header logo image


Page 33«..1020..32333435..40..»

Archive for the ‘Genetics’ Category

How Is Genetic Genealogy Shaping The Way Cold Cases Are Solved? – Oxygen

Thursday, October 24th, 2019

In early 2018, investigators in California uploaded DNA from one of the Golden State Killer crime scenes to GEDmatch, an open data personal genomics database and genealogy website. They found a mutual descendant of the suspects great-great-great grandparents, and enlisted genetic genealogists to construct a family tree.

This eventually led authorities to Joseph James DeAngelo, a 72-year-old former police officer. DeAngelo was arrested that April and charged with 13 counts of murder, including one committed during burglary and rape, and 13 counts of kidnapping for robbery, according to CNN. He is currently awaiting trial.

Since DeAngelos arrest, law enforcement has investigated numerous cold cases using genealogical DNA databases, whether from open-use platforms like GEDmatch or commercial websites like FamilyTreeDNA. While an impressive number of cases have been solved, finding usable matches that lead to an arrest is labor intensive and often difficult.

Its a steep learning curve, CeCe Moore, head of genetic genealogy at Parabon NanoLabs, told Oxygen.com. Its been two years since genetic genealogists have started to be used. There are some law enforcement officials who are getting good at it. We just need more people that really put the time in to do it because there are a limited number of us out there that can work these cases skillfully.

To help law enforcement identify unknown suspect or victim DNA from a crime scene, genealogists upload the DNA sample to a database like GEDmatch. Then, they reverse-engineer the family tree of the unidentified person by comparing it to the family trees of people with whom they share DNA.

Youre hoping your top match is going to be at least a third cousin, and you can go back to a second great-grandparent. I use a lot of supporting matches as well, said Moore, who has worked on more than 200 cases. If youve got a lot of really close genetic matches, its obviously going to be a lot easier.

While genetic genealogists have proved critical in helping investigations, arrests and convictions are ultimately the result of police work.

It sometimes can only narrow it down so far, and then you turn it over to the investigators, Moore said. Were really just a lead generator. Were trying to get tips, and then investigators have to determine whether thats a viable tip or not.

The key to more successful identifications is access to DNA databanks, according to Moore. As more people upload their genetic information, there will be more profiles to compare them to.

The techniques are already very powerful, and the science is very powerful. We just need more DNA to compare it to, Moore told Oxygen.com. Thats what it relies on. We need more data and more people who are willing to contribute their DNA profiles to the effort.

As forensic genealogy has grown in usage, however, many genealogical websites have wrestled with the privacy concerns of their users. Increasingly, websites only allow access to users who have manually opted-in to share their profiles with law enforcement, thus limiting the data pool.

While Moore is hopeful that more cold cases will be solved in the coming years thanks to breakthroughs in DNA technology, she is realistic about the challenges ahead.

It takes a lot of skill unless you get very, very lucky, she said. Theres been some cases over the last year or so that law enforcement agencies have been able to figure it who it is themselves without a genetic genealogist because it was such a close match, but the vast majority of the cases 99 percent of them are going to need someone who is experienced and skilled, or they're just going to be wasting their time spinning their wheels. Its not that it will lead them in the wrong direction, it just wont lead them anywhere.

For more cold case investigations, follow Paul Holes as he explores the physical and emotional "DNA" of crime scenes in "The DNA of Murder with Paul Holes," premiering Saturdaysat 7/6c on Oxygen.

Crime Time is your destination for true crime stories from around the world, breaking crime news, and information about Oxygen's original true crime shows and documentaries. Sign up for our Crime Time Newsletter and subscribe to our true crime podcast Martinis & Murder for all the best true crime content.

Follow this link:
How Is Genetic Genealogy Shaping The Way Cold Cases Are Solved? - Oxygen

Read More...

Here’s Why We’re Not At All Concerned With Seattle Genetics’s (NASDAQ:SGEN) Cash Burn Situation – Yahoo Finance

Thursday, October 24th, 2019

There's no doubt that money can be made by owning shares of unprofitable businesses. For example, biotech and mining exploration companies often lose money for years before finding success with a new treatment or mineral discovery. But while the successes are well known, investors should not ignore the very many unprofitable companies that simply burn through all their cash and collapse.

Given this risk, we thought we'd take a look at whether Seattle Genetics (NASDAQ:SGEN) shareholders should be worried about its cash burn. For the purposes of this article, cash burn is the annual rate at which an unprofitable company spends cash to fund its growth; its negative free cash flow. First, we'll determine its cash runway by comparing its cash burn with its cash reserves.

View our latest analysis for Seattle Genetics

A cash runway is defined as the length of time it would take a company to run out of money if it kept spending at its current rate of cash burn. When Seattle Genetics last reported its balance sheet in June 2019, it had zero debt and cash worth US$376m. Looking at the last year, the company burnt through US$172m. That means it had a cash runway of about 2.2 years as of June 2019. Importantly, though, analysts think that Seattle Genetics will reach cashflow breakeven before then. If that happens, then the length of its cash runway, today, would become a moot point. Depicted below, you can see how its cash holdings have changed over time.

NasdaqGS:SGEN Historical Debt, October 21st 2019

On balance, we think it's mildly positive that Seattle Genetics trimmed its cash burn by 4.3% over the last twelve months. On top of that, operating revenue was up 32%, making for a heartening combination Considering the factors above, the company doesnt fare badly when it comes to assessing how it is changing over time. While the past is always worth studying, it is the future that matters most of all. For that reason, it makes a lot of sense to take a look at our analyst forecasts for the company.

Seattle Genetics seems to be in a fairly good position, in terms of cash burn, but we still think it's worthwhile considering how easily it could raise more money if it wanted to. Generally speaking, a listed business can raise new cash through issuing shares or taking on debt. Commonly, a business will sell new shares in itself to raise cash to drive growth. By comparing a company's annual cash burn to its total market capitalisation, we can estimate roughly how many shares it would have to issue in order to run the company for another year (at the same burn rate).

Seattle Genetics has a market capitalisation of US$15b and burnt through US$172m last year, which is 1.2% of the company's market value. So it could almost certainly just borrow a little to fund another year's growth, or else easily raise the cash by issuing a few shares.

It may already be apparent to you that we're relatively comfortable with the way Seattle Genetics is burning through its cash. In particular, we think its cash burn relative to its market cap stands out as evidence that the company is well on top of its spending. Its weak point is its cash burn reduction, but even that wasn't too bad! It's clearly very positive to see that analysts are forecasting the company will break even fairly soon Taking all the factors in this report into account, we're not at all worried about its cash burn, as the business appears well capitalized to spend as needs be. Notably, our data indicates that Seattle Genetics insiders have been trading the shares. You can discover if they are buyers or sellers by clicking on this link.

Story continues

Of course, you might find a fantastic investment by looking elsewhere. So take a peek at this free list of interesting companies, and this list of stocks growth stocks (according to analyst forecasts)

We aim to bring you long-term focused research analysis driven by fundamental data. Note that our analysis may not factor in the latest price-sensitive company announcements or qualitative material.

If you spot an error that warrants correction, please contact the editor at editorial-team@simplywallst.com. This article by Simply Wall St is general in nature. It does not constitute a recommendation to buy or sell any stock, and does not take account of your objectives, or your financial situation. Simply Wall St has no position in the stocks mentioned. Thank you for reading.

Visit link:
Here's Why We're Not At All Concerned With Seattle Genetics's (NASDAQ:SGEN) Cash Burn Situation - Yahoo Finance

Read More...

Study: The Likelihood of Developing PTSD Following Trauma Is Party Determined by Genetics – The Swaddle

Thursday, October 24th, 2019

Our understanding of post-traumatic stress disorder (PTSD), until now, is that it is a mental health disorder that occurs as a consequence of exposure to extreme, life-threatening stress, and/or serious injury. This exposure, by definition, is requisite for the development of PTSD, but not all those who face trauma necessarily develop it the individual susceptibility to PTSD varies widely. Since the turn of the century, scientists have been trying to find evidence for genetic influence on PTSD risk, with the last decade witnessing concerted efforts to identify specific DNA variants that can influence ones genetic susceptibility to develop PTSD.

New research, findings of which were published in Nature Communications, has, for the first time, identified a clear biological pathway for the mental health disorder, despite a section of science still viewing it as an entirely social construction. In the largest and most diverse genetic study of PTSD to date, scientists from the University of California, San Diego, School of Medicine and more than 130 additional institutions have concluded that genetics do, in fact, play a role in determining whether or not a person will develop PTSD, similar to the biological pathways of depression and other forms of mental illness.

Our long-term goal is to develop tools that might help clinicians predict who is at greatest risk for PTSD and personalize their treatment approaches. We cant always protect people from trauma. But we can treat them in the best ways possible, at the best time, Dr. Caroline Nievergelt, the studys first author, associate professor of psychiatry at UC San Diego School of Medicine and associate director of neuroscience in the Center of Excellence for Stress and Mental Health at the Veterans Affairs San Diego Healthcare System, said in a press release.

In collaboration with the Psychiatric Genomics Consortiums PTSD working group and Cohen Veterans Bioscience, a non-profit organization dedicated to accelerating PTSD and traumatic brain injury research, the studys authors built a 12-country network of more than 200 researchers, assembling data and DNA samples from more than 20,000 people with PTSD and 170,000 control subjects (those who did not develop PTSD following trauma).

At more than 200,000 people, the latest studys sample size is 10 times larger than the firstPsychiatric Genomics Consortium PTSD study, published in 2017, and includes both civilians and members of the military. The release notes the cohort is also the most ancestrally diverse for any psychiatric genetics study to date, with more than 23,000 people with PTSD of European ancestry and more than 4,000 of African ancestry.

Related on The Swaddle:

Researchers Find Genetic Hotspot Behind Autism

Putting this large database through statistical analyses, the studys authors measured the effect of gene variants at millions of different points on chromosomes across the human genomes on someones chances of developing PTSD.

According to the findings, PTSDs heritability the level of influence genetics has on the variability of PTSD risk among people is between 5% and 20%. Scientists found that, like other psychiatric disorders and several human traits, the risk of developing PTSD following trauma is a highly polygenic trait. This means there exist thousands of genes at different loci on different chromosomes that make tiny contributions to the disorder and when expressed together, add up to the heritable trait. Scientists have found gene variants at six loci that were strongly associated with PTSD risk.

Three of the six loci were specific to certain ancestral backgrounds two European and one African and three were only detected in men. The six loci hint that inflammatory and immune mechanisms may be involved in the disorder, which is consistent with findings from previous studies, the release summarises.

Further exploring the relatively nascent belief in science that many psychiatric disorders and behavioral traits have important molecular similarities at DNA-level, the study also analyzed genetic correlations between PTSD and 235 other disorders behaviors and physical traits. They found a significant overlap with 21, including depression, schizophrenia, insomnia, asthma, and coronary artery disease. Additionally, a Parkinsons disease gene involved in dopamine regulation was also found to be associated with PTSD. Similar to other mental disorders, the genetic contribution to PTSD correlates with that for many other traits. Further research is needed to determine what this means whether some of the same genes that influence risk for PTSD also influence risk for other diseases like, for example, depression, Karestan Koenen, a senior author of the study and associate member of the Stanley Center for Psychiatric Research at MIT and Harvard University, and a professor of psychiatric epidemiology in the Harvard T.H. Chan School of Public Health, said in the release.

Koenen adds: Based on these findings, we can say with certainty that there is just as much of a genetic component to PTSD risk as major depression and other mental illnesses. Our limited ability to study the living human brain and uncover the biological roots of PTSD has contributed to the lack of treatments and the stigma around this debilitating condition. Genetics helps us make new discoveries, find opportunities for new therapies, and counter that stigma.

Read more:
Study: The Likelihood of Developing PTSD Following Trauma Is Party Determined by Genetics - The Swaddle

Read More...

23andMe Wants Everyone to Get Used to Sharing Their Genetic Data – Motherboard

Thursday, October 24th, 2019

What Anne Wojcicki, co-founder and CEO of 23andMe, would like you to consider is this: Once upon a time, people were kinda freaked out by the idea of typing their credit card info into a website. Now this is done basically all the time. Your phone is a digital wallet; your web browsers will memorize your card numbers for you. Might your bank information get cribbed or hacked? Has it already been? Yeah, probably so, but this is How We Live Now. Its weird to think it was ever not normal.

This little historic tale is how Wojcicki addresses the current hesitance a majority of people feel to submitting their genetic material to a database by spitting into a tube and mailing that tube off to a companys lab. In a brief discussion at the TIME 100 Health Summit in New York City on Thursday, Wojcicki explained that shes actually grateful for the 2030 percent of the population thats down to spit (according to a Twitter poll TIME ran ahead of Wojcickis talk). At-home DNA kits like 23andMe are still a relatively new technology. People simply need to grow accustomed to the ideas of genetic testing and sharing genomic data with public databases, so that researchers can observe patterns across the population, and ultimately make people healthier, she said.

Its essentially an argument for sharing DNA data as good for public health, like a new-age equivalent of getting vaccinated. To drive the point home further, Eric Lander, a geneticist and director of the Broad Institute of MIT (and someone who once sat around a table with Jeffrey Epstein), mentioned a potential breakthrough in treating angiosarcoma, a rare, highly fatal cancer by using using DNAthat, Lander argued, may never have happened if it werent for peoples willingness to fork over health and genetic information.

Its hard to find a sensible, non-demonic argument against something that could lead to expedited breakthroughs in cancer treatment. But what has to be kept in mind is that 23andMe is a private company. These anecdotesvirtuous as they may soundare marketing techniques. Wojcicki rightfully believes that no singular institution will be able to harvest the amount of DNA that her company has. According to 23andMes About page, more than 10 million people have spit into its tubes and thereby handed their genetic information over. Of those 10 million, 80 percent have opted in to participate in research, via that spit data. For context: The National Institute of Health is currently in the midst of enrolling its largest DNA-related study to date (All Of Us), which will reach full enrollment at one million participants. About a year into their recruiting efforts, theyre about a quarter of the way there, according to what NIH Director Francis Collins said in a separate panel at Thursdays TIME 100 Health Summit.

Wojcicki emphasized that only those who opt in for ancestry information have their data entered into public databases, which are subject to subpoena (the likes of which helped identify the Golden State Killer). She further emphasized that, because DNA is highly similar among family members, submitting your spit implicates your relatives genetic information. (23andMe consents around this.) Thats a hell of a lot of data thats sitting around for seemingly forever, and since 23andMe is, once again, a private company, theres no telling what happens to this info if/when the company goes under, or if they decide to change their policies.

What 23andMe is sitting on now is perhaps the most valuable pool of genetic data in the world. Earlier this year, the company partnered with TrialSpark, an NYC-based research company, in order to use its large database of data to fit its opted-in consumers to studies. Mind you, Wojcicki used to work on Wall Street. She is, at her core, a businessperson. Charging customers to have their data, and then partnering with another company with an interest in that data, sounds lucrative as fuck; a hell of a business deal.

Wojcicki and Lander concluded their talk on Thursday with a heartwarming sentiment: The virtue of something like 23andMe is that consumers (or participants, however youd prefer to look at it) have access to their genetic data, rather than submitting it to a study and never getting feedback. Wojcicki refers to this as empowering; its empowering to know whats going on in the little strands that make you the person you are. It also sounds very empowering, monetarily speaking, for those in the game, marketing spit kits and trading (totally consented for!!) genetic data.

Go here to see the original:
23andMe Wants Everyone to Get Used to Sharing Their Genetic Data - Motherboard

Read More...

New universe of miniproteins is upending cell biology and genetics – Science Magazine

Monday, October 21st, 2019

By Mitch LeslieOct. 17, 2019 , 2:00 PM

Mice put human runners to shame. Despite taking puny strides, the rodents can log 10 kilometers or more per night on an exercise wheel. But the mice that muscle biologist Eric Olson of the University of Texas Southwestern Medical Center in Dallas and colleagues unveiled in 2015 stood out. On a treadmill, the mice could scurry up a steep 10% grade for about 90 minutes before faltering, 31% longer than other rodents. Those iron mice differed from counterparts in just one small waythe researchers had genetically altered the animals to lack one muscle protein. That was enough to unleash superior muscle performance. "It's like you've taken the brakes off," Olson says.

Just as startling was the nature of the crucial protein. Muscles house some gargantuan proteins. Dystrophin, a structural protein whose gene can carry mutations that cause muscular dystrophy, has more than 3600 amino acids. Titin, which acts like a spring to give muscles elasticity, is the biggest known protein, with more than 34,000 amino acids. The protein disabled in the mice has a paltry 46. Although researchers have probed how muscles work for more than 150 years, they had completely missed the huge impact this tiny protein, called myoregulin, has on muscle function.

Olson and his colleagues weren't the only ones to be blindsided by Lilliputian proteins. As scientists now realize, their initial rules for analyzing genomes discriminated against identifying those pint-size molecules. Now, broader criteria and better detection methods are uncovering minuscule proteins by the thousands, not just in mice, but in many other species, including humans. "For the first time, we are about to explore this universe of new proteins," says biochemist Jonathan Weissman of the University of California, San Francisco.

Biologists are just beginning to delve into the functions of those molecules, called microproteins, micropeptides, or miniproteins. But their small size seems to allow them to jam the intricate workings of larger proteins, inhibiting some cellular processes while unleashing others. Early findings suggest microproteins bolster the immune system, control destruction of faulty RNA molecules, protect bacteria from heat and cold, dictate when plants flower, and provide the toxic punch for many types of venom. "There's probably going to be small [proteins] involved in all biological processes. We just haven't looked for them before," says biochemist Alan Saghatelian of the Salk Institute for Biological Studies in San Diego, California.

The venom of this predatory water bug has more than a dozen small proteins.

Small proteins also promise to revise the current understanding of the genome. Many appear to be encoded in stretches of DNAand RNAthat were not thought to help build proteins of any sort. Some researchers speculate that the short stretches of DNA could be newborn genes, on their way to evolving into larger genes that make full-size proteins. Thanks in part to small proteins, "We need to rethink what genes are," says microbiologist and molecular biologist Gisela Storz of the National Institute of Child Health and Human Development in Bethesda, Maryland.

Despite the remaining mysteries, scientists are already testing potential uses for the molecules. One company sells insecticides derived from small proteins in the poison of an Australian funnel-web spider. And a clinical trial is evaluating an imaging agent based on another minute protein in scorpion venom, designed to highlight the borders of tumors so that surgeons can remove them more precisely. Many drug companies are now searching for small proteins with medical potential, says biochemist Glenn King of the University of Queensland in St. Lucia, Australia. "It's one of the most rapidly growing areas."

Other short amino acidchains, often called peptides or polypeptides, abound in cells, but they are pared-down remnants of bigger predecessors. Myoregulin and its diminutive brethren, in contrast, are born small. How tiny they can be remains unclear. Fruit flies rely on a microprotein with 11 amino acids to grow normal legs, and some microbes may crank out proteins less than 10 amino acids long, notes microbial genomicist Ami Bhatt of Stanford University in Palo Alto, California. But even the largest small proteins don't measure up to average-size proteins such as alpha amylase, a 496amino-acid enzyme in our saliva that breaks down starch.

Few small proteins came to light until recently because of a criterion for identifying genes set about 20 years ago. When scientists analyze an organism's genome, they often scan for open reading frames (ORFs), which are DNA sequences demarcated by signals that tell the cell's ribosomes, its proteinmaking assembly lines, where to start and stop. In part to avoid a data deluge, past researchers typically excluded any ORF that would yield a protein smaller than 100 amino acids in eukaryotes or 50 amino acids in bacteria. In yeast, for example, that cutoff limited the list of ORFs to about 6000.

Relaxing that criterion reveals that cells carry vastly more ORFs. Earlier this year, Stanford postdoc Hila Sberro Livnat, Bhatt, and colleagues trawled genome fragments from the microbes that inhabit four parts of the human body, including the gut and skin. By searching for small ORFs that could encode proteins between five and 50 amino acids long, the researchers identified about 4000 families of potential microproteins. Almost half resemble no known proteins, but the sequence for one small ORF suggested that a corresponding protein resides in ribosomesa hint that it could play some fundamental role. "It's not just genes with esoteric functions that have been missed" when scientists overlooked small ORFs, Bhatt says. "It's genes with core functions."

For the first time, we are about to explore this universe of new proteins.

Other cells also house huge numbers of short ORFsyeast could make more than 260,000 molecules with between two and 99 amino acids, for example. But cells almost certainly don't use all those ORFs, and some of the amino acid strings they produce may not be functional. In 2011, after finding more than 600,000 short ORFs in the fruit fly genome, developmental geneticist Juan Pablo Couso of the University of Sussex in Brighton, U.K., and colleagues tried to whittle down the number. They reasoned that if a particular ORF had an identical or near-identical copy in a related species, it was less likely to be genomic trash. After searching another fruit fly's genome and analyzing other evidence that the sequences were being translated, the group ended up with a more manageable figure of 401 short ORFs likely to yield microproteins. That would still represent a significant fraction of the insects' protein repertoirethey harbor about 22,000 full-size proteins.

Weissman and colleagues found microproteins a second way, through a method they invented to broadly determine which proteins cells are making. To fashion any protein, a cell first copies a gene into messenger RNA. Then ribosomes read the mRNA and string together amino acids in the order it specifies. By sequencing mRNAs attached to ribosomes, Weissman and his team pinpoint which ones cells are actually turning into proteins and where on the RNAs a ribosome starts to read. In a 2011Cellstudy, he and his team applied that ribosome profiling method, also called Ribo-seq, to mouse embryonic stem cells and discovered the cells were making thousands of unexpected proteins, including many that would fall below the 100amino-acid cutoff. "It was quite clear that the standard understanding had ignored a large universe of proteins, many of which were short," Weissman says.

Saghatelian and his colleagues adopted a third approach to discover a trove of microproteins in our own cells. The researchers used mass spectrometry, which involves breaking up proteins into pieces that are sorted by mass to produce a distinctive spectrum for each protein. Saghatelian, his then-postdoc Sarah Slavoff, and colleagues applied the method to protein mixtures from human cells and then subtracted the signatures of known proteins. That approach revealed spectra for 86 previously undiscovered tiny proteins, the smallest just 18 amino acids long, the researchers reported in 2013 inNature Chemical Biology.

Being small limitsa protein's capabilities. Larger proteins fold into complex shapes suited for a particular function, such as catalyzing chemical reactions. Proteins smaller than about 50 to 60 amino acids probably don't fold, says chemist Julio Camarero of the University of Southern California in Los Angeles. So they probably aren't suited to be enzymes or structural proteins.

However, their diminutive size also opens up opportunities. "They are tiny enough to fit into nooks and crannies of larger proteins that function as channels and receptors," Olson says. Small proteins often share short stretches of amino acids with their larger partners and can therefore bind to and alter the activity of those proteins. Bound microproteins can also shepherd bigger molecules to new locationshelping them slip into cell membranes, for instance.

A microprotein in the poison of the deathstalker scorpion has been fused to a fluorescent dye to make tumors emit near-infrared light. (1) A tumor seen in visible light (2)Same tumor in visible and near-infrared light

Because of their attraction to larger proteins, small proteins may give cells a reversible way to switch larger proteins on or off. In a 2016 study inPLOS Genetics, plant developmental biologist Stephan Wenkel of the University of Copenhagen and colleagues genetically alteredArabidopsisplants to produce extra amounts of two small proteins. The plants normally burst into flower when the days are long enough, but when they overproduced the two microproteins, their flowering was postponed. The small proteins caused that delay by blocking a hefty protein called CONSTANS that triggers flowering. They tether CONSTANS to other inhibitory proteins that shut it down. "A cell uses things that help it survive. If a short protein does the job, that's fine," Saghatelian says.

Those jobs include other key tasks. In 2016, Slavoff, Saghatelian, and colleagues revealed that human cells manufacture a 68amino-acid protein they named NoBody that may help manage destruction of faulty or unneeded mRNA molecules. NoBody's name reflects its role in preventing formation of processing bodies (P-bodies), mysterious clusters in the cytoplasm where RNA breakdown may occur. When the protein is missing, more P-bodies form, thus boosting RNA destruction and altering the cell's internal structure. "It shows that small proteins can have massive effects in the cell," Slavoff says.

Muscles appear to depend on a variety of microproteins. During embryonic development, individual muscle cells merge into fibers that power contraction. The 84amino-acid protein myomixer teams up with a larger protein to bring the cells together, Olson's team reported in 2017 inScience. Without it, embryonic mice can't form muscles and are almost transparent.

Later in life, myoregulin steps in to help regulate muscle activity. When a muscle receives a stimulus, cellular storage depots spill calcium, triggering the fibers to contract and generate force. An ion pump called SERCA then starts to return the calcium to storage, allowing the muscle fibers to relax. Myoregulin binds to and inhibits SERCA, Olson's team found. The effect limits how often a mouse's muscles can contractperhaps ensuring that the animal has muscle power in reserve for an emergency, such as escaping a predator. Another small protein, DWORF, has the opposite effect, unleashing SERCA and enabling the muscle to contract repeatedly.

Even extensively studied organisms such as the intestinal bacteriumEscherichia coliharbor unexpected small proteins that have important functions. Storz and her team reported in 2012 that a previously undiscovered 49amino-acid protein called AcrZ helps the microbe survive some antibiotics by stimulating a pump that expels the drugs.

And the venom produced by a variety of organismsincluding spiders, centipedes, scorpions, and poisonous mollusksteems with tiny proteins. Many venom components disable or kill by blocking the channels for sodium or other ions that are necessary for transmission of nerve impulses. Small proteins "hit these ion channels with amazing specificity and potency," King says. "They are the major components of venoms and are responsible for most of the pharmacological and biological effects."

Australia's giant fish-killing water bug, for instance, doesn't just rely on sharp claws and lancelike mouthparts to subdue prey. It injects its victims with a brew of more than 130 proteins, 15 of which have fewer than 100 amino acids, King and colleagues reported last year.

Unlike hulking proteinssuch as antibodies, microproteins delivered by pill or injection may be able to slip into cells and alter their functions. Captopril, the first of a class of drugs for high blood pressure known as angiotensin-converting enzyme inhibitors was developed from a small protein in the venom of a Brazilian pit viper. But the drug, which the Food and Drug Administration approved for sale in the United States in 1981, was discovered by chance, before scientists recognized small proteins as a distinct group. So far, only a few microproteins have reached the market or clinical trials.

Cancer researchers are trying to capitalize on a microprotein in the poison of the deathstalker scorpion (Leiurus quinquestriatus) of Africa and the Middle East. The molecule has a mysterious attraction to tumors. By fusing it to a fluorescent dye, scientists hope to illuminate the borders of brain tumors so that surgeons can safely cut out the cancerous tissue. "It lights up the tumor. You can see the margins and if there are any metastases," King says. A clinical trial is now evaluating whether the dual molecule can help surgeons remove brain tumors in children.

How important small proteins will be for medicine is still unknown, but they have already upended several biological assumptions. Geneticist Norbert Hbner of the Max Delbrck Center for Molecular Medicine in Berlin and colleagues found dozens of new microproteins in human heart cells. The group traced them to an unexpected source: short sequences within long noncoding RNAs, a variety that was thought not to produce proteins. After identifying 169 long noncoding RNAs that were probably being read by ribosomes, Hbner and his team used a type of mass spectrometry to confirm that more than half of them yielded microproteins in heart cells, a result reported earlier this year inCell.

Bacteria such as Escherichia coli also churn out many microproteins, although their functions remain unclear in many cases.

The DNA sequences for other tiny proteins also occur in unconventional locations. For example, some lie near the ORFs for bigger proteins. Researchers previously thought those sequences helped manage the production of the larger proteins, but rarely gave rise to proteins themselves. Some coding sequences for recently discovered microproteins are even nested within sequences that encode other, longer proteins.

Those genomic surprises could illuminate how new genes arise, says evolutionary systems biologist Anne-Ruxandra Carvunis of the University of Pittsburgh in Pennsylvania. Researchers had thought most new genes emerge when existing genes duplicate or fuse, or when species swap DNA. But to Carvunis, microproteins suggest protogenes can form when mutations create new start and stop signals in a noncoding portion of the genome. If the resulting ORF produces a beneficial protein, the novel sequences would remain in the genome and undergo natural selection, eventually evolving into larger genes that code for more complex proteins.

In a 2012 study, Carvunis, who was then a postdoc in the lab of Marc Vidal at the Dana-Farber Cancer Institute in Boston, and colleagues found that yeast translate more than 1000 short ORFs into proteins, implying that these sequences are protogenes. In a new study, Carvunis and her team tested whether young ORFs can be advantageous for cells. They genetically altered yeast to boost output of 285 recently evolved ORFs, most of which code for molecules that are smaller than the standard protein cutoff or just over it. For almost 10% of the proteins, increasing their levels enhanced cell growth in at least one environment. The results, posted on the preprint server bioRxiv, suggest these sequences could be on their way to becoming full-fledged genes, Carvunis says.

Slavoff still recalls being astonished when, during her interview for a postdoc position with Saghatelian, he asked whether she would be willing to go hunting for small proteins. "I had never thought that there could be this whole size of proteins that was dark to us until then."

But the bet paid offshe now runs her own lab that is searching for microproteins. Recently, she unleashed some of her postdocs and graduate students on one of the most studied organisms, the K12 strain ofE. coli.The team soon uncovered five new microproteins. "We are probably only scratching the surface," she says.

Read the original post:
New universe of miniproteins is upending cell biology and genetics - Science Magazine

Read More...

Genetic testing kits ‘may wrongly reassure those at risk of cancer’ – The Guardian

Monday, October 21st, 2019

Consumer genetic tests could be giving false reassurance to those at heightened risk of cancers, according to findings presented at an international conference this week.

The study, by clinical genetic testing company Invitae, revealed that tests for breast and bowel cancer risk by direct-to-consumer companies such as 23andMe give negative results to the vast majority of those carrying DNA mutations in the genes under investigation.

These tests should not be taken at face value at all, whether they are positive or negative, said Edward Esplin from Invitae ahead of the annual meeting of the American Society of Human Genetics in Houston, Texas.

The data really underscores that there needs to be increased awareness that results from this type of screening may not be wrong but theyre woefully incomplete.

The research also showed that those from Asian and African-American backgrounds were more likely to carry mutations that were not designed to be detected by the consumer tests.

The research focused on DNA-based tests relating to breast, ovarian and bowel cancer that were recently approved by the US Food and Drug Administration.

The tests operate by a subject sending a DNA swab in the post for analysis and then receiving results with information about how their genetics could influence their health.

In the case of breast and ovarian cancer, the FDA has approved a screening test for three specific mutations on the BRCA1 and BRCA2 genes, which are most common in people of Ashkenazi Jewish heritage. However, these mutations are rare in people from other backgrounds.

Similarly, for bowel cancer, 23andMe offers FDA-authorised tests for two mutations, which are most common among individuals of northern European ancestry. The company explains the limitations of these tests to consumers and on its website.

Esplin said that despite this, consumers could be wrongly reassured by a negative result.

The study analysed the DNA of 270,806 patients who had been referred by healthcare providers for testing of the MUTYH gene, and 119,328 who had been referred for BRCA1/2 genetic testing.

It showed that for both tests, the majority of those carrying mutations would not be spotted, which Invitae describes as a clinical false-negative result.

For MUYTH, 40% of individuals with mutations in both copies of their MUTYH genes consistent with an almost 100% lifetime risk of bowel cancer had different mutations to those screened for in the FDA-approved test. This figure rose to 100% for those from Asian backgrounds and 75% for African-Americans.

For BRCA genes, 94% of non-Ashkenazi Jewish individuals and 19% of those of Ashkenazi heritage had a mutation that would be missed. Again, the figures were highest for those of Asian (98%) and African-American (99%) ancestry. Its performing a disproportionate disservice to individuals of these underrepresented groups, Esplin said.

A clinical false-negative result can be incorrectly reassuring, excluding a patient from receiving the preventive care they need based on their risk, he added. It could be the difference between preventing cancer and developing cancer.

In response to the findings, 23andMe said in a statement: The claims made by a competitor that we are returning clinical false negatives is incorrect and a false characterisation of 23andMes test. Our test is extremely accurate. As part of the FDA authorisation process weve demonstrated over 99% accuracy for the variants we test for in our health product.

The company said it makes clear to customers that it tests only for certain genetic variants and that customers should not forgo any recommended testing based on 23andMe results. 23andMe is not a diagnostic test, the company said. If an individual has a family history of cancer or other indications for clinical testing we always recommend consulting a healthcare provider first.

Prof Anneke Lucassen, a clinical geneticist at the University of Southampton, said that, in her experience, non-specialists would be likely to wrongly interpret negative results as an all-clear.

I do think the false-negative rate is an issue, not necessarily through the companies fault but through low general awareness, she said. Most people who come to clinic ask: Have I got the gene for breast cancer? and imagine its a single test, not that the test involves looking through around 20,000 letters of the genetic code to see if any one of them might be different.

View original post here:
Genetic testing kits 'may wrongly reassure those at risk of cancer' - The Guardian

Read More...

Genetic Tests For Psychiatric Drugs Now Covered By Some Insurers : Shots – Health News – NPR

Monday, October 21st, 2019

Myriad Genetics is among a handful of companies that make a genetic test to help doctors choose psychiatric medicines for patients. Evidence that the tests are effective has been called "inconclusive." Myriad Genetics hide caption

Myriad Genetics is among a handful of companies that make a genetic test to help doctors choose psychiatric medicines for patients. Evidence that the tests are effective has been called "inconclusive."

As a teenager, Katie Gruman was prescribed one mental health drug after another. None seemed to help her manage symptoms of anxiety and bipolar disorder, so she self-medicated with alcohol and illicit drugs.

It would take five years, and trying more than 15 different medications, before she found meds that actually helped.

Now 28 and in recovery, Gruman has been on the same drugs for years. But when a clinician recommended a genetic test to see which drugs work best for her, she took it.

Reading the test results "was definitely vindicating," she says. Medications that hadn't worked for her as a teenager were the same ones the results marked as bad fits.

She says she wishes she had taken the test as a teenager. "I could have avoided a lot of disaster in my life," she says.

Psychiatric medications are known to be hard to match to symptoms, and many patients like Gruman live through years of trial and error with their doctors.

Companies that make genetic tests like the one Gruman used say they can save patients and doctors from prolonged searching for the right medication and save insurance companies from paying for ineffective drugs. But many researchers say the tests don't have enough evidence backing them up. The Food and Drug Administration has warned that the tests could potentially steer patients towards the wrong medications. Nonetheless, UnitedHealthcare, the nation's largest insurer, began covering them October 1 for its 27 million individual and group plans.

Test makers hailed the announcement of United's coverage, the first from an insurance company to apply to all of its commercial plans across the country.

"We expect this to be a tipping point," says Shawn Patrick O'Brien, CEO of Genomind, a company that makes one of the tests. Other insurers will cover the tests "because they don't want to be uncompetitive in the marketplace," he predicts.

If the prediction is correct, it would likely fuel a market that has seen its largest test maker, Myriad Genetics, sell about 375,000 of its psychiatric medicine tests in the 2019 fiscal year, according to Jack Meehan, an industry analyst for Barclays. Myriad reported that it sold $113 million worth of the tests.

In addition to UnitedHealthcare's coverage, Myriad Genetics' test is covered by Medicare, a regional Blue Cross Blue Shield affiliate, and the insurance network for the grocery chain Kroger, a spokesperson says.

Genomind has discussed coverage with insurers including Anthem and Blue Cross Blue Shield, O'Brien says.

Debates over efficacy

As the field of genetic testing to help diagnose and treat disease grows, medicine has embraced certain tests, such as that for the BRCA gene linked to breast cancer. But many researchers say there is not enough evidence tying genetic variants to better outcomes for most psychiatric medications.

James Potash, the head of psychiatry at Johns Hopkins Medicine and an expert on psychiatric genetics, says of all the tests claiming to improve depression treatment, GeneSight's has the most proof. That isn't saying much, though.

"I wouldn't say there's no evidence that it works," he says. "It's just the evidence at this point is still weak."

The idea behind the tests is that in some cases, people can have different reactions to the same drug, even at the same dose, because they have different gene variants. Which variant a person has can affect how quickly or slowly a medicine moves through their body.

This link between genes and drug metabolism has been known for decades, says Francis McMahon, who leads genetic research into mood and anxiety disorders at the National Institutes for Mental Health.

Usually, the longer it takes your body to process a drug, the easier it is for that medication to have an effect. But in psychiatry, McMahon says, how fast someone processes a drug, or metabolizes it, and how well they respond to the drug "are sometimes not strongly related."

This skepticism is shared by some insurance companies. "Anthem considers these tests investigational and not medically necessary," says a spokesman for the carrier, which covers 41 million people. The Blue Cross Blue Shield Federal Employee Program, which covers about two-thirds of government workers and their families, said "there is not enough evidence at this time to determine the effect of genetic testing on health outcomes," according to a spokeswoman.

Test makers are also facing FDA objections that they haven't proven some of the claims underpinning genetic tests for medications, including that antidepressants work better with some gene variants.

"Changing drug treatment based on the results from such a genetic test could lead to inappropriate treatment decisions and potentially serious health consequences for the patient," the agency warned in late 2018. It told companies to stop naming specific drugs, in marketing materials or test results, for which its tests "claim to predict a patient's response" without "scientific or clinical evidence to support this use."

Most test makers complied. One, Inova Genomics Laboratory, stopped selling a range of tests, including its test for mental health disorders, after the FDA followed up with a warning letter in April.

Several mental health advocacy groups, including the National Alliance on Mental Illness, have sided with test makers in their dispute with the FDA. Keeping the names and types of medication off of genetic test reports, as the FDA has required, will "impede the ability of psychiatrists and other front-line health care professionals to personalize medication decisions" for patients with depression, the groups wrote the FDA in September.

Some have argued that genetic tests like these shouldn't be regulated by the FDA at all. Tests conducted in a lab are a medical service, not a medical device that's shipped like a product, says Vicky Pratt, president of the Association for Molecular Pathology. As a medical service, she says, clinical laboratories are already regulated by the Centers for Medicare and Medicaid Services.

"It would be redundant to have dual regulation by both the FDA and CMS," says Pratt.

Cost-benefit analysis

Research into the tests' efficacy is ongoing and continues to be debated.

Myriad hoped to bolster evidence for its test, GeneSight, in a study it funded that was published this year in the Journal of Psychiatric Research, but the results were mixed.

In the study, doctors used genetic tests to help prescribe medications for one group of patients with depression, while another group of patients received usual care. There was overall no difference between the groups in the study's primary measure of symptom improvement, though some patients showed improved response and remission rates.

Responding to criticisms of its clinical trial results, Myriad Genetics spokesman Ron Rogers says the trial population whose average participant had tried more than three unsuccessful medications for depression was uniquely difficult to treat. He says he expects to see stronger outcomes in a forthcoming review of the trial data.

In a statement on the use of genetic testing in psychiatry, the International Society of Psychiatric Genetics, calls the existing evidence "inconclusive," and notes that if 12 patients take such a test for antidepressants, just one will benefit from it.

A low rate of success means insurers will have to pay for a lot of tests for one useful result, says Barclays analyst Meehan. Meehan pointed to a letter about the recent GeneSight study that was published in the same journal, which found that 20 patients would need to take the test for one to recover as a result. At $2,000 for a GeneSight test, the authors wrote, that means patients and insurers would have to cover $40,000 worth of tests. (While competitor Genomind does not share pricing information, a spokeswoman confirmed that it has an active contract with the Department of Veterans to supply tests for $1,886.)

Still some clinicians value the tests. Skeptics often misunderstand how the tests should be used, argues Daniel Mueller, a professor at the University of Toronto who researches how genes and drugs interact. (Mueller is involved in research comparing Myriad's GeneSight to another test developed by a University of Toronto-affiliated hospital.) Most of the time, he says, doctors who order the test already plan to prescribe medication. The test is just another tool to help them decide which one to prescribe.

"It's not an alternative intervention," Mueller says. "It's additional information." He orders the test for most patients who do not respond to at least one antidepressant.

"If you think about the cost of depression and weeks of suffering that you can potentially avoid for some patients," Mueller says, he thinks anyone who can afford a test should take it. (Myriad says 95% of patients pay less than $330 for their test, the cost remaining after insurance and possible financial assistance; Genomind says most privately insured customers pay no more than $325.)

A lack of watertight evidence for the tests should not stop doctors from using it to inform their choice of medication, says Reyna Taylor, who leads public policy for the National Council for Behavioral Health, one of the advocacy groups that defended the tests in a letter to the FDA. "You use the science that you currently have," she says.

"Whether our providers choose to use [a genetic test] or not, we want them to have that choice," she adds.

Disagreement among experts hasn't dissuaded UnitedHealthcare from paying for the tests.

In a statement, UnitedHealthcare spokeswoman Tracey Lempner says they "frequently review our coverage policies to ensure they reflect the most current published evidence-based medicine and specialty society recommendations."

Graison Dangor is a journalist in Brooklyn.

Visit link:
Genetic Tests For Psychiatric Drugs Now Covered By Some Insurers : Shots - Health News - NPR

Read More...

Hacking Darwin: How the coming genetics revolution will play out – New Atlas

Monday, October 21st, 2019

Jamie Metzl is an extremely impressive man. Having held senior positions on Clinton's National Security Council and Department of State, and Joe Biden's Senate Foreign Relations Committee, he's also been Executive VP of the Asia Society, a Senior Fellow at the Atlantic Council and a former partner in Cranmere LLC, a global investment company. Today, while he's not running ultra-marathons, he's best known as a geopolitics expert, futurist and author.

Metzl writes in science fiction and scientific non-fiction, and his latest book, Hacking Darwin: Genetic Engineering and the Future of Humanity, delivers a serious, strongly-researched warning on what's likely to happen if we sleepwalk into the genetics age.

We spoke to Metzl at WCIT 2019 in Yerevan, Armenia, where he appeared as a keynote speaker, moderator and panel member.

Vahram Baghdasaryan/WCIT Yerevan 2019

"Right now were at this moment of super-convergence," Metzl tells us. "Its not any one technology thats determinative, its all these technologies happening at the same time, because theyre all influencing each other."

The first of these, Metzl outlines, is cheap sequencing of the human genome. Well need a ton of genetic information to be able to find the patterns needed to move forward, and while the cost of full genome sequencing is currently the limiting factor, it's dropping steeply, from around US$2.7 billion in 2003, to less than US$600 today. That's going to have to come down by another order or two of magnitude before it starts getting used as a matter of routine.

Secondly, 5G connectivity and the Internet of Things promises to teach us enormous amounts of information about people's health over the years, as wearable health analysis devices begin to stream back colossal piles of dynamic data about what's going on in people's bodies. Eventually, this will enable population-wide phenotypical research to be cross-checked against the genome to learn even more about how genes express themselves, individually and in concert with one another.

Thirdly, big data and analytics tools. The 2.9 billion haploid pairs making up each sequenced human genome represent about 725 megabytes of data, and dynamic health records will likely require even more storage space, in formats that can easily be cross-checked against each other at a massive scale.

Metzl notes that artificial intelligence or more precisely, deep learning is the only way we'll ever be able to meaningfully process such monstrous amounts of data, and its capabilities are rocketing forward daily. Perhaps when it's ready for serious commercial use, the speed and power of quantum computing will prove invaluable in quickly crunching through these petabytes of data.

Then of course, there are the wetter technologies: vastly improved IVF technologies that will soon enable us to generate egg and sperm cells from skin cells without needing invasive or embarrassing procedures to be carried out. Eventually, we'll have the capability to cheaply produce dozens, or even hundreds of embryos to sequence, select and implant.

And of course, gene editing tools. CRISPR/Cas9 editing is the most famous example of these, but it's already being compared to "genetic vandalism" due to its imprecise nature. More accurate and precise tools are constantly being discovered and refined to edit the genome of living subjects.

"We have to stitch together all these technologies," says Metzl, "and its already starting to happen. And itll happen increasingly until the end of time."

Vahram Baghdasaryan/WCIT Yerevan 2019

The first step, says Metzl, will be in healthcare. Our interactions with health care professionals will move from the current generalized model, to something more personal and precise as we start to understand what treatments work for people with certain genetic markers. Eventually, we'll have enough information to start engaging in predictive health care.

"You dont need to be perfect to make a huge impact on health care," says Metzl, "you just have to be better than the status quo, where nobody has that information, for it to be applied." It'll inch forward, offering probabilities rather than certainties as more and more is discovered.

Next and soon, we'll start seeing advanced embryo selection as a core part of any IVF treatment. Prospective parents will start having multiple embryos to choose from, each of which will have its genome fully sequenced so they'll be able to choose between offspring with a growing amount of information.

To begin with, this will allow parents to select against crippling genetic diseases, much the same as how parents who can afford the right scans can "select against" things like Down syndrome now.

But as science learns more and more about what individual genes, and combinations of them, do to the final outcome of a human, we'll quickly gain the ability to select for certain traits as well as against others. Will you want your child to be taller? More athletic, with a greater proportion of fast twitch muscle fibers? What about intelligence? Skin color? Eye color? Would you select for a child with a higher probability of living longer? Would you select for a child with a higher degree of extraversion, or a more even temperament?

All these things, and many more, are already known to have genetic underpinnings. And soon, parents will be able to choose between dozens, or potentially hundreds of their own biological embryos with this information at hand. All these possible kids are yours, so why wouldn't you choose the one that appears to have the best possible shot at life? Not doing so, says Metzl, could grow to be seen as a "crime against potential."

The disadvantages of having children the old-fashioned way will soon become apparent, as smarter, stronger, faster, healthier kids born from selection processes begin to dominate across a range of competitive situations, from sport to business to earning capacity and these advantages will multiply with subsequent generations, as more and more science is applied to the reproductive process.

"Embryo selection uses technologies that already exist," says Metzl. "IVF, embryo screening, and genome sequencing. Obviously we need to get better at all these things, but its happening very, very quickly."

And that's just using our naturally-occurring genetics. Soon afterward will follow precision gene editing, in which you select option J from your pre-implanted embryos, but make a few tweaks before you implant it. Here's where things start getting a little sketchy, as you're making edits to the germ line of the human species.

"Editing the genome requires the understanding that one gene might not just do one thing; it might do a lot of things," Metzl tells us. "If its a particularly harmful gene, then we know the alternative is deadly, so that decision will be easier. But when we move into the world of non-deadly single gene mutations, well, then the costs of not having a full understanding go higher."

Metzl says it's clear which direction things will go."We are going to do more and more complex genome editing," he tells us, "either to address risks, or to create enhancements - and there will be no natural boundary between the two. This is all about ethics. The science is advancing, theres nothing we can do to stop the science. The question is ethics."

The dawn of a new age of superhumans could nearly be upon us, in which a lucky, selected, edited few will have extraordinary genetic potentials in a wide range of areas. Sports could become almost meaningless, as it'll be impossible to tell a selected or edited human from a "natural born" one. Humanity will begin steering its own evolution for the first time in history, with some predictable results and some we can't see coming.

Negative results, says Metzl, could include everything from a gaping division between genetic haves and have-nots which could express itself within and between countries all the way up to eliminating all human life altogether. "We may make choices based on something we think is really good, like eliminating a terrible disease," says Metzl, "but then that genetic pattern that enabled that disease, in some other formulation, could be protective against some threat we cant even imagine, thats coming our way a thousand years from now. Thats why we need to be so respectful of our diversity. Genetic diversity, up to this point, has been our sole survival strategy. If we didnt have diversity, you could say wed still be single-celled organisms. We wouldnt, wed probably just have died. When the world changes around us, diversity is what helps us survive."

And then there's the potential of creating genetically engineered weapons. "Researchers in Canada spent $100,000 a couple of years ago," says Metzl, "to create essentially a weaponized version of horse pox in the lab, to show what could be done. Well, that could probably now be done for $20,000. In five years, you might be talking $2,000. These tools are agnostic. They dont come with their own value system. Just like nuclear power. We had to work out what are the OK uses, what are the not OK uses, and how do we structure things to we minimize the downsides."

Metzl wants people across the world to be informed about the technologies and capabilities that are barreling down the pipeline toward us, so meaningful efforts can be made to steer them in a direction that everyone can agree on, and set up clear redlines past which we agree not to venture. Each country, he says, needs to set up a national regulatory infrastructure to control the pace of these changes, and there also needs to be an international body with some teeth to make sure certain nations don't leap ahead and change the nature of humanity just due to lax regulations.

"This is always going to be changing," says Metzl. "The science is changing, the societal norms about what is and isnt OK are going to be changing too, and we need a dynamic process that can at least try to do a better job of keeping up with that rapid change."

Where does Metzl stand personally on how this next phase should be approached? "I'm a conservative person about this," he says. "I mean, four billion years of evolution is a lot. Life has made a lot of trade-offs. So if youre going against four billion years of evolution, you have to be humble. We know so little about the body. We cant let our hubris run away with us."

If you want to get informed on this incredibly complex, multilayered and potentially explosive technological revolution, Metzl's book Hacking Darwin: Genetic Engineering and the Future of Humanity (April 2019) is an outstanding summary with more examples and possible future situations laid out than you could possibly need, written in an engaging style designed to be accessible to anyone. I found it extremely enlightening and recommend it thoroughly.

Source: Jamie Metzl, WCIT Yerevan 2019

See the original post here:
Hacking Darwin: How the coming genetics revolution will play out - New Atlas

Read More...

Plotly Sponsors Development of Predictive Genetics Application in Partnership with McGill University – GlobeNewswire

Monday, October 21st, 2019

MONTREAL, Oct. 21, 2019 (GLOBE NEWSWIRE) -- Plotly, developer of the leading data science platform for creating analytic applications, today announced a partnership with McGill University to fund three Ph.D. interns in collaboration with Mitacs, a not-for-profit organization that fosters growth and innovation in Canada. The doctoral students will work with Sahir Rai Bhatnagar, Assistant Professor of Biostatistics, to create a predictive genetics application to better understand the genetic determinants of temporomandibular disorder (TMD). The tool will be based on a machine learning-driven analysis of the largest available dataset on TMD, which causes pain in the jaw.

Plotly and Mitacs are working together to provide financial support for the interns, and Plotly is contributing support for use of the companys powerful Dash software, an open source platform for building analytic web applications. Dr. Bhatnagars team will use Dash to interactively analyze the large dataset and visualize results from the machine learning model. These visualizations will provide key insight into which genetic components are driving the predictions. The team will create analytical applications in the R programming language for use by researchers studying pain and working to identify drug targets in order to develop more effective treatments.

This partnership expands an ongoing collaboration between Plotly and Mitacs. Last year, Plotly sponsored three bioinformatics interns at the Universit de Sherbrooke as they developed a visualization tool for the universitys CoBIUS Lab. The model enabled researchers to view DNA or complex molecules in 3D.

Plotly is delighted to work Mitacs to partner with McGill in support of technical talent in Qubec, said Jack Parmer, CEO of Plotly. Its important to us to give back to the communities were a part of, from open source data science to Canadian research teams. By contributing funding and use of our technology to these three promising biostatistics researchers, we hope to benefit not only the students, but patients across Qubec.

Dr. Bhatnagar commented: Funding from Plotly and Mitacs will help us bring our work out of the lab and to Canadas healthcare community. Dash will help our team visualize a trove of data and may give us, as well as the researchers at the Qubec Pain Research Network, more insight than ever before on temporomandibular disorder.

For 20 years, Mitacs has helped develop partnerships between Canadian industry and post-secondary institutions, and were happy to continue that work by joining with Plotly and biostatistics researchers at McGill University, said Eric Bosco, Chief Business Development Officer at Mitacs. Recognizing the data analysis capabilities of Plotly and biostatistics expertise of Dr. Bhatnagars team, we see this as a perfect match for both organizations to elevate their work and to help understand chronic pain alongside the Qubec Pain Research Network.

About MitacsMitacs is a not-for-profit organization that fosters growth and innovation in Canada for business and academia. Working with 70 universities, thousands of companies, and both federal and provincial governments, Mitacs builds partnerships that support industrial and social innovation in Canada. Open to all disciplines and all industry sectors, projects can span a wide range of areas, including manufacturing, business processes, IT, social sciences, design, and more. Mitacs is funded by the Government of Canada and the Government of Quebec, along with every other province, industry and not-for-profit partners, and academic partners.

About PlotlyFounded in 2013, Plotly is a data visualization company focused on taking data science out of the lab and into the business. Plotly makes it easy to create, deploy, and share interactive web apps, graphs, and visualizations in any programming language. Plotly's libraries are used by millions worldwide and embedded into mission critical applications across the Fortune 500.

ContactDanielle Toboni617-945-1915Plotly@LaunchSquad.com

See the original post here:
Plotly Sponsors Development of Predictive Genetics Application in Partnership with McGill University - GlobeNewswire

Read More...

Homology Medicines Presents Data from Investigational PKU and MLD Gene Therapy Programs that Demonstrate Preclinical Proof-of-Concept for Potential…

Monday, October 21st, 2019

BEDFORD, Mass., Oct. 21, 2019 (GLOBE NEWSWIRE) -- Homology Medicines Inc. (Nasdaq: FIXX), a genetic medicines company, announced today the presentation of preclinical data that support its investigational gene therapy programs for the treatment of metachromatic leukodystrophy (MLD) and phenylketonuria (PKU) at the American Society of Human Genetics (ASHG) 2019 Meeting.

For the first time, Homology presented preclinical data from the murine model and non-human primates that demonstrated that the HMI-202 gene therapy candidate crossed the blood-brain-barrier and the blood-nerve-barrier and increased levels of arylsulfatase a (ARSA) protein to therapeutic levels. In addition, preclinical data on gene therapy candidate HMI-102 showed that a single administration resulted in sustained Phe reduction and increased tyrosine and other important downstream biochemical metabolites in the PKU murine model.

The MLD presentation is part of a growing foundation of HMI-202 data to support a future IND filing, and the PKU preclinical data supported the initiation of our Phase 1/2 trial, which is ongoing and expected to report initial data by the end of this year, said Albert Seymour, Ph.D., Chief Scientific Officer of Homology Medicines. Taken together, these presentations demonstrate the potential of our genetic medicines platform, investigational PKU and MLD gene therapies and our continued focus on advancing these treatments to help patients and their families.

Highlights from the posters include:

HMI-202 gene therapy in development for MLD

HMI-102 investigational gene therapy for PKU

This poster received a Reviewers Choice Abstract award during the ASHG Meeting.

A 5-year retrospective study of individuals with PKU treated at two specialized U.S. clinics

For more information, please visit http://www.homologymedicines.com/publications.

About Phenylketonuria (PKU)PKU is a rare, inherited inborn error of metabolism caused by mutations in the PAH gene. The current standard of care is a highly restrictive diet, but it is not always effective, and there are currently no treatments available that address the genetic defect in PKU. If left untreated, PKU can result in progressive and severe neurological impairment. PKU affects approximately 16,500 people in the U.S., and an estimated 350 newborns are diagnosed each year.

About Metachromatic Leukodystrophy (MLD)MLD is a rare lysosomal storage disorder caused by mutations in the ARSA gene. ARSA is responsible for the creation of the arylsulfatase A (ARSA) protein, which is required for the breakdown of cellular components. In MLD, these cellular components accumulate and destroy myelin-producing cells in the peripheral and central nervous system leading to progressive and serious neurological deterioration. The late infantile form of the disorder is estimated to affect 1 in 40,000 people, and it is fatal within five to ten years after onset.

About Homology Medicines, Inc.Homology Medicines, Inc. is a genetic medicines company dedicated to transforming the lives of patients suffering from rare genetic diseases with significant unmet medical needs by curing the underlying cause of the disease. Homologys proprietary platform is designed to utilize its human hematopoietic stem cell-derived adeno-associated virus vectors (AAVHSCs) to precisely and efficiently deliver genetic medicinesin vivoeither through a gene therapy or nuclease-free gene editing modality across a broad range of genetic disorders. Homology has a management team with a successful track record of discovering, developing and commercializing therapeutics with a particular focus on rare diseases, and intellectual property covering its suite of 15 AAVHSCs. Homology believes that its compelling preclinical data, scientific expertise, product development strategy, manufacturing capabilities and intellectual property position it as a leader in the development of genetic medicines. For more information, please visitwww.homologymedicines.com.

Forward-Looking Statements This press release contains forward-looking statements within the meaning of the Private Securities Litigation Reform Act of 1995. All statements contained in this press release that do not relate to matters of historical fact should be considered forward-looking statements, including without limitation statements regarding our expectations surrounding the potential, safety, efficacy, and regulatory and clinical progress of our product candidates; plans and timing for the release of clinical data; our beliefs regarding our manufacturing capabilities; the potential of and related advancement of our novel platform and pipeline; our goal of delivering potential cures to patients; beliefs about preclinical data; our position as a leader in the development of genetic medicines; and the sufficiency of our cash, cash equivalents and short-term investments. These statements are neither promises nor guarantees, but involve known and unknown risks, uncertainties and other important factors that may cause our actual results, performance or achievements to be materially different from any future results, performance or achievements expressed or implied by the forward-looking statements, including, but not limited to, the following: we have and expect to continue to incur significant losses; our need for additional funding, which may not be available; failure to identify additional product candidates and develop or commercialize marketable products; the early stage of our development efforts; potential unforeseen events during clinical trials could cause delays or other adverse consequences; risks relating to the capabilities and potential expansion of our manufacturing facility; risks relating to the regulatory approval process; our product candidates may cause serious adverse side effects; inability to maintain our collaborations, or the failure of these collaborations; our reliance on third parties; failure to obtain U.S. or international marketing approval; ongoing regulatory obligations; effects of significant competition; unfavorable pricing regulations, third-party reimbursement practices or healthcare reform initiatives; product liability lawsuits; failure to attract, retain and motivate qualified personnel; the possibility of system failures or security breaches; risks relating to intellectual property and significant costs as a result of operating as a public company. These and other important factors discussed under the caption Risk Factors in our Quarterly Report on Form 10-Q for the quarter ended June 30, 2019 and our other filings with the SEC could cause actual results to differ materially from those indicated by the forward-looking statements made in this press release. Any such forward-looking statements represent managements estimates as of the date of this press release. While we may elect to update such forward-looking statements at some point in the future, we disclaim any obligation to do so, even if subsequent events cause our views to change.

Read this article:
Homology Medicines Presents Data from Investigational PKU and MLD Gene Therapy Programs that Demonstrate Preclinical Proof-of-Concept for Potential...

Read More...

Research presented by Invitae at the American Society of Human Genetics Meeting Pushes Science and Practice of Genetics Forward – Yahoo Finance

Monday, October 21st, 2019

-- Advances in classification, new approaches to genetics in cancer and implications of primary and secondary findings for clinical care among wide-ranging data presentations --

HOUSTON, Oct. 17, 2019 /PRNewswire/ -- Researchers fromInvitae Corporation (NVTA), a leading medical genetics company, are presenting data showing the increasing utility of genetic information at the American Society of Human Genetics (ASHG) annual meeting this week, ranging from comprehensive screening for cancer patients, to appropriate clinical follow up for women using non-invasive prenatal screening, to the limitations of direct to consumer genetic screening health reports.

The company's research includes three platform presentations and multiple poster sessions, many performed in collaboration with leading academic researchers. Among the data presented is a study evaluating the utility of combined germline testing and tumor profiling (somatic testing) in cancer patients. Germline and somatic testing are increasingly used in precision treatment of people with cancer, although frequently are ordered separately in clinical practice. Data presented at the meeting shows a substantial number of patients with medically significant variants in hereditary cancer syndrome genes in their tumor profile carry the same variant in their germline, thereby establishing a previously unknown risk of hereditary cancer and suggesting the value of combined or concurrent testing to inform precision medicine approaches.

"The research we are presenting at this year's ASHG meeting provides meaningful insight into both the science and practice of genetics, helping identify how we as clinicians can better use deep genetic insights to help a wide array of patients, whether they are cancer patients, women having a child or healthy adults seeking to better understand their risk of disease," said Robert Nussbaum, M.D., chief medical officer of Invitae. "We are proud and grateful to be able to join our colleagues from across genetic medicine in meaningful conversations that push genetic medicine forward."

Following are research from the company and collaborators to be presented at the meeting:

Story continues

Wednesday, October 16:

Poster presentation #819W | 2:00 3:00 pm Germline testing in colorectal cancer: Increased yield and precision therapy implications of comprehensive multigene panels. Presented by Shan Yang, PhD. Invitae.

Poster presentation #2427W | 2:00 3:00 pm Harmonizing tumor sequencing with germline genetic testing: identification of at-risk individuals for hereditary cancer disorders. Presented by Daniel Pineda-Alvarez, MD, FACMG, Invitae.

Poster presentation #606W | 3:00 4:00 pm A comprehensive evaluation of the importance of prenatal diagnostic testing in the era of increased utilization of non-invasive prenatal screening. Presented by Jenna Guiltinan, MS, LCGC, Invitae.

Thursday, October 17:

Platform presentation #235 | 5:00 pm, Room 370A, Level 3 Limitations of direct-to-consumer genetic screening for hereditary breast, ovarian and colorectal cancer risk. Presented by: Edward Esplin, MD, PhD, FACMG, FACP, Invitae.

Poster presentation #763T | 2:00 3:00 pm In-depth dissection of APC pathogenic variants: Spectrum of more than 400 pathogenic variants, challenges of variant interpretation, and new observations in a large clinical laboratory testing cohort. Presented by: Hio Chung Kang, PhD, Invitae.

Poster presentation #1399T | 2:00 3:00 pm Prediction of lethality and severity of osteogenesis imperfecta variants in the triple-helix regions of COL1A1 and COL1A2. Presented by: Vikas Pejaver, PhD, University of Washington.

Friday, October 18:

Platform presentation #264 | 9:00 am, Room 361D, Level 3 Million Veteran Program Return Of Actionable Results - Familial Hypercholesterolemia (MVP-ROAR-FH) Study: Considerations for variant return to mega-biobank participants. Presented by Jason Vassy, MD, MPH, VA, Boston Healthcare System.

Platform presentation #265 | 9:15 am, Room 361D, Level 3 Comprehensive secondary findings analysis of parental samples submitted for exome evaluation yields a high positive rate. Presented by Eden Haverfield, DPhil, FACMG, Invitae.

Poster presentation #698F | 2:00 3:00 pm Reporting of variants in genes with limited, disputed, or no evidence for a Mendelian condition among GenomeConnect participants. Presented by: Juliann Savatt, MS, LGC, Geisinger.

About InvitaeInvitae Corporation(NVTA)is a leading medical genetics company, whose mission is to bring comprehensive genetic information into mainstream medicine to improve healthcare for billions of people. Invitae's goal is to aggregate the world's genetic tests into a single service with higher quality, faster turnaround time, and lower prices. For more information, visit the company's website atinvitae.com.

Safe Harbor StatementsThis press release contains forward-looking statements within the meaning of the Private Securities Litigation Reform Act of 1995, including statements relating to the increasing utility of genetic information; the utility of combined germline and somatic testing; and the benefits of the company's research. Forward-looking statements are subject to risks and uncertainties that could cause actual results to differ materially, and reported results should not be considered as an indication of future performance. These risks and uncertainties include, but are not limited to: the applicability of clinical results to actual outcomes; the company's history of losses; the company's ability to compete; the company's failure to manage growth effectively; the company's need to scale its infrastructure in advance of demand for its tests and to increase demand for its tests; the company's ability to use rapidly changing genetic data to interpret test results accurately and consistently; security breaches, loss of data and other disruptions; laws and regulations applicable to the company's business; and the other risks set forth in the company's filings with the Securities and Exchange Commission, including the risks set forth in the company's Quarterly Report on Form 10-Q for the quarter ended June 30, 2019. These forward-looking statements speak only as of the date hereof, and Invitae Corporation disclaims any obligation to update these forward-looking statements.

Contact:Laura D'Angelopr@invitae.com(628) 213-3283

View original content to download multimedia:http://www.prnewswire.com/news-releases/research-presented-by-invitae-at-the-american-society-of-human-genetics-meeting-pushes-science-and-practice-of-genetics-forward-300940213.html

Read more from the original source:
Research presented by Invitae at the American Society of Human Genetics Meeting Pushes Science and Practice of Genetics Forward - Yahoo Finance

Read More...

Most genetic studies use only white participants this will lead to greater health inequality – The Conversation UK

Monday, October 21st, 2019

Few areas of science have seen such a dramatic development in the last decade as genomics. It is now possible to read the genomes of millions of people in so-called genome-wide association studies. These studies have identified thousands of small differences in our genome that are linked to diseases, such as cancer, heart disease and mental health.

Most of these genetic studies use data from white people over 78% of participants are of European descent. This doesnt mean that they represent Europe. In fact, only three nationalities make up most of the participants: the US, UK, and Iceland. Even though the UK and the US have very diverse populations, their non-white citizens have rarely been included in genetic research.

In recent years, efforts to collect multi-ethnic data have increased. One example is the UK Biobank, a collection of data from half a million British people accessible to any bona fide researcher. It includes some 35,000 DNA samples from people who are either non-European or mixed-race. Yet 92% of research papers on UK Biobank only used the data from the European-descent samples. So collecting data doesnt automatically solve the problem of non-white representation in research.

The under-representation of non-European groups is problematic for scientific and ethical reasons. The effects of gene variants that are present only in the unstudied groups remain unknown, which means important clues about the causes of diseases might be missed. Such undiscovered genes would not be included when testing for genetic diseases. So a person carrying one of them could wrongly get a negative genetic test result and might be told that they are not at increased risk of developing the disease.

Read more: How the genomics health revolution is failing ethnic minorities

Our recent work also shows that existing genetic findings might not apply equally to non-European populations. We found that some gene variants predicting high cholesterol in white populations do not lead to the same heart problems in people from rural Uganda. These findings should serve as a major warning to the field of genetics one cannot blindly apply findings from ancestrally European groups to everyone else.

It is important to support the global application of research because scientists have a moral responsibility to develop science for the benefit of the whole of humanity, not restricted by ethnic, cultural, economic or educational boundaries. Some 80% of the worlds population live in low and middle-income countries where healthcare and research are constrained by limited financial and human resources. We should not overlook this part of the world.

Studying different populations has advanced the medical field for everyones benefit. For example, the first disease gene mapped in humans was the gene for Huntingtons disease in 1983, identified through examining a large population of patients in villages surrounding Lake Maracaibo in Venezuela. The area was found to have the largest concentration of Huntingtons disease sufferers in the world, which helped them to find the gene.

More recently, a study of schizophrenia found new risk genes by using African and Latino American samples. Genetic risk scores based on results from these groups improved the ability to predict who would develop schizophrenia in all ethnic groups.

Read more: Decolonise science time to end another imperial era

Two things need to happen if we want to avoid increasing health disparities and instead share the medical benefits of genomic science across countries and ethnic groups. First, we need more large diverse studies. First steps in this direction are being taken by the Human Hereditary and Health in Africa Initiative. PAGE and All of Us are paving the way to recruit more diverse ethnic groups in the US, and East London Genes and Health focuses on people of South Asian origin in London.

And second, to make sure diverse ethnic data resources are widely used by researchers, the challenges of analysing genetic data from ancestrally diverse samples need to be addressed. While there are statistical solutions, more work is needed to make them easy to use and give clear guidance about the best approach.

Understanding how genetic risk and social inequality interact to influence disparities in disease risk and outcomes will be critical to improving public health for all.

Go here to read the rest:
Most genetic studies use only white participants this will lead to greater health inequality - The Conversation UK

Read More...

23andMe Wants Everyone to Get Used to Sharing Their Genetic Data – VICE

Monday, October 21st, 2019

This article originally appeared on VICE US.

What Anne Wojcicki, co-founder and CEO of 23andMe, would like you to consider is this: Once upon a time, people were kinda freaked out by the idea of typing their credit card info into a website. Now this is done basically all the time. Your phone is a digital wallet; your web browsers will memorize your card numbers for you. Might your bank information get cribbed or hacked? Has it already been? Yeah, probably so, but this is How We Live Now. Its weird to think it was ever not normal.

This little historic tale is how Wojcicki addresses the current hesitance a majority of people feel to submitting their genetic material to a database by spitting into a tube and mailing that tube off to a companys lab. In a brief discussion at the TIME 100 Health Summit in New York City on Thursday, Wojcicki explained that shes actually grateful for the 2030 percent of the population thats down to spit (according to a Twitter poll TIME ran ahead of Wojcickis talk). At-home DNA kits like 23andMe are still a relatively new technology. People simply need to grow accustomed to the ideas of genetic testing and sharing genomic data with public databases, so that researchers can observe patterns across the population, and ultimately make people healthier, she said.

Its essentially an argument for sharing DNA data as good for public health, like a new-age equivalent of getting vaccinated. To drive the point home further, Eric Lander, a geneticist and director of the Broad Institute of MIT (and someone who once sat around a table with Jeffrey Epstein), mentioned a potential breakthrough in treating angiosarcoma, a rare, highly fatal cancer by using using DNAthat, Lander argued, may never have happened if it werent for peoples willingness to fork over health and genetic information.

Its hard to find a sensible, non-demonic argument against something that could lead to expedited breakthroughs in cancer treatment. But what has to be kept in mind is that 23andMe is a private company. These anecdotesvirtuous as they may soundare marketing techniques. Wojcicki rightfully believes that no singular institution will be able to harvest the amount of DNA that her company has. According to 23andMes About page, more than 10 million people have spit into its tubes and thereby handed their genetic information over. Of those 10 million, 80 percent have opted in to participate in research, via that spit data. For context: The National Institute of Health is currently in the midst of enrolling its largest DNA-related study to date (All Of Us), which will reach full enrollment at one million participants. About a year into their recruiting efforts, theyre about a quarter of the way there, according to what NIH Director Francis Collins said in a separate panel at Thursdays TIME 100 Health Summit.

Wojcicki emphasized that only those who opt in for ancestry information have their data entered into public databases, which are subject to subpoena (the likes of which helped identify the Golden State Killer). She further emphasized that, because DNA is highly similar among family members, submitting your spit implicates your relatives genetic information. (23andMe consents around this.) Thats a hell of a lot of data thats sitting around for seemingly forever, and since 23andMe is, once again, a private company, theres no telling what happens to this info if/when the company goes under, or if they decide to change their policies.

What 23andMe is sitting on now is perhaps the most valuable pool of genetic data in the world. Earlier this year, the company partnered with TrialSpark, an NYC-based research company, in order to use its large database of data to fit its opted-in consumers to studies. Mind you, Wojcicki used to work on Wall Street. She is, at her core, a businessperson. Charging customers to have their data, and then partnering with another company with an interest in that data, sounds lucrative as fuck; a hell of a business deal.

Wojcicki and Lander concluded their talk on Thursday with a heartwarming sentiment: The virtue of something like 23andMe is that consumers (or participants, however youd prefer to look at it) have access to their genetic data, rather than submitting it to a study and never getting feedback. Wojcicki refers to this as empowering; its empowering to know whats going on in the little strands that make you the person you are. It also sounds very empowering, monetarily speaking, for those in the game, marketing spit kits and trading (totally consented for!!) genetic data.

More here:
23andMe Wants Everyone to Get Used to Sharing Their Genetic Data - VICE

Read More...

US Dairy Cows Are Very Genetically Similar. That’s Not Good : The Salt – NPR

Monday, October 21st, 2019

Unlike most dairy cows in America, which are descended from just two bulls, this cow at Pennsylvania State University has a different ancestor: She is the daughter of a bull that lived decades ago, called University of Minnesota Cuthbert. The bull's frozen semen was preserved by the U.S. Agriculture Department. Dan Charles/NPR hide caption

Unlike most dairy cows in America, which are descended from just two bulls, this cow at Pennsylvania State University has a different ancestor: She is the daughter of a bull that lived decades ago, called University of Minnesota Cuthbert. The bull's frozen semen was preserved by the U.S. Agriculture Department.

Chad Dechow, a geneticist at Pennsylvania State University who studies dairy cows, is explaining how all of America's cows ended up so similar to each other.

He brings up a website on his computer. "This is the company Select Sires," he says. It's one of just a few companies in the United States that sells semen from bulls for the purpose of artificially inseminating dairy cows.

Dechow chooses the lineup of Holstein bulls. This is the breed that dominates the dairy business. They're the black-and-white animals that give a lot of milk.

Dairy farmers can go to this online catalog and pick a bull, and the company will ship doses of semen to impregnate their cows. "There's one bull we figure he has well over a quarter-million daughters," Dechow says.

The companies rank their bulls based on how much milk their daughters have produced. Dechow picks one from the top of the list, a bull named Frazzled. "His daughters are predicted to produce 2,150 pounds more milk than daughters of the average bull," he says, reading from the website.

Farmers like to buy semen from top-ranked bulls, and the companies keep breeding even better bulls, mating their top performers with the most productive cows. "They keep selecting the same families over and over again," Dechow says.

A few years ago, Dechow and some of his colleagues at Penn State made a discovery that shocked a lot of people. All the Holstein bulls that farmers were using could trace their lineage back to one of just two male ancestors. "Everything goes back to two bulls born in the 1950s and 1960s," he says. "Their names were Round Oak Rag Apple Elevation and Pawnee Farm Arlinda Chief."

This doesn't mean that the bulls in the catalog are genetically identical. They still had lots of different mothers, as well as grandmothers. But it does show that this system of large-scale artificial insemination, with farmers repeatedly picking top-rated bulls, has made cows more genetically similar. Meanwhile, genetic traits that existed in Holstein cows a generation ago have disappeared.

"We've lost genetic variation," Dechow says. "Now, some of that variation was garbage that we didn't want to begin with. But some of it was valuable stuff."

To see what might have been lost, Dechow decided to do an experiment. He located some old semen from other bulls that were alive decades ago, with names like University of Minnesota Cuthbert and Zimmerman All-Star Pilot. You might call them heirloom bulls. The U.S. Agriculture Department keeps samples of their semen in deep-freeze storage in Fort Collins, Colo.

Dechow used that semen to impregnate some modern cows. They gave birth, and now it's possible to see some lost pieces of the Holstein family tree come to life in a barn at Penn State in the form of three cows.

Dechow leads the way to the barn. He points toward a cow that eyes us suspiciously. "Here is our old genetic lineage, [cow] number 2869," he says.

To the untrained eye, this cow looks pretty much like all the others. But Dechow sees things that others can't. "If you notice, if you look over her back see how that cow to her left is a little more bony?" he says.

Once Dechow points it out, the difference is plain to see. "So she definitely carries more body condition. She's a little bit fatter," he says.

Traditionally, dairy farmers didn't like cows with extra body fat. They thought the ideal cow was a skinny one, because she was turning all her feed into milk, not fat. So farmers chose bulls that tended to produce that kind of daughter.

"We've kind of selected for tall, thin, cows," Dechow says. "And that's a really bad combination. They're infertile, unhealthy. So we need to get away from that."

Dechow thinks the frozen semen from those long-forgotten heirloom bulls can bring back valuable genes that went missing maybe genes that would allow cows to thrive in warmer temperatures, for instance.

For this to work, though, farmers actually have to use those bulls, and they'll only do so if they're persuaded that the daughters will also produce lots of milk.

So Dechow is carefully monitoring his experimental cows. So far, he says, it's going pretty well. Two of the three cows are producing at least as much milk as the industry average.

Follow this link:
US Dairy Cows Are Very Genetically Similar. That's Not Good : The Salt - NPR

Read More...

National News Family tries to cure toddler with rare genetic disease Tomas Hoppough 9:36 AM – ABC15 Arizona

Monday, October 21st, 2019

A Denver family is trying to raise $3 million in order to cure their son with a rare genetic disease.

Doctors told Amber Freed that her 2-year-old son is one of 34 people in the world to have this rare neurological genetic disease.

The disease is so rare, it doesnt even have a name, Freed said. Its called SLC6A1, because that is the gene that it effects.

The disease causes Maxwell to have trouble moving and communicating, and soon it will only get worse.

The most debilitating part of the disease will begin between the ages of 3 and 4, Freed said. So, we are in a fight against time.

Maxwell has a twin sister named Riley.

I noticed early on that Maxwell wasnt progressing as much as Riley, Freed said. I noticed he couldnt use his hands. The doctors told me that every baby can use their hands. Thats when I realized there was something wrong with him.

After multiple visits to the doctor, Freed was able to find a genetic specialist to give Maxwell a diagnosis.

He looked at me and said, Something is very wrong with your son. I dont know if hes going to live, Freed said. My soul was just crushed. It was a sadness I didnt even know existed on earth. You never think something like this could happen. I left my career, and I had no other choice but to create my own miracle and to find a treatment forward to help Maxwell and all those others like him.

Freed searched for scientists trying to create a cure, which she found at the University of Texas Southwestern Medical Center in Dallas.

Were working with diseases where kids are born with a defective gene, said Steven Gray, an associate professor at UTSW in pediatrics. Our approach is to replace that gene to fix the condition at the level of their DNA. Were taking the DNA that those patients are missing and packaging that into a virus and use that virus as a molecular delivery truck to carry those genes back in their body and fix their DNA.

Its a rare disease, no one has ever heard of it, Freed said. But one rare disease messed with the wrong mother.

Freed said she has raised $1 million to help with research for the cure and will need an additional $3 million, in order to let Maxwell and many others continue to enjoy life.

I want Maxwell to have every opportunity that children should have in this life, Freed said. When he is having a good day, I just try and soak him in as much as I can. We dont take anything for granted in this house.

If you want to help donate for the cure, you can do so by visiting MilestonesforMaxwell.org or click on this GoFundMe page.

Read more here:
National News Family tries to cure toddler with rare genetic disease Tomas Hoppough 9:36 AM - ABC15 Arizona

Read More...

$2.1 million drug approved for Pearland toddler with rare genetic disease – KHOU.com

Monday, October 21st, 2019

PEARLAND, Texas A 2-year-old whose family has been fighting for a $2 million drug to battle her rare disease received approval for the drug Friday.

Krista James has Spinal Muscular Atrophy, or SMA, a life-threatening condition that severely impacts kids muscle movements. Her family has been fighting for the toddler to receive Zolgensma, a cutting-edge gene therapy that treats the disease at the genetic level.

RELATED: Pearland family fighting to get $2.1 million drug for toddler with rare genetic disease

Zolgensma is the most expensive drug to ever receive FDA approval. The treatment is priced at $2.125 million.

Despite Kristas doctor telling Medicaid its what the toddler needs, the request for coverage was denied until Friday, which happened to be Kristas 2nd birthday. Texas Health and Human Services approved the family's appeal.

The family told KHOU 11 they are beyond blessed and considered the approval the best birthday present ever.

ALSO POPULAR ON KHOU.COM

Send us a news tip | Download our app |#HTownRush Newsletter

Continue reading here:
$2.1 million drug approved for Pearland toddler with rare genetic disease - KHOU.com

Read More...

UCLH and UCL begin trial of genetic analysis in blood pressure patients – University College London Hospitals

Monday, October 21st, 2019

While this has been done as part of a number of studies in the past, the UCLH and UCL approach is unique in that they will also be returning this information to the patients as well as the doctors to understand how this information is useful and how it should be best communicated with doctors and patients.

For the UCLH BRCs AboutMe initiative, researchers will analyse patients DNA and other personal biological information to try to understand why some people have high blood pressure and others dont, and if it could be used to tailor treatment and whether it would be feasible for such testing to be rolled out across the NHS for other disease areas.

Some NHS patients have already had their DNA sequenced as part of the 100,000 Genomes Project done by Genomics England, but that project has so far focussed on rare diseases and cancer. UCLH and UCL researchers aim to extend this sequencing effort for common conditions. This effortand return of genomic information has not been done in the NHS before, within routine care.

The ultimate aim is that any NHS patient could have their DNA analysed in order to predict their chance of developing different conditions, prevent or reduce their risk of disease, and diagnose patients earlier with data returned to patients in a personalised report. Data from patients may also help in future with the development of drugs or tailoring of treatment.

For the initial study, research processes will be carried out in parallel with routine care to minimise disruption to patients. For instance, the doctor will seek the patients consent in the course of a clinic appointment, and the phlebotomist will take blood samples for research at the time of taking samples for standard tests. Blood samples will be processed by UCLHs laboratory.

The BRCs AboutMe initiative seeks to embed genomics and testing of other personal biological information into the NHS.

Researchers will be working with patients to look at how genetic information, including information on the risk of developing different conditions, can be best communicated to patients within the NHS environment.

See more here:
UCLH and UCL begin trial of genetic analysis in blood pressure patients - University College London Hospitals

Read More...

Food As Medicine: What Biochemistry And Genetics Are Teaching Us About How To Eat Right – Forbes

Thursday, October 10th, 2019

We often talk about genetics as if its set in stone. She just has good genes or He was born with it are common phrases.

However, over the past decade, biochemists and geneticists have discovered that your genetic expression changes over time. Based on environmental factors, certain genes may be strongly expressive while others are dormant.

In fact, a 2016 study of human longevity found that only 25% of health outcomes are attributable to genetics. The other 75% of outcomes are attributable to environmental factors. Among those environmental factors, diet and nutrition play a major role.

An entire branch of scientific research has now exploded around nutrigenomics, the study of the interaction between nutrition and genetics. Scientists now understand that genes set the baseline for how your body can function, but nutrition modifies the extent to which each gene is expressed.

As more data comes in about the types and quality of food that improve health outcomes, high-tech farmers are also entering the nutrigenomics conversation. Using precision agriculture, they hope to produce food thats targeted to deliver a nutrient-rich, genetically beneficial diet.

Implications Of Nutrigenomics

Researchers have found that theres no such thing as a perfect diet. Dietary recommendations are not one-size-fits-all. Each individual needs different nutritional choices for optimal health and gene expression. In addition, each person is different in the extent to which their genes and health are impacted by their diet.

Geneticists and nutritionists are working together to study the dietary levers that most impact genetic expression. If theyre successful, it may be possible to prevent and treat disease through individualized nutrition tailored to your genetic profile. Indeed, you may walk into a doctors office and leave with a dietary prescription customized to your DNA.

In the near future, instead of diagnosing and treating diseases caused by genome or epigenome damage, health care practitioners may be trained to diagnose and nutritionally prevent or even reverse genomic damage and aberrant gene expression, reports Michael Fenech, a research scientist at CSIRO Genome Health and Nutrigenomics Laboratory.

The initial results of nutrigenomics studies are promising. A healthy, personalized diet has the potential to prevent, mitigate, or even cure certain chronic diseases. Nutrigenomics has shown promise in preventing obesity, cancer and diabetes.

If Food Is Medicine, Food Quality Matters

Nutrient abundance or deficiency is the driving factor behind nutrigenomics. Foods that have grown in poor conditions have a lower nutritional density. In turn, eating low-quality foods can have a significant impact on human gene expression. In order to take advantage of the findings of nutrigenomics, consumers need access to high-quality, nutrient-dense foods.

Similar to human health, plant health is impacted by the combination of genes and nutrient intake. Healthy soil, correctly applied fertilization techniques, and other forms of environmental management lead to healthy crops.

However, applying these custom growing techniques at a large scale is a major challenge. Agriculture technology (AgTech) will play a big role in allowing farmers to precisely manage the growing conditions and nutrient delivery for their crops. In turn, this precision farming will make crops more nutritious and targeted for nutrigenomics-driven diets.

Making Food Thats Better For Us

Plant health relies on nutrient uptake from the soil. In order to ensure plants receive the nutrients they need, farmers need to precisely apply additives where theyre needed. With in-ground sensors, advanced mapping of crop quality across a field, and other technologies, farmers can target their applications of water and nutrients to match plant needs. The days of broadly applying generic fertilizer to entire fields are coming to an end.

Farmers play an integral role in providing access to diverse, nutritious food, explains Remi Schmaltz, CEO of Decisive Farming. Nutrient deficiency in plants and the soil can contribute to the deficiencies found in humans. The opportunity exists to address these deficiencies through precision nutrition delivered by the agriculture sector.

Additionally, CRISPR and other technologies allow us to experiment with the genetic makeup of plants, increasing nutrition and flavor, both pluses for consumers. In recent years, genetic modification has produced disease-resistant bananas, more flavorful tomatoes, lower gluten wheat, non-browning mushrooms and sustainable rice. While there has been a lot of skepticism over genetically-modified crops, multiple studies have shown that GMOs are safe for consumption and can even improve plant health and nutrition.

Using Biochemistry And Big Data To Create Better Food And Healthier People

Nutrigenomics will completely change how we think about health and disease prevention. Indeed, personalized diet recommendations that are tailored to your genes could be a new form of medicine for chronic illnesses.

Nevertheless, a key part of making nutrigenomics effective is having access to high-quality, nutrient-dense foods. AgTech is using the internet of things, AI, precision farming and gene editing to make nutrient-dense food more readily available. The benefits to public health from these efforts could change the way we think about medicine, longevity and what it means to be healthy.

Continue reading here:
Food As Medicine: What Biochemistry And Genetics Are Teaching Us About How To Eat Right - Forbes

Read More...

The thorny ethics of collecting genetic data – Quartz

Thursday, October 10th, 2019

In 2009, researchers collected DNA from four elderly men in Namibia, each from one of the many San indigenous communities scattered across southern Africa. A year later, analyses of the mens DNA were published in the journal Naturealongside that of South African human rights activist Desmond Tutu. The intention, in part, was to increase the visibility of southern, indigenous Africans in genetic-based medical research. Soon after, a nongovernmental organization representing indigenous minorities in Southern Africa took issue with the consent procedures used to gather the data and wrote to Natures editors accusing the papers authors of absolute arrogance, ignorance, and cultural myopia.

The San case highlights the thorny ethics of collecting genetic data. Yet today, to make medicine more equitable, scientists see the importance of sampling DNA from more diverse populations. Most genetic research uses DNA from descendants of Europeans, which means the related medical applicationssuch as genetic tests to see the likelihood of developing a certain disease, called polygenic risk assessments can only benefit those populations. In 2018 in the United States, for example, the National Institutes of Health launched All of Us, a research program that aims to collect DNA, electronic health records, and other data, from about one million Americans with emphasis on including many different groups of people.

When we do genetic studies, trying to understand the genetic basis of common and complex diseases, were getting a biased snapshot, said Alicia Martin, a geneticist at the Massachusetts General Hospital and the Broad Institute, a biomedical and genomics research center affiliated with Harvard and MIT.

Research to capture these snapshots, called genome-wide association studies, can only draw conclusions about the data thats been collected. Without studies that look at each underrepresented population, genetic tests and therapies cant be tailored to everyone. Still, projects intended as correctives, like All of Us and the International HapMap Project, face an ethical conundrum: Collecting that data could exploit the very people the programs intend to help.

Researchers with All of Us have already collected data from about 1,600 Native Americans, some of whom live in cities outside of sovereign lands, where tribal approval is not necessary for genetic research, according to Krystal Tsosie, a geneticist at Vanderbilt University who is co-leading a study in collaboration with a tribal community in North Dakota .

Obviously theres an interest in monetizing biomarkers collected from diverse populations and underrepresented populations, Tsoise said, so without adequate protections, the concern becomes about exploitation.

Medical genetic research generally works like this: Geneticists use powerful computers to compare the genomes of people affected by a particular disease to healthy controls. Researchers mark genetic patterns that are common in people with, say, diabetes, but not the controls, as associated with the disease. The more samples geneticists feed to the algorithms, the more likely that the findings reflect reality.

But studies restricted to descendants of Europeans will only find associations between diseases and variants that are common in European ancestry populations, said Martinif those variants are common enough to be found.

Scientists use the results to develop polygenic risk scores, which count the risky variants on someones genome to estimate their susceptibility to a disease. But if studies dont use the genomes of non-white populations, the tests wont pick up on the problematic variants in different groups of people. One 2019 Nature Genetics study, on which Martin was an author, determined that these blind spots reduce the accuracy of polygenic tests by approximately two and five times in South or East Asian, and black populations, respectively.

In many cases, the groups whose DNA is missing have worse health care outcomes compared to their white counterparts, and genetic medicine could worsen these disparities. I think theres a huge responsibility, said Martin. If we look at the history of the field, over the past decade weve gone from participants in genetic studies being 96% European ancestry to about 80 percent. Weve shifted gears a little bit, but not nearly enough to be able to serve minority populations. Jantina De Vries, a bioethicist at the University of Cape Town, agreed that representation in genomics research can bring health benefits, particularly if it is paired with measures to build research capacity so that, eventually, there are researchers at every level within the groups themselves.

Collecting broader genetic samples poses a host of challenges. Efforts to collect and study the genomes of indigenous peoples, for example, have been controversial since the early 90s. The first such project, called the Human Genome Diversity Project (HGDP), was meant to explore the full range of genome diversity within the human family by collecting DNA samples from about 500 distinct groups, with an emphasis on indigenous peoples that might soon vanish. Indigenous-rights organizations criticized the project, taking issue with being treated as mere objects of scientific interest and potential for commercialization. All of Us, more recently, has run into similar objections from the National Congress of American Indians.

The concerns are linked to the long history of exploitative encounters between researchers and vulnerable populations. The Tuskegee Studyin which the US Public Health Service withheld treatment from African American men with syphilislasted from 1932 until 1972, ending less than 20 years before the HGDP proposal. And in 1989, researchers from Arizona State University collected DNA samples from the Havasupai Tribe and reused the data for research to which the participants hadnt consented: on schizophrenia, inbreeding, and migration history. Tsosie said this context has created a climate in which weve seen tribes deciding to disengage from biomedical research completely.

All the geneticists and ethicists Undark spoke with agreed that community engagement is crucial to establish trust. But they didnt agree on the degree of the engagement. Some believed that gaining the consent of communities is necessary for ethical research, while others said it was enough to have respect and open dialogue between researchers and the people theyd like to study.

But both approaches are difficult in the context of collecting and analyzing genetic data, since geneticists take DNA from individuals to make conclusions about entire populations. For instance, the San paper in Nature extrapolated findings regarding individual genomes to discuss the genomes of the broader communities. Ones genome is not their own specifically; ones genome is informed by their recent ancestry, their family structures, and their more distant ancestry, said Tsosie. Geneticists are never talking about an individual thats siloed.

The gap between individual and collective consent is partly responsible for the continued friction between genetic science and indigenous peoples. Collective consent, said Tsosie, who is herself Navajo, is more culturally consistent with how tribal groups govern themselves. In 2017, Andries Steenkamp, a San leader, and Roger Chennells, a lawyer, wrote that the Nature study failed in this regard by only getting informed consent from the indigenous individuals who participated.

Not everyone agrees that collective consent can or should be a requirement for all genetic studies. For instance, de Vries said, it depends what sort of community were talking about, drawing a contrast between small, rural, communities and larger populations spread across several cities or countries. If were talking about the entire Yoruba population, who would you even talk to? she added. The Yoruba are an ethic group of more than 20 million individuals, most of whom live in Nigeria, with smaller populations in Benin, Togo, and across several diaspora communities. De Vries believes the onus lies on researchers to think in terms of respecting communities, rather than in terms of collective consent.

Gaining collective consent involves logistical hurdles, especially for large-scale projects. The NIHs All of Us program, for example, wasnt able to get input from each of the 573 federally-recognized tribes. According to Tsosie, during the planning stages, there was talk of gaining tribal input, but that plan seemed to be abandoned early on. The All of Us website does have a section on tribal engagement, but only offers formal consultation and listening sessions for ongoing projects, not guidance on how to approach these issues before a project starts.

Among non-indigenous policymakers and scientists, Tsosie noted, theres a magical notion that stakeholders from every tribe can be brought together in one room when, in reality, that is not how we make consensus decisions for ourselves.

Even more difficult than logistics, perhaps, may be conceptualizing the genetic studies to begin withfor example, deciding which people belong in which groups. One of the greatest political acts, acts of power, that we perform as human beings is dividing ourselves up for the purpose of knowing and governing ourselves, said Jenny Reardon, a sociologist who specializes in genomics at the University of California, Santa-Cruz.

Globally, indigenous peoples are so culturally distinct from one another that a single understanding of a community wont resonate with everyone. Finding a method for data collection that crosses all indigenous groups is going to be really hard, said Vanessa Hayes, a geneticist at the Garvan Institute of Medical Research and the University of Sydney in Australia who conducts fieldwork in South Africa. Because, straightaway, thats assuming all indigenous people are the same. Without common ground, scientists must do the hard work of understanding each unique community. As Hayes put it, every group that you work with, you have to respect that group, and take the time to understand what is important in that group.

Hayes was one of the authors of the 2010 Nature study on the San, and she was responsible for obtaining consent, gathering samples, and discussing the results with the community. While Steenkamp and Chennells suggested the researchers were hasty in their data collection and ignored governance structures, Hayes countered that, at the time of the study, shed already been working in these communities for more than a decade and they were working directly with government agencies. Shed been in contact with the Working Group of Indigenous Minorities in Southern Africa (WIMSA)the NGO which would eventually criticize the studybefore it began. But, she said, when I went back to the community and asked if they knew who WIMSA was, they said no. I asked them if they wanted WIMSA to represent them, and they said, Hell no.

(As an organization, WIMSA is currently being restructured. The South African San Council, which now represents the San communities of South Africa, declined an interview with Undark, citing a requirement for financial compensation and a signed contract.)

Hayes followed the principles of collective consent, she said, just at a lower level than formal institutions like WIMSA or the San Council: Their decision was made as a group. They are the group, they are the band, they are the family. She added, No one can represent them that is not them.

The difficulties in defining a group make collective consent even more challenging.

In the clearest of circumstances, where an established organization exists, approval processes can be difficult to navigate and can take months. But within some indigenous and minority groups, issues of representation remain controversial. Often, a scientist will have to invest a lot of time interacting with potential subjects in order to judge what consent procedures are appropriate. Few scientists have the necessary time and resources.

There is no easy way to choose which organizations to deal with, especially when there are internal disagreements about representation. Or, as Reardon put it: The folks that are trying to democratize the science are going to have the same problem as the people who were attempting to treat it as Were just going to go out and get these groups, and study them from a scientific perspective.

Although the repeated controversies surrounding research and indigenous groups may have slowed their inclusion in genetic science, the researchers Undark spoke with said ensuring these concerns are heard and addressed is a vital part of the work. Indigenous groups are demanding a greater say in research that concerns them, whether under the All of Us program or conducted by individual researchers in Africa. Resolving the ethical ambiguities is no easy task, but, as Hayes asked: Why should it be easy?

This article was originally published on Undark. Read the original article.

View original post here:
The thorny ethics of collecting genetic data - Quartz

Read More...

Innolea Created to Focus on Genetics and Genomics of Oil Crops – Seed World

Thursday, October 10th, 2019

Since January 1, 2019, the research activities in oilcrop genetics and genomics of the company Biogemma moved to a new company, Innolea, under the control of three major French seed companies in oil crops: Euralis Semences, Limagrain, RAGT Semences and the innovation fund for oil and protein crops, Sofiprotol.

Innolea is supported by experienced teams andknow-how developed for more than 20 years by Biogemma and benefit of theinfrastructure of Mondonville (Haute-Garonne) site, which is now the headquarter,as well as the support of its shareholders.

Innoleas direction was given to Bruno Grzes-Besset who was research coordinator in Biogemma, and the presidency to Jean-Marc Ferullo, the research director of Euralis Semences.

Biogemmas research programs in rapeseed and sunflower, the main oil crops in France and Europe, are continued in Innolea. The sunflower program is led by Delphine Fleury and the rapeseed program by Sbastien Faure.

To support the French oil industry, Innoleasactivities focus on the genetics of traits of interest with direct applicationsto breeding new varieties. The main research areas will be the exploration ofgenetic diversity of the main oil crops of France and Europe and the characterisationof beneficial genes.

By creating this new company, the French oil and protein industry and the seeds companies demonstrate their engagement in supporting competitive research in applied genomics and pre-breeding. This leading-edge knowledge will enable offering new varieties to the French market, with improved agronomy and particularly plant resistance to diseases and pests.

Read more:
Innolea Created to Focus on Genetics and Genomics of Oil Crops - Seed World

Read More...

Page 33«..1020..32333435..40..»


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