (GISAID) database for other researchers to study. The staff also cultures the virus and images it using electron microscopy. Thats not really a diagnostic test, but gives you some confirmation of what youre seeing in the laboratory, Dwyer says.

He adds that, so far, theres no suggestion of false positive findings, because every positive test has been confirmed with whole genome sequencing, viral culture, or electron microscopy. As for false negatives, he adds, it would be hard to know if any infected patients were mistakenly given the all-clear.

Not all countries have adopted the WHOs recommended diagnostic. The US Centers for Disease Control and Prevention (CDC), for instance, has developed its own assay that looks for three sequences in the N gene, which codes for the nucleocapsid phosphoprotein found in the viruss shell, also known as the capsid. The assay also contains primers for the RNA-dependent RNA polymerase gene. Dwyer says that the principles of testing are the same; its just the genetic targets that vary.

Mina says its not clear why the CDC chose to develop a different assay to that selected by the WHO and taken up by other countries. Was this actually based on superior knowledge that the CDC had, or was this more of an effort to just go our own route and have our own thing and feel good about developing our own test in the US versus the rest of the world? says Mina, who is also assistant professor of epidemiology at the Harvard School of Public Health. The CDC declined to respond to questions from The Scientist.

In the UK, testing for COVID-19 is being done by a range of accredited laboratories across the country. In the US, all laboratory testing for COVID-19 has until recently been done exclusively by the CDC. The turnaround time for a result has been 2472 hours. Mina argues that enabling hospitals to conduct their own on-site diagnostics could speed up the process. For instance, hospitals can generate flu results within an hour, Mina says, most commonly using assays that detect viral antigens. We spend a lot of money getting rapid turnaround tests in the hospital for flu, for example, because we have to know how to triage people.

The day or two or three that it takes to get COVID-19 results has had logistical ramifications for hospitals, Mina says. If we have a patient who we only suspect is positive, even if they are not positive, just the suspicion alone will lead us to have to find an isolation bed for them, he says.

There has been a move by the CDC to send out RT-PCR test kits to state health laboratories, says Molly Fleece, an infectious diseases physician at the University of Alabama at Birmingham. Hopefully, more laboratories around the country will be able to have access to these testing kits and be able to test specimens instead of having to send all the specimens to the CDC for testing, she says.

However, that plan hit a snag recently when one of the CDC kits reagents was found to be faulty. The agency has announced that the reagent is now being remanufactured.

There are now numerous companies working on commercial test kits in response to the rising diagnostic demands of the epidemic. Most are applying the same real-time PCR methods already in use, but others are taking a different approach. For instance, Mina and colleagues are trialling a diagnostic in partnership with Sherlock Biosciences, based in Cambridge, Massachusetts. The researchers are using CRISPR technology to tag the target SARS-CoV-2 sequences with a fluorescent probe.

Were not at that stage yet of rolling out the serology or antibody tests.

Dominic Dwyer,NSW Health Pathology at Westmead Hospital

In many ways its similar to real-time PCR but its just more sensitive and much more rapid, Mina says. Another CRISPR-based diagnostic protocol developed by researchers at the McGovern Institute at MIT uses paper strips to detect the presence of a target virus, and claims to take around one hour to deliver the result. It has not yet been tested on COVID-19 patient samples, and the institute has stressed the test still needs to be developed and validated for clinical use, for COVID-19 or any other viral disease. Meanwhile, Anglo-French biotech company Novacyte has announced the release of its real-time PCR diagnostic kit for COVID-19, which it says will deliver results in two hours.

A different diagnostics approach would be to devise blood tests for antibodies against the SARS-CoV-2 virus, a development that Mina says will be an important next step for monitoring the spread of the virus. Could we just start taking blood samples from people around the world and see how many people who had no symptoms or very minimal symptoms may have actually been exposed to this? Mina asks.

Dwyer says such approaches could help detect any false negatives that slip through the PCR-based protocols, but were not at that stage yet of rolling out the serology or antibody tests. Numerous groups are trying to isolate antibodies, some with more success than others. Researchers at Duke-NUS Medical School in Singapore have used antibody testing to demonstrate a link between two separate clusters of infections, and in patients who had cleared their symptoms at the time they were given the antibody test. Meanwhile, researchers in Taiwan are also working to identify a SARS-CoV-2 antibody that could be used for diagnostic testing, and they say such a test could deliver a result in a matter of minutes rather than hours.

Bianca Nogrady is a freelance science writer based in Sydney, Australia.

Visit link:
How SARS-CoV-2 Tests Work and Whats Next in COVID-19 Diagnostics - The Scientist

Read More...

Harvard and Guangzhou Institute of Respiratory Health Team to Fight SARS-CoV-2 – Harvard Magazine

Monday, March 9th, 2020

Ever since the earliest reports of a pneumonia-like illness spreading within Hubei province in China, the resemblance to the SARS outbreak of 2002-2003 has been uncanny: probable origins in the wild-animal markets of China; an illness that in some people resembles the common cold or a flu, but in others leads to pneumonia-like symptoms that can cause respiratory failure; community transmission that often occurs undetected; super-spreader events; and reported vertical transmission in high-rises or other living spaces where the waste systems are improperly engineered or drain catch-basins are dry, allowing aerosolized particles to pass from one floor of a building to another (see The SARS Scare for an in-depth description of the epidemiology and virology of the SARS outbreak of 2002-2003 and the four independent zoonotic transmissions of 2003-2004).

UPDATED 3-04-2020 at 12:57p.m. See below.

At first, this latest outbreak was referred to as a novel coronavirus, then in the media as COVID-19 (formally, the name for the disease in an infected person who has become sick, a distinction analogous to that between a person who is HIV positive and one who has developed AIDS). Now that the virus has been characterized and its relationship to SARS firmly established, its designation is SARS-CoV-2severe acute respiratory syndrome coronavirus 2.

Will public-health measures be sufficient to contain its spread? How infectious is it? What is the incubation period? Is this a pandemic? What role does the immune-system response play in the progression of the disease? Which populations are most at risk? Can scientists develop a vaccine, and how quickly? These are some of the questions that scientists worldwide are asking, and that a collaboration among Harvard University and Chinese researchers will address as part of a $115-million research initiative funded by China Evergrande Group, which has previously supported Universitygreen-buildings research at the Graduate School of Design, research onimmunologic diseases, and work inmathematics. (See below for the University press release describing the initiative.)

Harvard Magazinespoke with some of the researchers involved in fighting the first SARS outbreak, and those who will be collaborating with Chinese colleagues, in what is already a worldwide effort to control SARS-CoV-2.

Michael Farzan 82, Ph.D. 97, who in 2002 was an assistant professor of microbiology and molecular genetics at Harvard Medical School (HMS) studying the mechanism that viruses use to enter cells, was the first person to identify the receptor that SARS used to bind and infect human cells. SARS-CoV-2 is a close cousin to SARS, and uses the same human receptor, ACE2, reports Farzan, who is now co-chair of the department of immunology and microbiology at Scripps Research. The ACE2 receptor is expressed almost exclusively in the lungs, gastrointestinal tract, and the kidneys, which explains why the disease is so effectively transmitted via both the respiratory and fecal-oral routes.

But there are subtle differences in the new virus behind the current outbreak, he explained in an interview. The viruss receptor binding domainthe part that attaches to the human receptorhas undergone a lot of what we call positive selection, meaning there has been a good deal of evolutionary pressure on that region from natural antibodies, probably in bats or some other animal host that is a reservoir for this disease. So while the virus retains its ability to bind ACE2, Farzan explains, it no longer binds the same antibodies. That is unfortunate, because as the first SARS epidemic wound down, HMS professor of medicine Wayne Marasco had identified a single antibodyfrom what was then a 27-billion antibody librarythat blocked the virus from entering human cells. (Marasco is actively testing new antibodies, hoping to find one that will have the same effect on SARS-CoV-2. For more on Marascos work, see below.) Still, we are not starting from square one, says Farzan.

In animal studies,Remdesivir [a new and experimental antiviral drug] has seemed to work against SARS-like viruses, he says. Its effectiveness will probably hinge on getting it early enough, in the same way that the antiviral drug Tamifluis most effective against the seasonal flu when given to patients early in the course of infection.

And there is a reasonable hope that a vaccine canbe developed, Farzan adds, because the part of the virus that binds the human receptor is exposed and accessible, making it vulnerable to the immune systems antibodies. In addition, the viral genome is relatively stable. That means SARS CoV-2 wont evolve much over the course of an epidemic, so a vaccine that is relatively protective at the beginning of an epidemic will remain effective until its end.

Another reason for optimismdespite the long road to deploying any vaccine in humansis that the science that allows researchers to understand the viruss structure, life cycle, and vulnerabilities is progressing far more rapidly today than during the first SARS outbreak 17 years ago. So, too, is the understanding of the human immune response to the virus, and of the most effective public-health strategies based on the epidemiology of the disease.

When epidemiologists assess the severity of an epidemic, they want to know how effectively the disease can propagate in a population. The first measure they attempt to calculate is the reproductive number (R0)the number of people that an infected individual will in turn infect in an unexposed population, in the absence of interventions. When the reproductive number is greater than 1 (meaning each infected person in turn infects more than one other person), more and more people become infected, and an epidemic begins. Public-health interventions are therefore designed to lower the rate of transmission below 1, which eventually causes the epidemic to wind down. The second number epidemiologists focus on is the serial intervalhow long it takes one infected person at a particular stage of the disease to infect another person to the point of the same stage of the disease. The serial interval thus suggests how rapidly the disease can spread, which in turn determines whether public-health officials can identify and quarantine all known contacts of an infected individual to prevent their retransmitting the disease to others.

Epidemiologist Marc Lipsitch will be one of several Harvard scientists collaborating with Chinese colleagues to fight SARS-CoV-2Photograph by Kent Dayton

Marc Lipsitch, a professor of epidemiology at the Harvard Chan School of Public Health (HSPH), and director of the schoolsCenter for Communicable Disease Dynamics, helped lead one of the two teams that first calculated the reproductive number of SARS in the 2002-2003 outbreak. SARS had an R0 of 3, he recalls: each case led to three others. In that outbreak, about 10 percent of those who became sick died. The good news is that SARS CoV-2 appears to have a much lower R0 than SARS, ranging from the high ones to low twos, and only 1 percent to 2 percent of those who become sick have died. On the other hand, the serial intervalstill being worked outappears to be shorter, meaning the new virus has the potential to spread faster.

In the current epidemic, Lipsitch notes a further concern: the fact that the incubation-period distribution and the serial-interval distribution are almost identical. Thats a mathematical way of saying that people can start transmitting the virus even when they are pre-symptomatic, or just beginning to exhibit symptoms. That makes tracing and quarantining contacts of infected individualsa classic, frontline public-health measurenearly impossible.

Tracing, quarantining, and other public-health interventions, such as distancing measures (closing workplaces or asking employees to work from home, for example) proved sufficient to defeat SARS in the early 2000s. But with SARS-CoV-2, public-health measures alone may prove inadequate. Controlling this version of SARS may require antivirals, stopgap antibody therapies, and ultimately, vaccines, deployedtogetherwith robust public-health containment strategies.

Unfortunately, SARS-CoV-2 is almost certainly already a pandemic, Lipsitch continues: demonstrating sustained transmission in multiple locations that will eventually reach most, if not all places on the globe. The disease appears to be transmitting pretty effectively, probably in Korea, probably in Japan, and probably in Iran. He now estimates that 20 to 60 percent [figures updated 03-04-2020 at 12:57 p.m.]of the adult global population will eventually become infected.

That said, Infected is different from sick, he is careful to point out. Only some of those people who become infected will become sick. As noted above, only about 1 percent to 2 percent of those who have becomesickthus far have died, he says. But the number of people who areinfectedmay be far greater than the number of those who are sick. In a way, he says, thats really good news. Because if every person who had the disease was also sick, then that would imply gigantic numbers of deaths from the disease.

I'm very gratified, Lipsitch continues, to see that both China and Harvard recognize the complementarity between public health and epidemiology on the one hand, and countermeasure-development on the other hand. We can help target the use of scarce countermeasures [such as antivirals or experimental vaccines] better if we understand the epidemiology; and we will understand the epidemiology better if we have good diagnostics, which is one of the things being developed in this proposal. These approaches are truly complementary.

In the short term, Lipsitchwho has sought to expand the modeling activities of the Center for Communicable Disease Dynamics to better understand the current outbreaks epidemiologysays, It would be great toexpand collaborations with Chinese experts. Longer term, I see a really good opportunity for developing new methods for analyzing data better, as we have in previous epidemics. After the first SARS outbreak, for example, epidemiologists developed software for calculating the reproductive number of novel diseases; that software now runs on the desktop computers of epidemiologists around the world. And in 2009, during an outbreak of swine flu in Mexico, Lipsitch and others developed a method for using the incidence of the disease among awell-documented cohort of travelerswho had left Mexico, to estimate the extent of the disease among amuch larger and less well surveyedpopulation of Mexican residents.

What they found then was that the estimated number of cases in Mexican residents likely exceeded the number of confirmed cases by two to three orders of magnitude. The same method is being used to assess the extent of SARS-CoV-2 in China right nowso far without any hiccups. In the Mexican case, Lipsitchreports, the estimates suggested that severe cases of the disease were uncommon, since thetotal numberof cases was likely much larger than the number ofconfirmedcases. So I think we have learned from each epidemic how to do more things. And in between them, you solidify that less visible, less high-profile research that builds the foundation for doing better the next time. His group, for example, has been developing ways to make vaccine trials faster and better once a vaccine candidate exists.

A vaccine is the best long-term hope for controlling a disease like SARS-CoV-2. Higgins professor of microbiology and molecular genetics David Knipe, who like Lipsitch will participate in the newly announced collaboration, works on vaccine delivery from a molecular perspective. Knipe has developed methods to use the herpes simplex virus (HSV) as a vaccine vector and has even made HSV recombinants that express the SARS spike proteinthe part of the virus that binds the human ACE2 receptor. He now seeks to make HSV recombinants that express the new coronavirus spike protein as a potential vaccine vector.

But Knipe also studies the initial host-cell response to virus infection, which is sometimes called the innate immune response. And he has used HSV vectors that expressed the first SARS spike protein to study how it activates innate immune signaling. That is important because inSARS 1, initial symptoms lasted about a week, but it was the second phasecharacterized by a massive immune-system response that began to damage lung tissuethat led to low levels of oxygen saturation in the blood, and even death.The inflammation in the lungs is basically a cytokine storm, an overwhelming and destructive immune response thats the result of innate signaling, Knipe explains. So were going to look at that with the new coronavirus spike protein, as well. This could help to determine the actual mechanism of inflammation, and then we can screen for inhibitors of that that might be able to alleviate the disease symptoms.

The idea, he says, is to stop theinflammatoryresponse now killing people in the respiratory phase of the disease by targeting the specific molecular interaction between the virus and the host cell. This, he explains, aligns with one of the principal initial goals of the collaboration, which is to support research both in China and at Harvard to address the acute medical needs of infected individuals during the current crisis.

Another form of frontline defense against the virus is antibody therapy. In an epidemic, this type of therapy is usually administered as a prophylaxis to first responders at high risk of infection, or as treatment to patients who are already sick or to people who might be harmed by a vaccine, such as pregnant women, the elderly, or those with co-morbidities. Wayne Marasco, an HMS professor with a lab at the Dana Farber Cancer Institute, was the first to develop antibody therapies against SARS and MERS, a related coronavirus, in 2014. What he learned in those outbreaks was that using only a single antibody to bind the viruss receptor binding domainthe part of the virus that attaches to the human receptoris not enough to prevent escape through mutations that neutralize the therapy. You have to use combinations of antibodies to block the escape pathways, he explains. But the combinations have to be carefully designed to avoid the risk that the virus will evolve a gain of functionor the virus coming out of the patient is more pathogenic than the virus you started to treat.

During the MERS outbreak, Marasco led the Defense Advanced Research Projects Agencys 7-Day Biodefense program.DARPA would drop an unknown pathogen off at our doorstep, Marasco says, and we had seven days to develop a therapeutic that could be manufactured at scale. A second DARPA-funded project focused on reducing the cost of therapies to less than $10 a dose. The government has made efforts to streamline that process to get the production sped up and the cost decreased, he notes, although the efforts are independent of regulatory approval, which has a life of its own.

Marasco currently collaborates with an international team that can perform studiesincluding some that cant be done at Harvardthanks to ready access to a Biosafety Level 4 laboratory and to non-human primates for testing. The team is working to develop antibody therapies effective against SARS-CoV-2, but Marasco cautions that the situation is pretty worrisome with a disease that has a long latency period when people show no symptoms, and when public-health officials cannot identify source cases (as in Italy and in the single case of apparent community transmission in California reported February 26).

The problem in getting ahead of this now, he continues, is funding. Government resources are generally a redistribution of funds that have previously been granted to projects such as the Ebola outbreak in West Africa, or come as administrative supplements to preexisting grants. But with the pace of this epidemic, a lack of resources is limiting what can get done and how quickly it can be accomplished. Beyond the creation of therapeutics, there are all kinds of epidemiologic considerations that require rapid funding, from investigating modes of transmission to field testing for infection.

In the near term, the way to treat masses of patients, he says, is to take blood plasma from someone who has recovered and administer it to an infected person. The convalescents antibodies then fight the infection. The FDA would never approve it, he notes, but it does work. Ultimately, the treatment of choiceand the most cost-effective approach, he says, will be a vaccine.

In the last days of 2019 and the first days after the New Year, we started hearing about a pneumonia-like illness in China, says Dan Barouch, an HMS professor of medicine and of immunology known for his anti-HIV work, whose lab has developed a platform for rapid vaccine development. (During the Zika virus outbreak of 2016, for example, his group was the first to report, within a month, a vaccine protective in animal models.) When the genome of the virus was released on Friday, January 10, we started reviewing the sequence that same evening, working through the weekend. By Monday morning, we were ready to grow it.

His concern about this latest outbreak was that the rate of spread seemed to be very rapid. In addition, the outbreak had the clinical features of an epidemic. We reasoned that this might make it difficult to control solely by public-health measures, he says, particularly because the virus can be transmitted by asymptomatic individuals. Thus, if the epidemic is still spreading toward the end of this year or early 2021, by which point a vaccine might be available, Barouch explains, such a remedy could prove essential. Historically, when viral epidemics don't self-attenuate, the best method of control is a vaccine.

Although Barouchs Beth Israel Deaconess Medical Center lab is working on DNA and RNA vaccines, a new technology that has the potential to cut vaccine development times in half, large-scale manufacturing using so-called nucleotide vaccines is unproven. That's why I think there needs to be multiple parallel vaccine efforts, he emphasizes. Ultimately, we don't know which one will be the fastest and most protective. At the moment, he reports, there are at least a half dozen scientifically distinct vaccine platforms that are being developed and he believes that vaccine development for this pathogen will probably go faster than for any other vaccine target in human history.

Ever since I graduated from medical school, he points out, there have been new emerging or re-emerging infectious disease outbreaks of global significance with a surprising and disturbing sense of regularity. There is Ebola. There was Zika. There were SARS, MERS; the list keeps growing. With climate change, increasing globalization, increasing travel, and population shifts, the expectation is that epidemics will not go away, and might even become more frequent.

In this global context, Barouch emphasizes the importance of a collaborative response that involves governments, physicians, scientists in academiaandin industry, and public-health officials. It has to be a coordinated approach, he says. No one group can do everything. But I do think that the world has a greater sense of readiness this time to develop knowledge, drugs, and therapeutics very rapidly. The scientific knowledge that will be gained from the vaccine efforts [will] be hugely valuable in the biomedical research field, against future outbreaks, and in the development of a vaccine to terminate this epidemic.

University provost Alan Garber, a physician himself, adds that Global crises of such magnitude demand scientific and humanitarian collaborations across borders. Harvard and other institutions in the Boston area conduct research on diagnostics, virology, vaccine and therapeutics development, immunology, epidemiology, and many other areas.With its tremendous range of expertise and experience, our community can be an important resource for any effort to address a major global infectious disease outbreak. Our scientists and clinicians feel an obligation to be part of a promising collaboration to overcome the worldwide humanitarian crisis posed by this novel virus.

UPDATED 3-03-2020 AT 12:10 p.m.TO INCLUDE A REPORT FROM THE MEETING WITH CHINESE COLLEAGUES

In a closed-door meeting that took place Monday, March 2, 2020, at Harvard Medical School, nearly 80 Boston-area scientists gathered to discuss with colleagues from China participating via video link how to respond to COVID-19 disease and the SARS-CoV-2 virus that causes it. This was the first meeting to take place as a result of the collaboration with scientists at theGuangzhou Institute of Respiratory Health announced on Monday, February 24.In attendance locally were experts from Harvard Medical School (HMS), the Harvard T.H. Chan School of Public Health, the HMS-affiliated hospitals, the Ragon Institute, Boston University, the Broad Institute, MIT, the Wyss Institute, as well as representatives from industry. The workshop, convened by HMS dean George Q. Daley, was a planning session to map out the process for coordinating on collaborative projects, designed to allow the participants to meet, form working groups by research area, and determine next steps.

The collaboration harnesses the strengths of the Boston scientific and biomedical ecosystem, the events organizers said in a statement, with the critical experience of Chinese scientists, who are providing on-the-ground insight into diagnostics and care for patients on the frontlines.

This public health crisis, they continued, is an opportunity to catalyze an unprecedented level of collaboration among various scientific efforts across Boston and Cambridge to address both the acute, most pressing challenges of this particular epidemic but also to establish a framework for future collaborations and create a more nimble rapid-response system for other epidemics.

The meeting was organized according to areas of research interest, need, and opportunity including:

The meeting demonstrated the need to establish a collaborative regional response capacity, not only for this outbreak, but for other future emerging infectious diseases, said the organizers. They are now working to create an organizational structure that will formalize the working groups in each of the above areas, and allow for the optimal deployment of resources including disciplinary and clinical expertise, shared core facilities, and funding.

The official Harvard press release follows:

Harvard University Scientists to Collaborate with Chinese Researcherson Development of Novel Coronavirus Therapies, Improved Diagnostics

At a glance:

Since its identification in December, the novel coronavirus has quickly evolved into a global threat, taking a toll on human health, overwhelming vulnerable health care systems and destabilizing economies worldwide.

To address these challenges, Harvard University scientists will join forces with colleagues from China on a quest to develop therapies that would prevent new infections and design treatments that would alleviate existing ones.

The U.S. efforts will be spearheaded by scientists at Harvard Medical School, led by DeanGeorge Q. Daley, working alongside colleagues from the Harvard T.H. Chan School of Public Health. Harvard Medical School will serve as the hub that brings together the expertise of basic scientists, translational investigators and clinical researchers working throughout the medical school and its affiliated hospitals and institutes, along with other regional institutions and biotech companies.

The Chinese efforts will be led by Guangzhou Institute of Respiratory Health and Zhong Nanshan, a renowned pulmonologist and epidemiologist. Zhong is also head of the Chinese 2019n-CoV Expert Taskforce and a member of the Chinese Academy of Engineering.

Through a five-year collaborative research initiative, Harvard University and Guangzhou Institute for Respiratory Health will share $115 million in research funding provided by China Evergrande Group, aFortuneGlobal 500 company in China.

We are confident that the collaboration of Harvard and Guangzhou Institute of Respiratory Health will contribute valuable discoveries to this worldwide effort, said Harvard University President Lawrence Bacow. We are grateful for Evergrandes leadership and generosity in facilitating this collaboration and for all the scientists and clinicians rising to the call of action in combating this emerging threat to global well-being.

Evergrande is honored to have the opportunity to contribute to the fight against this global public health threat, said Hui Ka Yan, chair of the China Evergrande Group. We thank all the scientists who responded so swiftly and enthusiastically from the Harvard community and are deeply moved by Harvard and Dr. Zhongs teams dedication and commitment to this humanitarian cause. We have the utmost confidence in this global collaborative team to reach impactful discoveries against the outbreak soon.

While formal details of the collaboration are being finalized, the overarching goal of the effort is to elucidate the basic biology of the virus and its behavior and to inform disease detection and therapeutic design. The main areas of investigation will include:

With the extraordinary scale and depth of relevant clinical and scientific capabilities in our community, Harvard Medical School is uniquely positioned to convene experts in virology, infectious disease, structural biology, pathology, vaccine development, epidemiology and public health to confront this rapidly evolving crisis, Daley said. Harnessing our science to tackle global health challenges is at the very heart of our mission as an institution dedicated to improving human health and well-being worldwide.

We are extremely encouraged by the generous gesture from Evergrande to coordinate and supportthe collaboration and by the overwhelmingly positive response from our Harvard colleagues, said Zhong, who in 2003 identified another novel pathogen, the severe acute respiratory syndrome (SARS) coronavirus and described the clinical course of the infection.

We look forward to leveraging each of our respective strengths to address the immediate and longer-term challenges and a fruitful collaboration to advance the global well-being of all people, Zhong added.

Harvard University ProvostAlan M. Garbersaid outbreaks of novel infections can move quickly, with a deadly effect.

This means the response needs to be global, rapid and driven by the best science. We believe that the partnershipwhich includes Harvard and its affiliated institutions, other regional and U.S.-based organizations and Chinese researchers and clinicians at the front linesoffers the hope that we will soon be able to contain the threat of this novel virus, Garber said. The lessons we learn from this outbreak should enable us to respond to infectious disease emergencies more quickly and effectively in the future.

Read the original here:
Harvard and Guangzhou Institute of Respiratory Health Team to Fight SARS-CoV-2 - Harvard Magazine

Read More...

Mutations in top autism gene linked to changes in brain structure – Spectrum

Monday, March 9th, 2020

Close inspection: A neuronal tract that connects the brains hemispheres is absent or thinner in people with TBR1 mutations (bottom) than in controls (top).

People with mutations in a gene called TBR1 have unusual features in several brain regions, along with autism traits and developmental delay, according to a new study1.

TBR1 encodes a protein that is involved in brain development. It controls the expression of several other autism-linked genes that lay out the structure of the cerebral cortex, the brains outer layer and locus of higher cognition.

Studies in mice have suggested that deletion or mutation of TBR1 results in structural abnormalities in the brain2. The layers of the cerebral cortex in these mice may be disorganized, and connections in the amygdala a structure involved in processing emotions may be missing.

However, few studies have examined how TBR1 mutations affect people.

Mutations in the gene have been linked to intellectual disability and autism, but more subtle features have not been well documented, says lead investigator Sophie Nambot, a clinician who specializes in medical genetics at the Centre Hospitalier Universitaire Dijon Bourgogne in France.

Previous research documented about 13 people with TBR1 mutations, but the reports included minimal information about outward characteristics, and few include brain-scan data, Nambot says.

The new study adds 25 previously unreported individuals to the literature, along with detailed descriptions of their physical, neurological and genetic features. The findings were published in January in the European Journal of Human Genetics.

It greatly increases the number of individuals and types of mutations in the TBR1 gene that are associated with developmental disorders, says John Rubenstein, professor of psychiatry at the University of California, San Francisco, who was not involved in the study.

Nambots team identified people with TBR1 mutations by contacting national and international health networks, such as GeneMatcher and DECIPHER. They ultimately recruited 25 people, ranging in age from 2 to 29 years, at 22 sites.

Scientists at the centers gathered information about these individuals development and autism traits, such as communication difficulties, lack of eye contact and restricted interests.

All 25 have intellectual disability or moderate-to-severe developmental delays; 19 show autism traits.

The researchers also scanned the individuals genomes to identify small and large mutations in TBR1. Combining their data with those from previous studies, the team identified 29 different single-nucleotide variants.

Magnetic resonance images from seven of the people revealed previously unidentified structural differences in their brains, Nambot says. These differences are in the cerebral cortex; the anterior commissure, a neuronal tract that connects the two hemispheres of the brain; and the hippocampus, an area involved in learning and memory.

In two of the people, the ridges in the cerebral cortex are unusually broad and thick, and in three people, the hippocampus is malformed.

The hippocampal problems are very likely to affect learning and memory, says Robert Hevner, director of neuropathology at the University of California, San Diego. The findings may explain why people with TBR1 mutations have developmental delays, he says. They also support prior studies suggesting that TBR1 affects a type of cell that helps organize the cortex and the hippocampus during fetal development.

Notably, in all seven people, the anterior commissure is thin or absent.

Research in mice demonstrates a similar phenomenon, says Yi-Ping Hsueh, distinguished research fellow in neuroscience at the Institute of Molecular Biology in Taiwan, who was not involved in the new study. In 2014, Hsuehs team found the anterior commissure to be partially missing in mice lacking one copy of TBR1, resulting in social and cognitive problems2.

Unlike mouse studies, however, the new study does not quantify volume differences in brain structures, which makes it difficult to gauge what should be considered atypical, Hsueh says.

The researchers caution that their sample is small, especially given the range of TBR1 mutations present. Still, they say, their findings may help scientists classify genetic subtypes of autism.

Continue reading here:
Mutations in top autism gene linked to changes in brain structure - Spectrum

Read More...

Why Companies Should Consider Genomic Evidence In Defending Toxic Torts – Lexology

Monday, March 9th, 2020

What is Genomics?

Genomics is the study of all the genes within an organism, including humans, and how all of those genes are interrelated and influence the organism. Genetics is primarily focused on single genes. Genomics involves sequencing and analysis of genomes through the use of DNA sequencing and bioinformatics.1

How are Genomics Being Used in Toxic Tort Litigation?

The use of genomics to defend toxic tort claims is emerging. For example, defendants in recent cases involving asbestos, benzene, low-dose radiation and other substances have used genomics successfully. These successes tend to occur after an early and careful look at the facts of a case.

For example, defendants recently and successfully used a genomic causation defense in two mesothelioma cases involving males, who developed peritoneal mesothelioma in their 30s, with little or no known exposure to asbestos fibers. The early age of cancer onset was a material fact suggesting a genetically caused cancer. Another fact suggesting that genomics should be considered is the presence of a family history of cancer, especially early onset cancers or multiple cancers in particular family members. Contrary to what one might assume, the family history does not need to involve the same cancer or the same organ. That is because adverse germline mutations (ones that are inherited) can cause cancers located on different organs.

What can Genomics Revealabout Cancers?

As explained by members of ToxicoGenomica, a multidisciplinary group of scientists and lawyers that provide services for using genomic and systems biology data in civil litigation, the application of Next Generation Sequencing (NGS) technologies in toxic tort cases can reveal the presence or absence of evidence of toxicant exposure and damage that is locked within the genetic blueprint of an individual, and in appropriate cases, can assess alternative causation.

In other words, the process can reveal objective evidence showing that adverse gene mutations were sufficient to cause a cancer without regard to exposure to a particular substance (such as asbestos fibers).

How is the Genomic Information Obtained?

The initial steps in the gene sequencing process are not complex. The first step is to obtain a source from which laboratory technicians can obtain DNA that meets standard criteria. Typically, DNA is either extracted from small samples of fresh blood (about two teaspoons) or from tumor biopsy tissue samples that include some non-cancerous tissue.

In some situations, old biopsy material may serve as a source of DNA, even if the biopsy sample was taken for reasons not involving cancer.

Reasons Why Using Genomic Evidence is a Good Idea.

1) To break up purported class actions. By exposing the material physical differences between plaintiffs, class claims can be defeated.

2) To force plaintiffs to back down. In toxic tort litigation, some of the most financially successful plaintiffs firms use templates for their cases. This allows them to recycle the same theories and defenses with only a modest investigation of the individualized facts. Firms using such a model are disinclined to invest the time and money needed to understand genomics and the genomics of a particular plaintiff. Instead, they may dismiss claims or settle for a relatively small amount.

According to a lawyer affiliated with ToxicoGenomica, we are seeing an increase in cases settling quickly after disclosure of a high quality expert report that proves up the existence of combinations of adverse germline mutations inherited by a person with cancer and a family history of cancer.

3) To dispute exposure. Some forms of genomic analysis can be used to generate data to show whether a plaintiff has or has not been exposed to a particular toxicant. For example, increasing numbers of studies evaluate the presence or absence of exposures based on patterns involving small or large segments of RNA. Studies of this sort are increasingly used to assess genomic changes related to use of tobacco.2

4) To combat the scope of general causation. Genomic evidence can provide previously unknown, but objective evidence, to dispute general long-held beliefs about general causation. For example, for years, juries were unpersuaded by the argument that mesothelioma could be idiopathic, meaning cause unknown. Todays jurors, raised on TV shows such as CSI, expect and want to see objective scientific data. Recently, researchers used new scientific tools (e.g., CRISPR) to create genetically engineered mice that developed mesotheliomas without asbestos exposure when given mutations that promoted general cancer development, supporting the argument that mesotheliomas can be idiopathic or at least not related to asbestos exposure.3 These new genomic and molecular tools, such as CRISPR, are of paramount importance for litigation. It allows researchers to perform studies and experiments that were previously not possible to determine the impact of various substances on humans and even a given individual. Risk managers, lawyers and others do not need to know the nuances of how to use these tools, but they should know of their existence. To read more on this subject, see my three-part Article Using Genetic Evidence to Defend Against Toxic Tort Claims, co-authored with Dr. Whitney Christian and initially printed in the Intellectual Property & Technology Law Journal (2017). https://www.bclplaw.com/images/content/9/9/v2/99117/IP-Reprint-Article-complete.pdf

5) To demonstrate alternative causation. Genomics can provide objective evidence of the presence or absence of adverse germline mutations known to play a causative role in the hundreds of known familial cancer syndromes. Persons who inherit these adverse germline mutations are genetically predisposed to develop cancers and/or other conditions attributable to inherited mutations.4

6) To demonstrate a plaintiffs susceptibility to or resistance to a particular toxicant.

The presence of a protective mutation (an allele) may make it less likely that the substance in question actually caused the disease. For example, in a benzene case, the plaintiffs expert claimed that the plaintiff likely had a genome that made him more susceptible to a disease when benzene was metabolized. The scientists at ToxicoGenomica assessed his relevant genes and found the opposite, that he had a favorable genome that helped his body resist possible adverse effects. After receiving the data generated by ToxicoGenomica, the plaintiffs expert changed his mind and agreed that: 1) the plaintiff was not susceptible and 2) had a robust genome with respect to metabolism of benzene.

7) To contest specific causation. Genomics increasingly can identify somatic signature mutations patterns or non-inherited mutations in tumors. These patterns of somatic mutations increasingly can be used to provide objective evidence showing that a particular toxicant caused the tumor. Extensive somatic mutation pattern analysis of lung cancers in smokers has produced large amounts of data objectively showing that particular lung cancers arose from tobacco smoking instead of something else. In other instances, the pattern may suggest that the tumor arose because of an inherited mutation in a gene such as KRAS.5

8) To tackle specific causation. In some cases, gene expression profiling can be used to determine with more precision whether a persons disease was or was not caused by a specific toxicant. Exposure to certain substances leaves behind genetic fingerprints. If those fingerprints are missing, then it is reasonable to conclude that the toxicant did not cause the disease.

This technique was used successfully in a case involving naturally occurring radioactive materials. The defendants were able to show that the footprints were missing and that the plaintiffs gene expression instead supported that view that she had developed cancer from inherited mutations. 6

Caution for the Defense Attorney. While genomic evidence can help defendants, it can certainly be used to support a plaintiffs claim and provide evidence of both exposure and causation. As a result, it would be wise for counsel to assess carefully whether genomics is appropriate in a given case.

An example of genomics aiding plaintiffs lies in a federal multi-district litigation involving a diabetes drug known as Actos, which was alleged to cause bladder cancers in some people. The MDL court held an extensive Daubert hearing, which included expert opinions on how genomics could provide a logical explanation for why cancers developed in an unusually short time period (less than 1 year). The court decided to admit the genomic evidence, which made plaintiffs claims much more plausible. After that ruling, and after some other trial losses, defendants subsequently agreed to pay over $2 billion in settlements.

View post:
Why Companies Should Consider Genomic Evidence In Defending Toxic Torts - Lexology

Read More...

Supervisory Clinical Laboratory Scientist Job in Fort Leonard Wood, Missouri – Department of the Army – LemonWire

Monday, March 9th, 2020

The United States government is a massive employer, and is always looking for qualified candidates to fill a wide variety of open employment positions in locations across the country. Below youll find a Qualification Summary for an active, open job listing from the Department of the Army. The opening is for a Supervisory Clinical Laboratory Scientist in Fort Leonard Wood, Missouri Feel free to browse this and any other job listings and reach out to us with any questions!

Supervisory Clinical Laboratory Scientist Fort Leonard Wood, MissouriU.S. Army Medical Command, Department of the ArmyJob ID: 222868Start Date: 03/03/2020End Date: 03/16/2020

Qualification SummaryWho May Apply: US Citizens In order to qualify, you must meet the education and experience requirements described below. Experience refers to paid and unpaid experience, including volunteer work done through National Service programs (e.g., Peace Corps, AmeriCorps) and other organizations (e.g., professional; philanthropic; religious; spiritual; community; student; social). You will receive credit for all qualifying experience, including volunteer experience. Your resume must clearly describe your relevant experience; if qualifying based on education, your transcripts will be required as part of your application. Additional information about transcripts is in this document. Basic Requirement for Supervisory Clinical Laboratory Scientist: A. A Bachelors or graduate/higher level degree from a regionally accredited college/university including courses in biological science, chemistry and mathematics, AND successful completion of a Medical Laboratory Scientist/Clinical Laboratory Scientist program accredited by the National Accrediting Agency for Clinical Laboratory Sciences (NAACLS) or an accrediting body recognized by the U.S. Department of Education at the time the degree was obtained. OR B. At least a full 4-year course of study that included 12 months in a college or hospital-based medical technology program or medical technology school approved by a recognized accrediting organization. The professional medical technology curriculum may have consisted of a 1-year post- bachelors certificate program or the last 1 or 2 years of a 4-year program of study culminating in a bachelors in medical technology. OR C. A bachelors or graduate/higher level degree from an accredited college/university that included 16 semester hours (24 quarter hours) of biological science (with one semester in microbiology), 16 semester hours (24 quarter hours) of chemistry (with one semester in organic or biochemistry), one semester (one quarter) of mathematics, AND five years of full time acceptable clinical laboratory experience in Blood Banking, Chemistry, Hematology, microbiology, Immunology and Urinalysis/Body Fluids. This combination of education and experience must have provided knowledge of the theories, principles, and practices of medical technology equivalent to that provided by the full 4-year course of study described in A or B above. All science and mathematics courses must have been acceptable for credit toward meeting the requirements for a science major at an accredited college or university. Acceptable experience is responsible professional or technician experience in a hospital laboratory, health agency, industrial medical laboratory, or pharmaceutical house; or teaching, test development, or medical research program experience that provided an understanding of the methods and techniques applied in performing professional clinical laboratory work. Certification/licensure as a medical technologist (generalist) obtained through written examination by a nationally recognized credentialing agency or State licensing body is a good indication that the quality of experience is acceptable. Evaluation of Education and Experience: The four major areas of clinical laboratory science are microbiology, clinical chemistry, hematology, and immunohematology (blood banking). Qualifying course work in these areas includes bacteriology, mycology, mycobacteriology, tissue culture, virology, parasitology, endocrinology, enzymology, toxicology, urinalysis, coagulation, hemostasis, cell morphology, immunology, serology, immunoserology, immuno-deficiency, hemolysis, histocompatibility, cyto-genetics, and similar disciplines or areas of laboratory practice. Related fields include physiology, anatomy, molecular biology, cell biology, embryology, pathology, genetics, pharmacology, histology, cytology, nuclear medicine, epidemiology, biostatistics, infection control, physics, statistics, and similar areas of science where the work is directly related to the position to be filled. Exemption: You are exempt from the basic requirements above if you are a current federal employee occupying a position in the 0644 occupational series and have been continuously employed in this occupational series since September 27, 2017 or before. Note: You will be required to provide appropriate documentation to the respective Human Resources Office to validate your status. In addition to meeting the basic requirement above, to qualify for this position you must also meet the qualification requirements listed below: Experience required: To qualify based on your work experience, your resume must describe one year of specialized experience which includes providing various verification duties in a laboratory. This definition of specialized experience is typical of work performed at the next lower grade/level position in the federal service (GS-09). OR Education: I have at least two and a half years (45 semester hours) of progressively higher level graduate education leading towards a Ph.D. or equivalent doctoral degree from an accredited college or university that is directly related to the work of the position. OR Combination of Education and Experience: A combination of education and experience may be used to qualify for this position as long as the computed percentage of the requirements is at least 100%. To compute the percentage of the requirements, divide your total months of experience by 12. Then divide the total number of completed graduate semester hours (or equivalent) beyond the first year (total graduate semester hours minus 18) by 18. Add the two percentages. Experience or graduate education must have been in (1) the general field of medical technology; (2) one of the disciplines or specialized areas of medical technology; or (3) a field directly related and applicable to medical technology or the position to be filled. AND Certification: Certification from the American Society for Clinical Pathology (ASCP), American Medical Technologist (AMT) or other board or registry deemed comparable by the Office of the Assistant Secretary of Defense for Health Affairs (OASD(HA)) or their designee as a Medical Technologist (MT) or Medical Lab Scientist (MLS) is required.

If youd like to submit a resume or apply for this position, please contact Premier Veterans at abjobs@premierveterans.com. All are free to apply!

Apply

Post a job on LemonWire. Email jobs@lemonwire.com.Want to advertise on listings like this? Email ads@lemonwire.com.

Continued here:
Supervisory Clinical Laboratory Scientist Job in Fort Leonard Wood, Missouri - Department of the Army - LemonWire

Read More...

Predicting Immunotherapy Success – BSA bureau

Tuesday, February 25th, 2020

Overexpression of certain immunoproteasome subunits in melanoma are tied to anti-cancer immune activity

One of the frustrations with anti-cancer therapy is that no one drug fits all: Most work well in some people but have little effect in other patients with the same type of cancer. This is as true of the newer immunotherapy treatments as it is of older types of chemotherapy.

Weizmann Institute of Science researchers have now identified new markers that can help predict which patients have a better chance for a positive response to immunotherapy treatments. Their findings were reported in Nature Communications.

For Prof. Yardena Samuels and her research group partnered with Prof. Eytan Ruppins lab in the National Institutes of Health (USA), basing their research on the understanding that learning to predict which treatments are most likely to work is the first step to creating a personalized approach to curing cancer. The focus of their work is melanoma a collection of skin cancers that are often hard to treat, and which may be made up of varied tumor cells containing hundreds of different mutations. A certain percentage of melanomas have in recent years been successfully treated with immunotherapy drugs known as checkpoint inhibitors, which work by removing internal obstacles that trip up the bodys own immune system and keep it from attacking the cancer. Unfortunately, for others, these drugs remain ineffective.

To understand the differences in response between different people, the research team, led by postdoctoral fellow Dr. Shelly Kalaora in Samuels group and Joo Sang Li in the Ruppin group, first analyzed data from 470 melanoma patients that has been made available in the Cancer Genome Atlas (TCGA). They were particularly interested in differences in survival rates of patients in whom particular subunits of immunoproteasomes are overexpressed compared to those with low expression of these subunits. These are a variation on the proteasome subunits normally expressed in most cells (except for immune cells). Proteasomes are protein complexes that function as cutters, reducing long proteins to short pieces called peptides. These peptides are later presented on the cells surface by molecules called human leucocyte antigens (HLA). HLA peptides are basically bits of information small molecules displayed on the outsides of cells that report about new threats that the immune system needs to assess and address.

The immunoproteasome is assembled out of altered subunits and thus produces a unique collection of HLA peptides. The team thought that a particular change and overexpression in the HLA peptide repertoire might result in better recognition of the tumor cells by the immune system and thus better elimination of cancer cells.

To test this idea, the researchers cultured lines of tumor cells from melanoma patients in which they overexpressed the immunoproteasome subunits and identified the HLA peptides presented in each situation. By testing the response of the immune cells from the same patients, they showed that the newly formed HLA peptides were indeed more reactive compared to the HLA peptides presented by cells without this overexpression.

Could two subunits the research identified in particular and the unique HLA-bound peptides these produce be a predictor of immunotherapy success? The experiments showed that in tumor cells in which the subunits were overexpressed, the various immune system components that directly fight the cancer were more prevalent and more active than average. Indeed, looking back at the details of cancer patients in the database, the team reported that the expression levels of the two subunits were excellent predictors of the outcome better than the tumor mutational burden, a biomarker that is currently used in the clinic. The researchers in Samuelss and Ruppins lab suggest that expression of the immunoproteasome may be used as a biomarker for predicting better outcomes in melanoma and, together with mutational load testing, may improve patient matching to currently available immunotherapy.

Prof. Yardena Samuels is the head of the EKARD Institute for Cancer Diagnosis Research; and head of the Weizmann-Brazil Tumor Bank. Her research is also supported by the Laboratory in the name of M.E.H Fund established by Margot and Ernst Hamburger; the Green Family Charitable Foundation; the Wagner-Braunsberg Family Melanoma Research Fund; the Jean-Jacques Brunschwig Fund for the Molecular Genetics of Cancer; the Comisaroff Family Trust; the Rising Tide Foundation; the Fundacin Ramn Areces; the Meyer Henri Cancer Endowment; Ted and Sylvia Quint; Jean Jacques Roboh; and the European Research Council. Prof Samuels is the incumbent of the Knell Family Professorial Chair.

Original post:
Predicting Immunotherapy Success - BSA bureau

Read More...

Oncology Start-up OncoLens Named to Top 10 List of Innovative Technology Companies in Georgia – PR Web

Tuesday, February 25th, 2020

ATLANTA (PRWEB) February 24, 2020

OncoLens is pleased to announce it has been named one of the Top 10 Innovative Technology Companies in Georgia by the Technology Association of Georgia (TAG).

Oncology focused, OncoLens is a SaaS-based tumor board platform to help cancer centers connect members of their care teams, as they make treatment decisions for their patients. Centers can increase their referrals and streamline workflows. They can also reduce tumor board prep time by 90% and time to initiate patient treatment from 14 to 2 days.

OncoLens utilizes some of the latest technologies in conjunction with its own proprietary algorithms to identify treatment options for the cancer patient, said Anju Mathew, Co-Founder and CEO, OncoLens. The platform identifies clinical trials, evidence-based guidelines and personalized treatment options based on the tumor genotype. These options are proposed to the multi-disciplinary cancer care team to help them make the best decision possible for the cancer patient.

Better patient outcomes can result from OncoLens robust functionality. Cancer care teams can engage research staff early and automatically screen cases available for clinical trials. Physicians are able to access real time data for clinical decision-making, while the system works in the background to facilitate data/document collection, including EMR clinical data, laboratory and molecular genetics test results and radiology images. The platform also supports regulatory reporting for associations such as the Commission on Cancer (CoC), NAPBC and NAPRC.

The Top 10 were selected from TAGs Top 40 Innovative Technology Companies competition for showing the highest degree of innovation, the broadest scope and financial impact of their innovations, and the greatest effect of such innovation in promoting Georgia's technology industry throughout the U.S. and globally.

As a Top 10 honoree, OncoLens will present at The Summit, hosted by TAG, March 3-4, at Cobb Galleria, in Atlanta. It is Georgias largest technology showcase. We look forward to sharing the future of innovative cancer care at TAGs event, said Mathew.

###About OncoLens OncoLens develops technologies that improve cancer treatment planning, simplify tumor board management, facilitate survivorship care planning, and automate accreditations and quality reporting through an intelligent workflow engine that streamlines business processes and enables more informed clinical decision-making. For more information, visit http://www.oncolens.com.

Share article on social media or email:

Follow this link:
Oncology Start-up OncoLens Named to Top 10 List of Innovative Technology Companies in Georgia - PR Web

Read More...

University researchers show how a protein found – Mirage News

Tuesday, February 25th, 2020

Researchers at the University of Toronto have found that a receptor expression-enhancing protein contributes to normal heart development and function by regulating the sarcoplasmic reticulum, a network of tubules found in cardiac muscle cells.

The sarcoplasmic reticulum is key in the development and progression of heart disease, governing biochemical changes, structural remodeling and deterioration. But how this membrane-bound system organizes itself is still mostly unknown especially in cells with a highly differentiated or diverse network such as heart muscle cells, or cardiomyocytes.

Our findings show that a protein called REEP5 plays a critical role in regulating cellular stress responses in heart muscle cells, says Frank Shin-Haw Lee, a PhD student in the lab of Anthony Gramolini, an associate professor of physiology in U of Ts Faculty of Medicine who is based at the Ted Rogers Centre for Heart Research.

When REEP5 is depleted, it destabilizes the heart and reduces the amount of blood the heart can pump on each contraction, says Lee. When we removed this protein in both mice and zebrafish, it distorted the structure and shape of cardiomyocytes and led to cardiac dysfunction.

The journal Nature Communications published the findings this week.

When cardiomyocytes are under sustained stress from general dysfunction or disease, cellular pathways through the sarcoplasmic reticulum can lead to cell death and heart failure. Lee says that REEP5 is vital to the formation of the sarcoplasmic reticulum and to how it responds to stress and regulates calcium, which is essential for heart health.

A better understanding of how REEP5 functions in the heart may elucidate how heart failure develops amidst a sarcoplasmic reticulum in stress, Lee says.

The Gramolini lab worked with several other Toronto researchers on the study, including Ian Scott, a professor of molecular genetics at U of T and a senior scientist at the Hospital for Sick Children. The work builds on previous collaborative research from the labs of Gramolini, Scott and medical biophysics professor Thomas Kislinger in 2015, which created a blueprint of critical cell-surface and membrane-associated proteins in the heart.

Medical student Sina Hadipour-Lakmehsari was a co-first author on the current paper with Lee, and he says the findings may provide insight into heart disease in patients.

It is clearly an important protein for cardiac development and function and, combined with future human studies, it may help us unearth new potential therapies, Hadipour-Lakmehsari says, adding that the lab can continue to look at REEP5 in genetic studies to help shed light on diseases whose causes remain unknown.

This study is among the first in the world to show that the REEP5 protein plays an essential role in the stress responses that often lead to heart failure, adds Gramolini, who is also a scientist at Toronto General Hospital Research Institute, University Health Network. Deciphering the complex layers of heart function on a cellular level will help us generate new therapeutic and preventative strategies for heart failure.

The study recieved support from the Ted Rogers Centre Innovation Fund, the Natural Sciences and Engineering Research Council of Canada and the Canadian Institutes of Health Research, among others.

Continued here:
University researchers show how a protein found - Mirage News

Read More...

Page 20«..10..19202122..30..»


2024 © StemCell Therapy is proudly powered by WordPress
Entries (RSS) Comments (RSS) | Violinesth by Patrick